1
|
García-Martínez J, Salto R, Girón MD, Pérez-Castillo ÍM, Bueno Vargas P, Vílchez JD, Linares-Pérez A, Manzano M, García-Córcoles MT, Rueda R, López-Pedrosa JM. Supplementation with a Whey Protein Concentrate Enriched in Bovine Milk Exosomes Improves Longitudinal Growth and Supports Bone Health During Catch-Up Growth in Rats. Nutrients 2024; 16:3814. [PMID: 39599602 PMCID: PMC11597726 DOI: 10.3390/nu16223814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Undernutrition impairs linear growth while restoration of nutritional provisions leads to accelerated growth patterns. However, the composition of the nutrition provided is key to facilitating effective catch-up growth without compromising bone quantity, quality, and long-term health. METHODS We evaluated the role of a whey protein concentrate enriched in bovine milk exosomes (BMEs) in modulating the proliferative properties of human chondrocytes in vitro and studied how these effects might impact bone quantity and quality measured as longitudinal tibia growth, bone mineral content (BMC) and density (BMD), and trabecular micro-CT parameters in stunted rats during catch-up growth. RESULTS BMEs promoted proliferation in C28/I2 human chondrocytes mediated by mTOR-Akt signaling. In a stunting rat model, two-week supplementation with BMEs during refeeding was associated with improved tibia BMD, trabecular microstructure (trabecular number (Tb. N.) and space (Tb. Sp.)), and a more active growth plate (higher volume, surface, and thickness) compared to non-supplemented stunted rats. Positive effects on physis translated to significantly longer tibias without compromising bone quality when extending the refeeding period for another two weeks. CONCLUSIONS Overall, BME supplementation positively contributed to longitudinal bone growth and improved bone quantity and quality during catch-up growth. These findings might be relevant for improving diets aimed at addressing the nutritional needs of children undergoing undernutrition during early life.
Collapse
Affiliation(s)
- Jorge García-Martínez
- Abbott Nutrition R&D, Abbott Laboratories, 18004 Granada, Spain; (Í.M.P.-C.); (P.B.V.); (M.M.); (M.T.G.-C.); (R.R.); (J.M.L.-P.)
| | - Rafael Salto
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Campus de Cartuja, 18071 Granada, Spain; (R.S.); (M.D.G.); (J.D.V.); (A.L.-P.)
| | - María D. Girón
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Campus de Cartuja, 18071 Granada, Spain; (R.S.); (M.D.G.); (J.D.V.); (A.L.-P.)
| | - Íñigo M. Pérez-Castillo
- Abbott Nutrition R&D, Abbott Laboratories, 18004 Granada, Spain; (Í.M.P.-C.); (P.B.V.); (M.M.); (M.T.G.-C.); (R.R.); (J.M.L.-P.)
| | - Pilar Bueno Vargas
- Abbott Nutrition R&D, Abbott Laboratories, 18004 Granada, Spain; (Í.M.P.-C.); (P.B.V.); (M.M.); (M.T.G.-C.); (R.R.); (J.M.L.-P.)
| | - Jose D. Vílchez
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Campus de Cartuja, 18071 Granada, Spain; (R.S.); (M.D.G.); (J.D.V.); (A.L.-P.)
| | - Azahara Linares-Pérez
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Campus de Cartuja, 18071 Granada, Spain; (R.S.); (M.D.G.); (J.D.V.); (A.L.-P.)
| | - Manuel Manzano
- Abbott Nutrition R&D, Abbott Laboratories, 18004 Granada, Spain; (Í.M.P.-C.); (P.B.V.); (M.M.); (M.T.G.-C.); (R.R.); (J.M.L.-P.)
| | - María T. García-Córcoles
- Abbott Nutrition R&D, Abbott Laboratories, 18004 Granada, Spain; (Í.M.P.-C.); (P.B.V.); (M.M.); (M.T.G.-C.); (R.R.); (J.M.L.-P.)
| | - Ricardo Rueda
- Abbott Nutrition R&D, Abbott Laboratories, 18004 Granada, Spain; (Í.M.P.-C.); (P.B.V.); (M.M.); (M.T.G.-C.); (R.R.); (J.M.L.-P.)
| | - José M. López-Pedrosa
- Abbott Nutrition R&D, Abbott Laboratories, 18004 Granada, Spain; (Í.M.P.-C.); (P.B.V.); (M.M.); (M.T.G.-C.); (R.R.); (J.M.L.-P.)
| |
Collapse
|
2
|
Azrad-Daniel S, Cupa-Galvan C, Farca-Soffer S, Perez-Zincer F, Lopez-Acosta ME. Unusual presentation of Loeys-Dietz syndrome: A case report of clinical findings and treatment challenges. World J Clin Cases 2022; 10:12247-12256. [PMID: 36483799 PMCID: PMC9724511 DOI: 10.12998/wjcc.v10.i33.12247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/21/2022] [Accepted: 10/26/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Loeys-Dietz syndrome (LDS) is a rare autosomal dominant syndrome characterized by heterozygous mutations causing multisystemic alterations. It was recently described in 2005, and today at least six different subtypes have been identified. Classically presenting with aortic root enlargement or aneurysms and craniofacial and skeletal abnormalities, with specific arterial tortuosity at any site. The differential diagnosis of LDS includes atypical Marfan syndrome, vascular Ehlers-Danlos syndrome, Shprintzen-Goldberg craniosynostosis, and familial aortic aneurysm and dissection syndrome.
CASE SUMMARY We present a case study of a 35-year-old female who came to the emergency department due to lower gastrointestinal bleeding and severe abdominal pain. Computed tomography revealed vascular tortuosity in almost every abdominal vein.
CONCLUSION This case report will help us analyze the infrequent presentation of LDS type 4 and the numerous complications that it implies, underlying the importance of publishing more cases in order to expand our knowledge and offer better treatment for these patients. Differential diagnosis, clinical presentation and treatment options for this syndrome are discussed in this article.
Collapse
Affiliation(s)
- Shely Azrad-Daniel
- Department of Internal Medicine, Hospital Angeles Lomas, Huixquilucan 52763, Mexico
| | | | - Sion Farca-Soffer
- Department of Internal Medicine, Hospital Angeles Lomas, Huixquilucan 52763, Mexico
| | | | | |
Collapse
|
3
|
Furlan AG, Spanou CES, Godwin ARF, Wohl AP, Zimmermann LMA, Imhof T, Koch M, Baldock C, Sengle G. A new MMP-mediated prodomain cleavage mechanism to activate bone morphogenetic proteins from the extracellular matrix. FASEB J 2021; 35:e21353. [PMID: 33629769 DOI: 10.1096/fj.202001264r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 12/04/2020] [Accepted: 12/23/2020] [Indexed: 11/11/2022]
Abstract
Since their discovery as pluripotent cytokines extractable from bone matrix, it has been speculated how bone morphogenetic proteins (BMPs) become released and activated from the extracellular matrix (ECM). In contrast to TGF-βs, most investigated BMPs are secreted as bioactive prodomain (PD)-growth factor (GF) complexes (CPLXs). Recently, we demonstrated that PD-dependent targeting of BMP-7 CPLXs to the extracellular fibrillin microfibril (FMF) components fibrillin-1 and -2 represents a BMP sequestration mechanism by rendering the GF latent. Understanding how BMPs become activated from ECM scaffolds such as FMF is crucial to elucidate pathomechanisms characterized by aberrant BMP activation and ECM destruction. Here, we describe a new MMP-dependent BMP-7 activation mechanism from ECM-targeted pools via specific PD degradation. Using Edman sequencing and mutagenesis, we identified a new and conserved MMP-13 cleavage site within the BMP-7 PD. A degradation screen with different BMP family PDs and representative MMP family members suggested utilization of the identified site in a general MMP-driven BMP activation mechanism. Furthermore, sandwich ELISA and solid phase cleavage studies in combination with bioactivity assays, single particle TEM, and in silico molecular docking experiments provided evidence that PD cleavage by MMP-13 leads to BMP-7 CPLX disintegration and bioactive GF release.
Collapse
Affiliation(s)
- Ariane G Furlan
- Center for Biochemistry, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Chara E S Spanou
- Center for Biochemistry, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alan R F Godwin
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Alexander P Wohl
- Center for Biochemistry, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Laura-Marie A Zimmermann
- Center for Biochemistry, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thomas Imhof
- Institute for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Clair Baldock
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Gerhard Sengle
- Center for Biochemistry, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), Cologne, Germany
| |
Collapse
|
4
|
Controlling BMP growth factor bioavailability: The extracellular matrix as multi skilled platform. Cell Signal 2021; 85:110071. [PMID: 34217834 DOI: 10.1016/j.cellsig.2021.110071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 01/23/2023]
Abstract
Bone morphogenetic proteins (BMPs) belong to the TGF-β superfamily of signaling ligands which comprise a family of pluripotent cytokines regulating a multitude of cellular events. Although BMPs were originally discovered as potent factors extractable from bone matrix that are capable to induce ectopic bone formation in soft tissues, their mode of action has been mostly studied as soluble ligands in absence of the physiologically relevant cellular microenvironment. This micro milieu is defined by supramolecular networks of extracellular matrix (ECM) proteins that specifically target BMP ligands, present them to their cellular receptors, and allow their controlled release. Here we focus on functional interactions and mechanisms that were described to control BMP bioavailability in a spatio-temporal manner within the respective tissue context. Structural disturbance of the ECM architecture due to mutations in ECM proteins leads to dysregulated BMP signaling as underlying cause for connective tissue disease pathways. We will provide an overview about current mechanistic concepts of how aberrant BMP signaling drives connective tissue destruction in inherited and chronic diseases.
Collapse
|
5
|
Simonds MM, Schlefman AR, McCahan SM, Sullivan KE, Rose CD, Brescia AMC. The culture microenvironment of juvenile idiopathic arthritis synovial fibroblasts is favorable for endochondral bone formation through BMP4 and repressed by chondrocytes. Pediatr Rheumatol Online J 2021; 19:72. [PMID: 33980237 PMCID: PMC8117630 DOI: 10.1186/s12969-021-00556-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/16/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND We examined influences of conditioned media from chondrocytes (Ch) on juvenile idiopathic arthritis synovial fibroblasts (JFLS) and potential for JFLS to undergo endochondral bone formation (EBF). METHODS Primary cells from three control fibroblast-like synoviocytes (CFLS) and three JFLS were cultured in Ch-conditioned media and compared with untreated fibroblast-like synoviocytes (FLS). RNA was analyzed by ClariomS microarray. FLS cells cultured in conditioned media were exposed to either TGFBR1 inhibitor LY3200882 or exogenous BMP4 and compared with FLS cultured in conditioned media from Ch (JFLS-Ch). Media supernatants were analyzed by ELISA. RESULTS In culture, JFLS downregulate BMP2 and its receptor BMPR1a while upregulating BMP antagonists (NOG and CHRD) and express genes (MMP9, PCNA, MMP12) and proteins (COL2, COLX, COMP) associated with chondrocytes. Important TGFβ superfamily member gene expression (TGFBI, MMP9, COL1A1, SOX6, and MMP2) is downregulated when JFLS are cultured in Ch-conditioned media. COL2, COLX and COMP protein expression decreases in JFLS-Ch. BMP antagonist protein (NOG, CHRD, GREM, and FST) secretion is significantly increased in JFLS-Ch. Protein phosphorylation increases in JFLS-Ch exposed to exogenous BMP4, and chondrocyte-like phenotype is restored in BMP4 presence, evidenced by increased secretion of COL2 and COLX. Inhibition of TGFBR1 in JFLS-Ch results in overexpression of COL2. CONCLUSIONS JFLS are chondrocyte-like, and Ch-conditioned media can abrogate this phenotype. The addition of exogenous BMP4 causes JFLS-Ch to restore this chondrocyte-like phenotype, suggesting that JFLS create a microenvironment favorable for endochondral bone formation, thereby contributing to joint growth disturbances in juvenile idiopathic arthritis.
Collapse
Affiliation(s)
- Megan M. Simonds
- grid.239281.30000 0004 0458 9676Nemours Biomedical Research, Nemours/Alfred I. duPont Hospital for Children, 1701 Rockland Rd, Wilmington, DE 19803 USA
| | - Amanda R. Schlefman
- grid.413611.00000 0004 0467 2330Rheumatology, Johns Hopkins All Children’s Hospital, St. Petersburg, FL USA
| | - Suzanne M. McCahan
- grid.239281.30000 0004 0458 9676Nemours Biomedical Research, Nemours/Alfred I. duPont Hospital for Children, 1701 Rockland Rd, Wilmington, DE 19803 USA
| | - Kathleen E. Sullivan
- grid.239552.a0000 0001 0680 8770Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Carlos D. Rose
- grid.239281.30000 0004 0458 9676Division of Rheumatology, Nemours/Alfred I. duPont Hospital for Children, 1701 Rockland Rd, Wilmington, DE 19803 USA
| | - Anne Marie C. Brescia
- grid.239281.30000 0004 0458 9676Division of Rheumatology, Nemours/Alfred I. duPont Hospital for Children, 1701 Rockland Rd, Wilmington, DE 19803 USA
| |
Collapse
|
6
|
TGF-β Activity of a Demineralized Bone Matrix. Int J Mol Sci 2021; 22:ijms22020664. [PMID: 33440877 PMCID: PMC7827646 DOI: 10.3390/ijms22020664] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 12/25/2022] Open
Abstract
Allografts consisting of demineralized bone matrix (DBM) are supposed to retain the growth factors of native bone. However, it is not clear if transforming growth factor β1 (TGF-β1) is maintained in the acid-extracted human bone. To this aim, the aqueous solutions of supernatants and acid lysates of OraGRAFT® Demineralized Cortical Particulate and OraGRAFT® Prime were prepared. Exposing fibroblasts to the aqueous solution caused a TGF-β receptor type I kinase-inhibitor SB431542-dependent increase in interleukin 11 (IL11), NADPH oxidase 4 (NOX4), and proteoglycan 4 (PRG4) expression. Interleukin 11 expression and the presence of TGF-β1 in the aqueous solutions were confirmed by immunoassay. Immunofluorescence further confirmed the nuclear translocation of Smad2/3 when fibroblasts were exposed to the aqueous solutions of both allografts. Moreover, allografts released matrix metalloprotease-2 activity and blocking proteases diminished the cellular TGF-β response to the supernatant. These results suggest that TGF-β is preserved upon the processing of OraGRAFT® and released by proteolytic activity into the aqueous solution.
Collapse
|
7
|
Vieira JS, Cunha EJ, de Souza JF, Chaves LHK, de Souza JL, Giovanini AF. Alendronate disturbs femoral growth due to changes during immunolocalization of transforming growth factor-β1 and bone morphogenetic protein-2 in epiphyseal plate. World J Exp Med 2020; 10:1-9. [PMID: 31942441 PMCID: PMC6960019 DOI: 10.5493/wjem.v10.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 11/26/2019] [Accepted: 12/15/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The epiphyseal growth plate is an important anatomical segment localized on the ends of a long bone. Despite the abovementioned atractive reasons for alendronate’s use, few data on the effect of alendronate during epiphyseal growth exist.
AIM Verify the effect of alendronate on the growth epiphyseal plate, and compare its effect with the size of the femur during the double-staining of the immunolocalization of transforming growth factor-β1 (TGF-β1) and bone morphogenetic protein-2 (BMP2) in endochondral ossifing in specimens that have received alendronate.
METHODS Forty newborn rats were randomly divided into two groups: a control group (were given applications of 1 mg/kg physiologic saline) and a group that received Alendronate (a dose of 2.5 mg/kg). These groups were then divided into two subgroups for euthanasia in two and 12 d of life. After euthanasia, the femurs were removed, and the femoral bones were measured linearly between the apex of the greater trochanter until the lower intercondylar midlle face to verify the probable bone growth between 3 and 12 d in control and alednroanto treated rats. Posteriorly, the surgical pieces were also sent to the histopathology laboratory to produce histological slides. The obtained slides were stained with hematoxylin and eosin to measure each of the cartilage zones in endochondral development. and other slides were immunohistochemically tested for anti- TGF-β1 and BMP-2 antibodies to investigate the immunolocalization of these proteins in the epiphyseal plaque area.
RESULTS On the third day, some diferences between the control group and specimens treated with alendronate were verified. Macroscopiccaly, we found similarities in size between the femoral bones when we compared the control group with the specimens that received alendronate. On the 12th day, the bone size of the mice receiving the drug was significantly smaller than those of the control group. These results coincide with changes in the TGF-β1 and BMP-2 expression. In the specimens that received alendronate, the TGF-β1 was expressed in some sites of trabecular bone that was neoformed, peripherally to the bone marrow area. The BMP-2 was also positive in proliferative chondrocytes and hypertrofic chondrocytes. On the 12th day, all layers of chondrocytes exhibited positivity for BMP-2 in the specimens that received alendronate. In the interface between the trabecular bone and cartilage, an area of disorganized bone deposition was evident. Neoformed bone also appeared to be different at 12 d. In the control group, BMP-2 was positive in an intense area of bone trabeculae, whereas the alendronate-treated group showed TGF-β1 positive trabeculae and a greater bone area.
CONCLUSION Alendronate alters the immunolocalization of TGF-β1 and BMP-2 simultaneously, a condition that changes the usual histological aspects of the cartilage zone and impairs epiphysis growth and femur growth.
Collapse
|
8
|
Cigarette Smoke Induces the Risk of Metabolic Bone Diseases: Transforming Growth Factor Beta Signaling Impairment via Dysfunctional Primary Cilia Affects Migration, Proliferation, and Differentiation of Human Mesenchymal Stem Cells. Int J Mol Sci 2019; 20:ijms20122915. [PMID: 31207955 PMCID: PMC6628373 DOI: 10.3390/ijms20122915] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/27/2019] [Accepted: 06/12/2019] [Indexed: 12/18/2022] Open
Abstract
It is well established that smoking has detrimental effects on bone integrity and is a preventable risk factor for metabolic bone disorders. Following orthopedic surgeries, smokers frequently show delayed fracture healing associated with many complications, which results in prolonged hospital stays. One crucial factor responsible for fracture repair is the recruitment and differentiation of mesenchymal stem cells (MSCs) at early stages, a mechanism mediated by transforming growth factor β (TGF-β). Although it is known that smokers frequently have decreased TGF-β levels, little is known about the actual signaling occurring in these patients. We investigated the effect of cigarette smoke on TGF-β signaling in MSCs to evaluate which step in the pathway is affected by cigarette smoke extract (CSE). Single-cell-derived human mesenchymal stem cell line (SCP-1 cells) were treated with CSE concentrations associated with smoking up to 20 cigarettes a day. TGF-β signaling was analyzed using an adenovirus-based reporter assay system. Primary cilia structure and downstream TGF-β signaling modulators (Smad2, Smad3, and Smad4) were analyzed by Western blot and immunofluorescence staining. CSE exposure significantly reduced TGF-β signaling. Intriguingly, we observed that protein levels of phospho-Smad2/3 (active forms) as well as nuclear translocation of the phospho-Smad3/4 complex decreased after CSE exposure, phenomena that affected signal propagation. CSE exposure reduced the activation of TGF-β modulators under constitutive activation of TGF-β receptor type I (ALK5), evidencing that CSE affects signaling downstream of the ALK5 receptor but not the binding of the cytokine to the receptor itself. CSE-mediated TGF-β signaling impaired MSC migration, proliferation, and differentiation and ultimately affected endochondral ossification. Thus, we conclude that CSE-mediated disruption of TGF-β signaling in MSCs is partially responsible for delayed fracture healing in smokers.
Collapse
|
9
|
Vieira JS, Cunha EJ, de Souza JF, Sant'Ana RD, Zielak JC, Costa-Casagrande TA, Giovanini AF. Alendronate induces postnatal maxillary bone growth by stimulating intramembranous ossification and preventing premature cartilage mineralization in the midpalatal suture of newborn rats. Int J Oral Maxillofac Surg 2019; 48:1494-1503. [PMID: 31054875 DOI: 10.1016/j.ijom.2019.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 03/25/2019] [Accepted: 04/01/2019] [Indexed: 12/21/2022]
Abstract
Cleft palate is a common malformation of craniofacial development, and postnatal deficiencies in palate formation may occur. The aim of this study was to determine whether alendronate treatment could induce maxillary mineralization and thus reduce the need for surgical procedures. The effects of alendronate on maxillary bone development, the midpalatal suture, and the levels of transforming growth factor beta-1 (TGF-β1), bone morphogenetic protein 2 (BMP-2), collagen I and II, and V-ATPase were evaluated in newborn rats. Thirty newborn rats were placed in a control group and 30 in a group that received intraperitoneal alendronate (2.5 mg/kg/day). The animals were euthanized on day 7 or 12, and the heads were subjected to histological and immunohistochemical analyses. Specimens from rats that received alendronate presented larger bone matrix deposition in areas of intramembranous ossification of the maxillary bone when compared to controls. Furthermore, higher levels of TGF-β1, BMP-2, and collagen I were observed, whereas osteoclasts showed no V-ATPase. The alendronate group also showed higher levels of TGF-β1 and collagen II in the midpalatal suture, whereas BMP-2 levels were lower than in controls. These results coincided with an expansion of the chondroid. In conclusion, alendronate increased the intramembranous ossification in the maxillary bone in association with increased expression of TGF-β1, BMP-2, and collagen I and decreased V-ATPase. The drug induced an expansion of chondrocytes and a decrease in mineral bone deposition despite the high levels of TGF-β1 in this area. Alendronate may therefore be useful in the treatment of diseases affecting bone growth.
Collapse
Affiliation(s)
- J S Vieira
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil
| | - E J Cunha
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil
| | - J F de Souza
- Department of Stomatology, School of Dentistry, Federal University of Paraná, UFPR, Paraná, Brazil
| | - R D Sant'Ana
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil
| | - J C Zielak
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil
| | - T A Costa-Casagrande
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil
| | - A F Giovanini
- Graduate Programme in Clinical Dentistry, Positivo University, Curitiba, Paraná, Brazil.
| |
Collapse
|
10
|
Ripmeester EGJ, Timur UT, Caron MMJ, Welting TJM. Recent Insights into the Contribution of the Changing Hypertrophic Chondrocyte Phenotype in the Development and Progression of Osteoarthritis. Front Bioeng Biotechnol 2018; 6:18. [PMID: 29616218 PMCID: PMC5867295 DOI: 10.3389/fbioe.2018.00018] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/08/2018] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis (OA) is an extremely prevalent age-related condition. The economic and societal burden due to the cost of symptomatic treatment, inability to work, joint replacement, and rehabilitation is huge and increasing. Currently, there are no effective medical therapies that delay or reverse the pathological manifestations of OA. Current treatment options are, without exception, focused on slowing down progression of the disease to postpone total joint replacement surgery for as long as possible and keeping the associated pain and joint immobility manageable. Alterations in the articular cartilage chondrocyte phenotype might be fundamental in the pathological mechanisms of OA development. In many ways, the changing chondrocyte phenotype in osteoarthritic cartilage resembles the process of endochondral ossification as seen, for instance, in developing growth plates. However, the relative contribution of endochondral ossification to the changing chondrocyte phenotype in the development and progression of OA remains poorly described. In this review, we will discuss the current knowledge regarding the cartilage endochondral phenotypic changes occurring during OA development and progression, as well as the molecular and environmental effectors driving these changes. Understanding how these molecular mechanisms determine the chondrocyte cell fate in OA will be essential in enabling cartilage regenerative approaches in future treatments of OA.
Collapse
Affiliation(s)
- Ellen G J Ripmeester
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Ufuk Tan Timur
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Marjolein M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
11
|
Krajewska-Włodarczyk M, Owczarczyk-Saczonek A, Placek W, Osowski A, Wojtkiewicz J. Articular Cartilage Aging-Potential Regenerative Capacities of Cell Manipulation and Stem Cell Therapy. Int J Mol Sci 2018; 19:E623. [PMID: 29470431 PMCID: PMC5855845 DOI: 10.3390/ijms19020623] [Citation(s) in RCA: 16] [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: 01/28/2018] [Revised: 02/11/2018] [Accepted: 02/16/2018] [Indexed: 12/13/2022] Open
Abstract
Changes in articular cartilage during the aging process are a stage of natural changes in the human body. Old age is the major risk factor for osteoarthritis but the disease does not have to be an inevitable consequence of aging. Chondrocytes are particularly prone to developing age-related changes. Changes in articular cartilage that take place in the course of aging include the acquisition of the senescence-associated secretory phenotype by chondrocytes, a decrease in the sensitivity of chondrocytes to growth factors, a destructive effect of chronic production of reactive oxygen species and the accumulation of the glycation end products. All of these factors affect the mechanical properties of articular cartilage. A better understanding of the underlying mechanisms in the process of articular cartilage aging may help to create new therapies aimed at slowing or inhibiting age-related modifications of articular cartilage. This paper presents the causes and consequences of cellular aging of chondrocytes and the biological therapeutic outlook for the regeneration of age-related changes of articular cartilage.
Collapse
Affiliation(s)
- Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, Municipal Hospital in Olsztyn, 10-900 Olsztyn, Poland.
- Department of Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Waldemar Placek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Adam Osowski
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Laboratory for Regenerative Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| |
Collapse
|
12
|
Bundens G, Buckley A, Milton L, Behling K, Chmielewski S, Cho E, Lozano-Torres X, Selim A, Lackman R, George-Weinstein M, Miller L, D'Angelo M. Measuring clinically relevant endpoints in a serum-free, three-dimensional, primary cell culture system of human osteoarthritic articular chondrocytes. Exp Cell Res 2017; 357:310-319. [PMID: 28583763 DOI: 10.1016/j.yexcr.2017.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/09/2017] [Accepted: 06/01/2017] [Indexed: 11/24/2022]
Abstract
Osteoarthritis (OA) is characterized by degeneration of articular cartilage within the joint, inflammation and pain. The purpose of this study was to develop a primary, serum free cell culture system of human osteoarthritic articular chondrocytes (HOACs) with which to study manifestations of the disease process. Joint tissues were obtained from OA patients undergoing total knee arthroplasty (TKA). HOACs isolated from the femoral condyles and tibial plateau of the same side were combined, plated in three-dimensional, alginate beads and cultured for five days in serum, hormone and protein free medium. More living cells were obtained from the femoral condyles than the tibial plateau. The optimal plating density was 2.5 × 106 cells/ml of alginate. The amounts of DNA, RNA, proteoglycans and total collagen were similar in cultures prepared from the sides of least and greatest pathology. More type 1 than type 2 collagen was detected in the medium on days 2 and 5. A greater percentage of type 1 than type 2 collagen was degraded. The inflammatory cytokine interleukin-1 beta was present in the medium and alginate associated matrix. Although variation in the metabolic profiles between subjects was observed, HOACs from all patients continued to reflect the OA phenotype for five days in culture. This serum free, three-dimensional primary culture system of HOACs provides a platform with which to measure clinically relevant endpoints of OA and screen potential disease modifying OA therapeutics.
Collapse
Affiliation(s)
- Grace Bundens
- Cooper Medical School of Rowan University, 402 South Broadway, Camden, NJ 08103, USA.
| | - Andrea Buckley
- Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA.
| | - LaBraya Milton
- Cooper University Hospital, Three Cooper Plaza, Camden, NJ 08103, USA.
| | - Kathryn Behling
- Cooper Medical School of Rowan University, 402 South Broadway, Camden, NJ 08103, USA; Cooper University Hospital, Three Cooper Plaza, Camden, NJ 08103, USA.
| | - Sarah Chmielewski
- Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA.
| | - Ellen Cho
- Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA.
| | - Xiomara Lozano-Torres
- Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA.
| | - Abdulhafez Selim
- Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA.
| | - Richard Lackman
- Cooper University Hospital, Three Cooper Plaza, Camden, NJ 08103, USA.
| | | | - Lawrence Miller
- Cooper University Hospital, Three Cooper Plaza, Camden, NJ 08103, USA.
| | - Marina D'Angelo
- Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA.
| |
Collapse
|
13
|
Szychlinska MA, Stoddart MJ, D'Amora U, Ambrosio L, Alini M, Musumeci G. Mesenchymal Stem Cell-Based Cartilage Regeneration Approach and Cell Senescence: Can We Manipulate Cell Aging and Function? TISSUE ENGINEERING PART B-REVIEWS 2017; 23:529-539. [PMID: 28514935 DOI: 10.1089/ten.teb.2017.0083] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aging is the most prominent risk factor triggering several degenerative diseases, such as osteoarthritis (OA). Due to its poor self-healing capacity, once injured cartilage needs to be reestablished. This process might be approached through resorting to cell-based therapies and/or tissue engineering. Human mesenchymal stem cells (MSCs) represent a promising approach due to their chondrogenic differentiation potential. Presently, in vitro chondrogenic differentiation of MSCs is limited by two main reasons as follows: aging of MSCs, which determines the loss of cell proliferative and differentiation capacity and MSC-derived chondrocyte hypertrophic differentiation, which limits the use of these cells in cartilage tissue regeneration approach. The effect of aging on MSCs is fundamental for stem cell-based therapy development, especially in older subjects. In the present review we focus on homeostasis alterations occurring in MSC-derived chondrocytes during in vitro aging. Moreover, we deal with potential cell aging regulation approaches, such as cell stimulation through telomerase activators, mechanical strain, and epigenetic regulation. Future investigations in this field might provide new insights into innovative strategies for cartilage regeneration and potentially inspire novel therapeutic approaches for OA treatment.
Collapse
Affiliation(s)
- Marta A Szychlinska
- 1 Human Anatomy and Histology Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Catania, Italy
| | - Martin J Stoddart
- 2 Musculoskeletal Regeneration, AO Research Institute Davos , Davos Platz, Switzerland
| | - Ugo D'Amora
- 3 Institute of Polymers , Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Luigi Ambrosio
- 3 Institute of Polymers , Composites and Biomaterials, National Research Council of Italy, Naples, Italy .,4 Department of Chemical Science and Materials Technology, National Research Council of Italy , Rome, Italy
| | - Mauro Alini
- 2 Musculoskeletal Regeneration, AO Research Institute Davos , Davos Platz, Switzerland
| | - Giuseppe Musumeci
- 1 Human Anatomy and Histology Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Catania, Italy .,5 Department of Health, Institut des Etudes Universitaries , UniPoliSI, Veyras, Switzerland
| |
Collapse
|
14
|
Levin M, Udi Y, Solomonov I, Sagi I. Next generation matrix metalloproteinase inhibitors - Novel strategies bring new prospects. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017. [PMID: 28636874 DOI: 10.1016/j.bbamcr.2017.06.009] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Enzymatic proteolysis of cell surface proteins and extracellular matrix (ECM) is critical for tissue homeostasis and cell signaling. These proteolytic activities are mediated predominantly by a family of proteases termed matrix metalloproteinases (MMPs). The growing evidence in recent years that ECM and non-ECM bioactive molecules (e.g., growth factors, cytokines, chemokines, on top of matrikines and matricryptins) have versatile functions redefines our view on the roles matrix remodeling enzymes play in many physiological and pathological processes, and underscores the notion that ECM proteolytic reaction mechanisms represent master switches in the regulation of critical biological processes and govern cell behavior. Accordingly, MMPs are not only responsible for direct degradation of ECM molecules but are also key modulators of cardinal bioactive factors. Many attempts were made to manipulate ECM degradation by targeting MMPs using small peptidic and organic inhibitors. However, due to the high structural homology shared by these enzymes, the majority of the developed compounds are broad-spectrum inhibitors affecting the proteolytic activity of various MMPs and other zinc-related proteases. These inhibitors, in many cases, failed as therapeutic agents, mainly due to the bilateral role of MMPs in pathological conditions such as cancer, in which MMPs have both pro- and anti-tumorigenic effects. Despite the important role of MMPs in many human diseases, none of the broad-range synthetic MMP inhibitors that were designed have successfully passed clinical trials. It appears that, designing highly selective MMP inhibitors that are also effective in vivo, is not trivial. The challenges related to designing selective and effective metalloprotease inhibitors, are associated in part with the aforesaid high structural homology and the dynamic nature of their protein scaffolds. Great progress was achieved in the last decade in understanding the biochemistry and biology of MMPs activity. This knowledge, combined with lessons from the past has drawn new "boundaries" for the development of the next-generation MMP inhibitors. These novel agents are currently designed to be highly specific, capable to discriminate between the homologous MMPs and ideally administered as a short-term topical treatment. In this review we discuss the latest progress in the fields of MMP inhibitors in terms of structure, function and their specific activity. The development of novel highly specific inhibitors targeting MMPs paves the path to study complex biological processes associated with ECM proteolysis in health and disease. This article is part of a Special Issue entitled: Matrix Metalloproteinases edited by Rafael Fridman.
Collapse
Affiliation(s)
- Maxim Levin
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yael Udi
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY 10065, USA
| | - Inna Solomonov
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
15
|
Bai X, Li YY, Zhang HY, Wang F, He HL, Yao JC, Liu L, Li SS. Role of matrix metalloproteinase-9 in transforming growth factor-β1-induced epithelial-mesenchymal transition in esophageal squamous cell carcinoma. Onco Targets Ther 2017; 10:2837-2847. [PMID: 28652766 PMCID: PMC5476773 DOI: 10.2147/ott.s134813] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT) is thought to be a crucial event during the early metastasis of tumor cells. Transforming growth factor (TGF)-β1 is involved in the process of EMT in a variety of human malignancies. Matrix metalloproteinase (MMP)-9 plays an important role in tumor invasion and metastasis, and its expression is regulated by various growth factors, including TGF-β1, in different cell types. To date, the role of MMP-9 in TGF-β1-induced EMT in esophageal squamous cell carcinoma (ESCC) remains unclear. In this study, we aimed to elucidate the mechanism underlying MMP-9-mediated TGF-β1 induction of EMT in ESCC. We analyzed the expression of MMP-9, E-cadherin, and vimentin, in ESCC cells (EC-1), before and after the treatment with exogenous TGF-β1 or a broad spectrum MMP inhibitor, GM6001. Additionally, we analyzed the activity of MMP-9 in these cells and performed MMP-9 knockdown experiments. The results obtained in this study demonstrated that the treatment of EC-1 cells with TGF-β1 can induce EMT, together with the upregulation of vimentin and downregulation of E-cadherin expression in a time-dependent manner. The treatment with GM6001 was shown to attenuate TGF-β1-induced EMT. Furthermore, the exposure of EC-1 cells to TGF-β1 increased the expression and activity of MMP-9, while MMP-9 knockdown blocked TGF-β1-induced EMT and inhibited cell invasiveness and migration. Additionally, treatment with the recombinant human MMP-9 was shown to induce EMT and enhance ESCC cell invasion and metastasis. The obtained data suggest that the regulation of MMP-9 by TGF-β1 may represent a novel mechanism underlying TGF-β1-induced EMT in ESCC.
Collapse
Affiliation(s)
- Xue Bai
- Department of Pathology, Basic Medical College of Zhengzhou University
| | - Yun-Yun Li
- Department of Pathology, Basic Medical College of Zhengzhou University.,Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Hong-Yan Zhang
- Department of Pathology, Basic Medical College of Zhengzhou University
| | - Feng Wang
- Department of Pathology, Basic Medical College of Zhengzhou University
| | - Hong-Liu He
- Department of Pathology, Basic Medical College of Zhengzhou University
| | - Jin-Chao Yao
- Department of Pathology, Basic Medical College of Zhengzhou University
| | - Ling Liu
- Department of Pathology, Basic Medical College of Zhengzhou University
| | - Shan-Shan Li
- Department of Pathology, Basic Medical College of Zhengzhou University
| |
Collapse
|
16
|
Yamamoto Y, Ihara M. Disruption of transforming growth factor-β superfamily signaling: A shared mechanism underlying hereditary cerebral small vessel disease. Neurochem Int 2016; 107:211-218. [PMID: 28034724 DOI: 10.1016/j.neuint.2016.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/02/2016] [Accepted: 12/07/2016] [Indexed: 11/20/2022]
Abstract
Cerebral small vessel disease (SVD) is not only one of the leading causes of cognitive impairment but also an important contributory factor in Alzheimer's disease. SVD and related white matter changes are common in the elderly, but the underlying pathogenic mechanism remains unclear. The end-stage pathology of SVD often involves replacement of vascular smooth muscle cells with collagenous or other nontensile fibrillary material. Recent studies on hereditary SVD have revealed a close relationship between small vessel pathology and disruption of transforming growth factor-β (TGF-β) superfamily signaling. TGF-β superfamily members, such as TGF-β and bone morphogenetic proteins, are multifunctional proteins that regulate production of extracellular matrix proteins, which in turn control the bioavailability of TGF-β superfamily members and modulate their signaling activities. This article reviews hereditary disorders with small vessel pathology and their relation to TGF-β superfamily signaling.
Collapse
Affiliation(s)
- Yumi Yamamoto
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, 5-7-1 Fujishirodai, Suita, Osaka, 565-8565, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, 5-7-1 Fujishirodai, Suita, Osaka, 565-8565, Japan.
| |
Collapse
|
17
|
Matrix metalloproteinases in inflammatory bowel disease: an update. Mediators Inflamm 2015; 2015:964131. [PMID: 25948887 PMCID: PMC4408746 DOI: 10.1155/2015/964131] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 09/07/2014] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are known to be upregulated in inflammatory bowel disease (IBD) and other inflammatory conditions, but while their involvement is clear, their role in many settings has yet to be determined. Studies of the involvement of MMPs in IBD since 2006 have revealed an array of immune and stromal cells which release the proteases in response to inflammatory cytokines and growth factors. Through digestion of the extracellular matrix and cleavage of bioactive proteins, a huge diversity of roles have been revealed for the MMPs in IBD, where they have been shown to regulate epithelial barrier function, immune response, angiogenesis, fibrosis, and wound healing. For this reason, MMPs have been recognised as potential biomarkers for disease activity in IBD and inhibition remains a huge area of interest. This review describes new roles of MMPs in the pathophysiology of IBD and suggests future directions for the development of treatment strategies in this condition.
Collapse
|
18
|
Fontana P, Genesio R, Casertano A, Cappuccio G, Mormile A, Nitsch L, Iolascon A, Andria G, Melis D. Loeys–Dietz syndrome type 4, caused by chromothripsis, involving the TGFB2 gene. Gene 2014; 538:69-73. [DOI: 10.1016/j.gene.2014.01.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 10/29/2013] [Accepted: 01/03/2014] [Indexed: 12/20/2022]
|
19
|
Albro MB, Nims RJ, Cigan AD, Yeroushalmi KJ, Alliston T, Hung CT, Ateshian GA. Accumulation of exogenous activated TGF-β in the superficial zone of articular cartilage. Biophys J 2013; 104:1794-804. [PMID: 23601326 DOI: 10.1016/j.bpj.2013.02.052] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 01/07/2013] [Accepted: 02/19/2013] [Indexed: 11/28/2022] Open
Abstract
It was recently demonstrated that mechanical shearing of synovial fluid (SF), induced during joint motion, rapidly activates latent transforming growth factor β (TGF-β). This discovery raised the possibility of a physiological process consisting of latent TGF-β supply to SF, activation via shearing, and transport of TGF-β into the cartilage matrix. Therefore, the two primary objectives of this investigation were to characterize the secretion rate of latent TGF-β into SF, and the transport of active TGF-β across the articular surface and into the cartilage layer. Experiments on tissue explants demonstrate that high levels of latent TGF-β1 are secreted from both the synovium and all three articular cartilage zones (superficial, middle, and deep), suggesting that these tissues are capable of continuously replenishing latent TGF-β to SF. Furthermore, upon exposure of cartilage to active TGF-β1, the peptide accumulates in the superficial zone (SZ) due to the presence of an overwhelming concentration of nonspecific TGF-β binding sites in the extracellular matrix. Although this response leads to high levels of active TGF-β in the SZ, the active peptide is unable to penetrate deeper into the middle and deep zones of cartilage. These results provide strong evidence for a sequential physiologic mechanism through which SZ chondrocytes gain access to active TGF-β: the synovium and articular cartilage secrete latent TGF-β into the SF and, upon activation, TGF-β transports back into the cartilage layer, binding exclusively to the SZ.
Collapse
Affiliation(s)
- Michael B Albro
- Department of Mechanical Engineering, Columbia University, New York, New York, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Shivakumar HR, Batra J, Upasi AP, Rai KK. Evaluation of Elongated Styloid Process in Patients with Oral Submucous Fibrosis Using Panoramic Radiographs. J Maxillofac Oral Surg 2013. [PMID: 26225028 DOI: 10.1007/s12663-013-0529-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Oral submucous fibrosis (OSF) is an insidious disease affecting the oral cavity, pharynx, and upper digestive tract. It is characterized by a juxtaepithelial inflammatory reaction followed by fibroelastic change in the lamina propria and associated epithelial atrophy. Higher levels of TGF-β present in patients with OSF could be responsible for impetus to remnants of Reichert's cartilage present in styloid complex leading to partial or complete ossification of associated ligaments. So, a study was conducted to evaluate the elongation of the styloid process in patients with OSF by using panoramic radiographs. MATERIALS AND METHODS Panoramic radiographs of patients with OSF were studied from 2007-2011. The apparent lengths of styloid process were measured with the help of divider and steel metric ruler. The length of the styloid process and/or ossification of stylomandibular ligaments which were longer than 30 mm were considered. RESULTS Out of 47 patients, 35 patients (34 males & 1 female) met the inclusion criteria. Eleven patients (31.4%) were found to have elongated styloid processes which included 10 male patients and 1 female patient. CONCLUSION It had been estimated that between 2 % and 4% of the general population presents radiographic evidence of an ossified portion of the styloid complex. The high incidence of elongation of styloid process (31.4 %) in patients with oral submucous fibrosis highlights that progressive OSF might have some influence on elongation of styloid process.
Collapse
Affiliation(s)
- H R Shivakumar
- Department of Oral and Maxillofacial Surgery, Bapuji Dental College and Hospital, Davangere, Karnataka 577004 India
| | - Jitender Batra
- Department of Oral and Maxillofacial Surgery, Post Graduate Institute of Dental Sciences, Rohtak, Haryana 124001 India
| | - Amarnath P Upasi
- Department of Oral and Maxillofacial Surgery, Bapuji Dental College and Hospital, Davangere, Karnataka 577004 India
| | - Kirthi Kumar Rai
- Department of Oral and Maxillofacial Surgery, Bapuji Dental College and Hospital, Davangere, Karnataka 577004 India
| |
Collapse
|
21
|
Morrison C, Mancini S, Cipollone J, Kappelhoff R, Roskelley C, Overall C. Microarray and proteomic analysis of breast cancer cell and osteoblast co-cultures: role of osteoblast matrix metalloproteinase (MMP)-13 in bone metastasis. J Biol Chem 2011; 286:34271-85. [PMID: 21784845 PMCID: PMC3190775 DOI: 10.1074/jbc.m111.222513] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 07/20/2011] [Indexed: 12/20/2022] Open
Abstract
Dynamic reciprocal interactions between a tumor and its microenvironment impact both the establishment and progression of metastases. These interactions are mediated, in part, through proteolytic sculpting of the microenvironment, particularly by the matrix metalloproteinases, with both tumors and stroma contributing to the proteolytic milieu. Because bone is one of the predominant sites of breast cancer metastases, we used a co-culture system in which a subpopulation of the highly invasive human breast cancer cell line MDA-MB-231, with increased propensity to metastasize to bone, was overlaid onto a monolayer of differentiated osteoblast MC3T3-E1 cells in a mineralized osteoid matrix. CLIP-CHIP® microarrays identified changes in the complete protease and inhibitor expression profile of the breast cancer and osteoblast cells that were induced upon co-culture. A large increase in osteoblast-derived MMP-13 mRNA and protein was observed. Affymetrix analysis and validation showed induction of MMP-13 was initiated by soluble factors produced by the breast tumor cells, including oncostatin M and the acute response apolipoprotein SAA3. Significant changes in the osteoblast secretomes upon addition of MMP-13 were identified by degradomics from which six novel MMP-13 substrates with the potential to functionally impact breast cancer metastasis to bone were identified and validated. These included inactivation of the chemokines CCL2 and CCL7, activation of platelet-derived growth factor-C, and cleavage of SAA3, osteoprotegerin, CutA, and antithrombin III. Hence, the influence of breast cancer metastases on the bone microenvironment that is executed via the induction of osteoblast MMP-13 with the potential to enhance metastases growth by generating a microenvironmental amplifying feedback loop is revealed.
Collapse
Affiliation(s)
- Charlotte Morrison
- From the Centre for Blood Research and
- Departments of Oral Biological and Medical Sciences
| | - Stephanie Mancini
- Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Jane Cipollone
- Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Reinhild Kappelhoff
- From the Centre for Blood Research and
- Departments of Oral Biological and Medical Sciences
| | - Calvin Roskelley
- Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Christopher Overall
- From the Centre for Blood Research and
- Departments of Oral Biological and Medical Sciences
- Biochemistry and Molecular Biology, and
| |
Collapse
|
22
|
Attia M, Mohamed A, Huet E, Eric H, Delbé J, Jean D, Ledoux D, Dominique L, Menashi S, Suzanne M, Martelly I, Isabelle M. Extracellular matrix metalloproteinase inducer (EMMPRIN/CD147) as a novel regulator of myogenic cell differentiation. J Cell Physiol 2010; 226:141-9. [PMID: 20648565 DOI: 10.1002/jcp.22315] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Matrix metalloproteinases (MMPs) are thought to play an important role in skeletal muscle cell growth and differentiation. In view of the MMP inducing function of EMMPRIN/CD147, its role in myogenic cell differentiation was investigated. EMMPRIN level increased during differentiation of both rat primary myoblasts derived from satellite cells and mouse C2.7 myogenic cells and was associated with an alteration in its molecular forms. In parallel, expression of pro-MMP-9 gradually decreased and that of pro-MMP-2 and active MMP-2 increased. While small interfering RNA (siRNA) inhibition of EMMPRIN expression accelerated cell differentiation, exogenously added recombinant EMMPRIN inhibited differentiation by an MMP-mediated mechanism, as the MMP inhibitor marimastat abrogated EMMPRIN's effect. Our results further suggest that EMMPRIN regulates differentiation through an MMP activation of transforming growth factor beta (TGFβ), a known inhibitor of myoblast's differentiation, as the increased activation and signaling of TGFβ by EMMPRIN was attenuated in the presence of marimastat. EMMPRIN inhibition may thus represent a novel strategy in the treatment of muscular degenerative disorders.
Collapse
Affiliation(s)
- Mohamed Attia
- Laboratoire CRRET, CNRS EAC 7149, Université Paris-Est Créteil, Créteil, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Palmer GD, Piton AH, Thant LM, Oliveira SM, D’Angelo M, Attur MG, Abramson SB, Teixeira CC. F-spondin regulates chondrocyte terminal differentiation and endochondral bone formation. J Orthop Res 2010; 28:1323-9. [PMID: 20839318 PMCID: PMC3245523 DOI: 10.1002/jor.21130] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This study examines the role of F-spondin, an extracellular matrix protein of osteoarthritic cartilage, during chondrocyte maturation in embryonic growth plate cartilage. In chick tibia, F-spondin expression localized to the hypertrophic and calcified zones of the growth plate. Functional studies using tibial organ cultures indicated that F-spondin inhibited (∼35%, p = 0.02), and antibodies to F-spondin increased (∼30%, p < 0.1) longitudinal limb growth relative to untreated controls. In cell cultures, induction of chondrocyte maturation, by retinoic acid (RA) or transforming growth factor (TGF)-β treatment led to a significant upregulation of F-spondin (p < 0.05). F-spondin transfection increased mineral deposition, alkaline phosphatase (AP) and matrix metalloproteinase (MMP)-13 mRNA levels (p < 0.05), and AP activity following RA stimulation, compared to mock transfected controls. Using AP as a differentiation marker we then investigated the mechanism of F-spondin promaturation effects. Blocking endogenous F-spondin via its thrombospondin (TSR) domain inhibited RA induced AP activity 40% compared to controls (p < 0.05). The stimulatory effect of F-spondin on AP expression was also inhibited following depletion of TGF-β from culture supernatants. Our findings indicate that F-spondin is expressed in embryonic cartilage, where it has the capacity to enhance chondrocyte terminal differentiation and mineralization via interactions in its TSR domain and TGF-β dependent pathways.
Collapse
Affiliation(s)
- Glyn D. Palmer
- Division of Rheumatology, New York University School of Medicine, Hospital for Joint Diseases, New York, New York
| | - Alejandro H. Piton
- Department of Anatomy and Center for Chronic Disorders of Aging, Philadelphia, College of Osteopathic Medicine, Philadelphia, Pennsylvania 19131
| | - Lwin Mon Thant
- Department of Anatomy and Center for Chronic Disorders of Aging, Philadelphia, College of Osteopathic Medicine, Philadelphia, Pennsylvania 19131
| | - Serafim M. Oliveira
- Department of Anatomy and Center for Chronic Disorders of Aging, Philadelphia, College of Osteopathic Medicine, Philadelphia, Pennsylvania 19131
| | - Marina D’Angelo
- Department of Anatomy and Center for Chronic Disorders of Aging, Philadelphia, College of Osteopathic Medicine, Philadelphia, Pennsylvania 19131
| | - Mukundan G. Attur
- Division of Rheumatology, New York University School of Medicine, Hospital for Joint Diseases, New York, New York
| | - Steven B. Abramson
- Division of Rheumatology, New York University School of Medicine, Hospital for Joint Diseases, New York, New York
| | - Cristina C. Teixeira
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York 10010,Department of Anatomy and Center for Chronic Disorders of Aging, Philadelphia, College of Osteopathic Medicine, Philadelphia, Pennsylvania 19131
| |
Collapse
|
24
|
Taylor MA, Parvani JG, Schiemann WP. The pathophysiology of epithelial-mesenchymal transition induced by transforming growth factor-beta in normal and malignant mammary epithelial cells. J Mammary Gland Biol Neoplasia 2010; 15:169-90. [PMID: 20467795 PMCID: PMC3721368 DOI: 10.1007/s10911-010-9181-1] [Citation(s) in RCA: 185] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Accepted: 04/22/2010] [Indexed: 12/14/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is an essential process that drives polarized, immotile mammary epithelial cells (MECs) to acquire apolar, highly migratory fibroblastoid-like features. EMT is an indispensable process that is associated with normal tissue development and organogenesis, as well as with tissue remodeling and wound healing. In stark contrast, inappropriate reactivation of EMT readily contributes to the development of a variety of human pathologies, particularly those associated with tissue fibrosis and cancer cell invasion and metastasis, including that by breast cancer cells. Although metastasis is unequivocally the most lethal aspect of breast cancer and the most prominent feature associated with disease recurrence, the molecular mechanisms whereby EMT mediates the initiation and resolution of breast cancer metastasis remains poorly understood. Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine that is intimately involved in regulating numerous physiological processes, including cellular differentiation, homeostasis, and EMT. In addition, TGF-beta also functions as a powerful tumor suppressor in MECs, whose neoplastic development ultimately converts TGF-beta into an oncogenic cytokine in aggressive late-stage mammary tumors. Recent findings have implicated the process of EMT in mediating the functional conversion of TGF-beta during breast cancer progression, suggesting that the chemotherapeutic targeting of EMT induced by TGF-beta may offer new inroads in ameliorating metastatic disease in breast cancer patients. Here we review the molecular, cellular, and microenvironmental factors that contribute to the pathophysiological activities of TGF-beta during its regulation of EMT in normal and malignant MECs.
Collapse
Affiliation(s)
- Molly A Taylor
- Case Comprehensive Cancer Center, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
25
|
Rodríguez D, Morrison CJ, Overall CM. Matrix metalloproteinases: what do they not do? New substrates and biological roles identified by murine models and proteomics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1803:39-54. [PMID: 19800373 DOI: 10.1016/j.bbamcr.2009.09.015] [Citation(s) in RCA: 369] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 09/17/2009] [Accepted: 09/24/2009] [Indexed: 12/14/2022]
Abstract
The biological roles of the matrix metalloproteinases (MMPs) have been traditionally associated with the degradation and turnover of most of the components of the extracellular matrix (ECM). This functional misconception has been used for years to explain the involvement of the MMP family in developmental processes, cell homeostasis and disease, and led to clinical trials of MMP inhibitors for the treatment of cancer that failed to meet their endpoints and cast a shadow on MMPs as druggable targets. Accumulated evidence from a great variety of post-trial MMP degradomics studies, ranging from transgenic models to recent state-of-the-art proteomics screens, is changing the dogma about MMP functions. MMPs regulate cell behavior through finely tuned and tightly controlled proteolytic processing of a large variety of signaling molecules that can also have beneficial effects in disease resolution. Moreover, net proteolytic activity relies upon direct interactions between the different protease and protease inhibitor families, interconnected in a complex protease web, with MMPs acting as key nodal components. Such complexity renders simple interpretation of Mmp knockout mice very difficult. Indeed, the phenotype of these models reveals the response of a complex system to the loss of one protease rather than necessarily a direct effect of the lack of functional activity of a protease. Such a shift in the MMP functional paradigm, together with the difficulties associated with current methods of studying proteases this highlights the need for new high content degradomics approaches to uncover and annotate MMP activities in vivo and identify novel interactions within the protease web. Integration of these techniques with specifically designed animal models for final validation should lay the foundations for the development of new inhibitors that specifically target disease-related MMPs and/or their upstream effectors that cause deleterious effects in disease, while sparing MMP functions that are protective.
Collapse
Affiliation(s)
- David Rodríguez
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | | | | |
Collapse
|
26
|
Estrogen/estrogen receptor alpha signaling in mouse posterofrontal cranial suture fusion. PLoS One 2009; 4:e7120. [PMID: 19771170 PMCID: PMC2743190 DOI: 10.1371/journal.pone.0007120] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Accepted: 08/25/2009] [Indexed: 01/22/2023] Open
Abstract
Background While premature suture fusion, or craniosynostosis, is a relatively common condition, the cause is often unknown. Estrogens are associated with growth plate fusion of endochondral bones. In the following study, we explore the previously unknown significance of estrogen/estrogen receptor signaling in cranial suture biology. Methodology/Principal Findings Firstly, estrogen receptor (ER) expression was examined in physiologically fusing (posterofrontal) and patent (sagittal) mouse cranial sutures by quantitative RT-PCR. Next, the cranial suture phenotype of ER alpha and ER beta knockout (αERKO, βERKO) mice was studied. Subsequently, mouse suture-derived mesenchymal cells (SMCs) were isolated; the effects of 17-β estradiol or the estrogen antagonist Fulvestrant on gene expression, osteogenic and chondrogenic differentiation were examined in vitro. Finally, in vivo experiments were performed in which Fulvestrant was administered subcutaneously to the mouse calvaria. Results showed that increased ERα but not ERβ transcript abundance temporally coincided with posterofrontal suture fusion. The αERKO but not βERKO mouse exhibited delayed posterofrontal suture fusion. In vitro, addition of 17-β estradiol enhanced both osteogenic and chondrogenic differentiation in suture-derived mesenchymal cells, effects reversible by Fulvestrant. Finally, in vivo application of Fulvestrant significantly diminished calvarial osteogenesis, inhibiting suture fusion. Conclusions/Significance Estrogen signaling through ERα but not ERβ is associated with and necessary for normal mouse posterofrontal suture fusion. In vitro studies suggest that estrogens may play a role in osteoblast and/or chondrocyte differentiation within the cranial suture complex.
Collapse
|
27
|
Differential effects of TGF-beta1 and TGF-beta3 on chondrogenesis in posterofrontal cranial suture-derived mesenchymal cells in vitro. Plast Reconstr Surg 2009; 123:31-43. [PMID: 19116522 DOI: 10.1097/prs.0b013e3181904c19] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Transforming growth factor (TGF)-beta1 has been associated with cranial suture fusion, whereas TGF-beta3 has been associated with suture patency. The mouse posterofrontal suture, analogous to the human metopic suture, fuses through endochondral ossification. METHODS TGF-beta1 and TGF-beta3 expression in the posterofrontal suture was examined by immunohistochemistry. Next, the authors established cultures of suture-derived mesenchymal cells from the posterofrontal suture and examined the cellular responses to TGF-beta1 and TGF-beta3. Proliferation in response to TGF-beta isoforms was examined by bromodeoxyuridine incorporation. High-density micromass culture of posterofrontal mesenchymal cells was used to study the effect of TGF-beta1 and TGF-beta3 on chondrogenic differentiation. RESULTS TGF-beta1 but not TGF-beta3 protein was highly expressed in chondrocytes within the posterofrontal suture. Significant increases in posterofrontal cell proliferation were observed with TGF-beta3 but not TGF-beta1. TGF-beta1 led to significant increases in chondrogenic-specific gene expression (including Sox9, Col II, Aggrecan, and Col X) as compared with moderate effects of TGF-beta3. TGF-beta1 increased cellular adhesion molecule expression (N-cadherin and fibronectin) and promoted cellular condensation, whereas TGF-beta3 increased cellular proliferation (PCNA expression). Finally, TGF-beta1 and, to a lesser extent, TGF-beta3 induced the expression of fibroblast growth factors (FGF-2 and FGF-18). CONCLUSIONS TGF-beta1 and TGF-beta3 exhibit marked differences in their effects on chondrogenesis in posterfrontal suture-derived mesenchymal cells, influencing different stages of chondrogenic differentiation. TGF-beta3 significantly increased cellular proliferation, whereas TGF-beta1 induced precartilage condensation, promoting chondrocyte differentiation.
Collapse
|
28
|
Transforming growth factor-beta1 stimulates chondrogenic differentiation of posterofrontal suture-derived mesenchymal cells in vitro. Plast Reconstr Surg 2009; 122:1649-1659. [PMID: 19050517 DOI: 10.1097/prs.0b013e31818cbf44] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Evidence from animal studies has associated transforming growth factor (TGF)-beta signaling with both normal and premature cranial suture fusion. However, the mechanisms whereby this pleiotropic cytokine mediates suture fusion remain uncertain. The authors established cultures of suture-derived mesenchymal cells from normally fusing (posterofrontal) and patent (sagittal) sutures and examined the in vitro effects of TGF-beta1 on these distinct cell populations. METHODS Skulls were harvested from 80 5-day-old mice. Posterofrontal and sagittal sutures were dissected, and cultures of suture-derived mesenchymal cells were established. The mitogenic, osteogenic, and chondrogenic effects of recombinant TGF-beta1 were then assessed on posterofrontal and sagittal suture-derived mesenchymal cells (1 to 10 ng/ml). Quantitative real-time polymerase chain reaction was used to examine the effects of TGF-beta1 on gene expression. RESULTS TGF-beta1 significantly decreased proliferation of both posterofrontal and sagittal suture-derived mesenchymal cells, by bromodeoxyuridine incorporation assays (n = 6). TGF-beta1 also inhibited osteogenesis in both suture-derived mesenchymal cells determined by alkaline phosphatase activity and mineralization (n = 3 for all assays). During chondrogenic differentiation, TGF-beta1 markedly increased expression of chondrocyte-specific gene markers in posterofrontal suture-derived mesenchymal cells (Sox9, Col II, Aggrecan, and Col X) (p <or= 0.05). In contrast, TGF-beta1 did not increase chondrocyte-specific gene expression in sagittal suture-derived mesenchymal cells (n = 3). CONCLUSIONS Posterofrontal suture-derived mesenchymal cells retain significant capability for both osteogenic and chondrogenic differentiation in vitro. TGF-beta1 induces in vitro chondrogenesis in posterofrontal but not sagittal suture-derived mesenchymal cells. Future studies will focus on elucidating the mechanisms whereby TGF-beta signaling mediates chondrogenesis in posterofrontal suture-derived mesenchymal cells.
Collapse
|
29
|
Extracorporeal Shock Wave-Mediated Changes in Proliferation, Differentiation, and Gene Expression of Human Osteoblasts. ACTA ACUST UNITED AC 2008; 65:1402-10. [DOI: 10.1097/ta.0b013e318173e7c2] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
30
|
Yang M, Wang X, Zhang L, Yu C, Zhang B, Cole W, Cavey G, Davidson P, Gibson G. Demonstration of the interaction of transforming growth factor beta 2 and type X collagen using a modified tandem affinity purification tag. J Chromatogr B Analyt Technol Biomed Life Sci 2008; 875:493-501. [PMID: 18952512 PMCID: PMC2653427 DOI: 10.1016/j.jchromb.2008.10.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Revised: 08/13/2008] [Accepted: 10/01/2008] [Indexed: 12/20/2022]
Abstract
Like other members of the transforming growth factor beta (TGF-beta) family of growth factors, the biological activity of TGF-beta2 is believed to be regulated by the formation and dissociation of multiprotein complexes. To isolate the molecular complex formed by TGF-beta2 secreted by hypertrophic chondrocytes we have used expression of TGF-beta2 fused with the humanized, tandem affinity purification (hTAP) tag and mass spectrometry for the identification of interacting proteins. The hTAP synthetic gene was assembled by systematically replacing the rare codons of the original TAP tag with codons most preferred in highly expressed human genes to circumvent the poor translation efficiency of the original TAP tag in animal cells. TGF-beta2 was shown to interact with Type X collagen and this interaction confirmed using V5 tagged TGF-beta2. Functional interaction was suggested by the inhibition of TGF-beta2 activity by type X collagen in culture and the influence of a mutation in type X collagen on the distribution of TGF-beta2 in growth cartilage.
Collapse
Affiliation(s)
- Maozhou Yang
- Bone and Joint Center, Henry Ford Hospital, Michigan, 48202
| | - Xinli Wang
- Bone and Joint Center, Henry Ford Hospital, Michigan, 48202
| | - Liang Zhang
- Bone and Joint Center, Henry Ford Hospital, Michigan, 48202
| | - Chiyang Yu
- Bone and Joint Center, Henry Ford Hospital, Michigan, 48202
| | - Bingbing Zhang
- Bone and Joint Center, Henry Ford Hospital, Michigan, 48202
| | - William Cole
- Division of Genetic and Genomic Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Grey Cavey
- Van Andel Institute, 333 Bostick NE, Grand Rapids, Michigan 49503
| | - Paula Davidson
- Van Andel Institute, 333 Bostick NE, Grand Rapids, Michigan 49503
| | - Gary Gibson
- Bone and Joint Center, Henry Ford Hospital, Michigan, 48202
| |
Collapse
|
31
|
Illman SA, Lohi J, Keski-Oja J. Epilysin (MMP-28)--structure, expression and potential functions. Exp Dermatol 2008; 17:897-907. [PMID: 18803661 DOI: 10.1111/j.1600-0625.2008.00782.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Epilysin (MMP-28) is the newest member of the matrix metalloproteinase (MMP) family of extracellular proteases. Together the MMPs can degrade almost all components of the extracellular matrix (ECM). MMPs also regulate cell behaviour by releasing growth factors and biologically active peptides from the ECM by modulating cell surface receptors and adhesion molecules and by regulating the activity of mediators of the inflammatory pathways. Epilysin differs from most other MMPs as it is expressed in a number of normal tissues, suggestive of functions in tissue homeostasis. The epilysin homologue in Xenopus laevis (XMMP-28) is expressed in neural tissues, where it cleaves the neural cell adhesion molecule. Enhanced expression of epilysin has been observed in basal keratinocytes during wound healing and in different forms of cancer. There are, however, also reports on the downregulation of epilysin in malignant cells. The roles of epilysin in cancer seem to vary based on tumor type and stage of the disease. Importantly, epilysin can induce stable epithelial to mesenchymal transition (EMT) when overexpressed in epithelial lung carcinoma cells. Transforming growth factor beta (TGF-beta) is a crucial mediator of this process, which was characterized by the loss of E-cadherin and increased cell migration and invasion. Current results suggest a plausible interaction between epilysin and TGF-beta also under physiological circumstances, where epilysin activity may not induce EMT but, instead, trigger less permanent changes in TGF-beta signalling and cell motility.
Collapse
Affiliation(s)
- Sara A Illman
- Department of Pathology, Haartman Institute and Biomedicum Helsinki, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | | | | |
Collapse
|
32
|
Illman SA, Lehti K, Keski-Oja J, Lohi J. Epilysin (MMP-28) induces TGF-beta mediated epithelial to mesenchymal transition in lung carcinoma cells. J Cell Sci 2006; 119:3856-65. [PMID: 16940349 DOI: 10.1242/jcs.03157] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Epilysin (MMP-28) is the newest member of the matrix metalloproteinase (MMP) family. Although it is expressed in a number of tissues, no biological substrates or functions for this enzyme have been identified yet. We have expressed recombinant epilysin in A549 lung adenocarcinoma cells and found that this resulted in stable and irreversible epithelial to mesenchymal transition (EMT) accompanied by loss of cell surface E-cadherin, proteolytic processing of latent TGF-beta-complexes and increased levels of active TGF-beta. The cascade of events leading to the onset of EMT is prevented by the MMP inhibitor GM6001 or antibodies neutralizing the activity of TGF-beta. Once EMT had occurred the cell phenotype could, however, not be reversed by the MMP-inhibitor. Importantly, the expression of epilysin also resulted in upregulation of MT1-MMP and gelatinase-B (MMP-9) and in the collagen invasive activity of A549 cells. Further, we found that epilysin and the recombinant hemopexin domain were targeted to the surface of epithelial cells. This cell surface interaction was sensitive to the proteolytic activity of MT1-MMP, and was lost after EMT. Current results indicate that epilysin can induce EMT and cell invasion through a TGF-beta-dependent mechanism suggesting novel biological roles for this enzyme in the regulation of epithelial cell function and in the induction of carcinogenesis.
Collapse
Affiliation(s)
- Sara A Illman
- Department of Pathology, Haartman Institute and Biomedicum Helsinki, University of Helsinki and Helsinki University Hospital, FIN-00014 Helsinki, Finland
| | | | | | | |
Collapse
|
33
|
van Donkelaar CC, Huiskes R. The PTHrP-Ihh feedback loop in the embryonic growth plate allows PTHrP to control hypertrophy and Ihh to regulate proliferation. Biomech Model Mechanobiol 2006; 6:55-62. [PMID: 16691414 DOI: 10.1007/s10237-006-0035-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Accepted: 01/06/2006] [Indexed: 10/24/2022]
Abstract
Growth plate and long bone development is governed by biochemical signaling pathways of which the PTHrP-Ihh system is the best known. Other factors, such as BMPs, FGFs and mechanical loading, may interact with this system. This study aims at elucidating the relative importance of PTHrP and Ihh for controlling proliferation, and hypertrophy in fetal growth plate cartilage. We assessed the question why reduced Ihh expression leads to more pronounced effects on the number of non-hypertrophic cells and total bone formation, compared to PTHrP down-regulation. Using few basic equations, constituted from literature data, this paper shows how the PTHrP-Ihh feedback system can control different aspects of tissue differentiation at distinct locations. In particular, it is shown that (mechanical or biochemical) perturbations will affect proliferation via Ihh-related parameters, whereas changes in PTHrP-related parameters selectively interact with hypertrophy. This is contra-intuitive, since PTHrP acts to keep cells proliferating. In this context, the critical PTHrP level for keeping cells proliferating has been reconsidered. In addition, an explanation is provided for the aforementioned difference in effect between reduced Ihh and PTHrP expression.
Collapse
Affiliation(s)
- C C van Donkelaar
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600MB, Eindhoven, The Netherlands.
| | | |
Collapse
|
34
|
Hartenstein B, Dittrich BT, Stickens D, Heyer B, Vu TH, Teurich S, Schorpp-Kistner M, Werb Z, Angel P. Epidermal development and wound healing in matrix metalloproteinase 13-deficient mice. J Invest Dermatol 2006; 126:486-96. [PMID: 16374453 PMCID: PMC2767339 DOI: 10.1038/sj.jid.5700084] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Degradation of the extracellular matrix, which is an indispensable step in tissue remodelling processes such as embryonic development and wound healing of the skin, has been attributed to collagenolytic activity of members of the matrix metalloproteinase family (MMPs). Here, we employed mmp13 knockout mice to elucidate the function of MMP13 in embryonic skin development, skin homeostasis, and cutaneous wound healing. Overall epidermal architecture and dermal composition of non-injured skin were indistinguishable from wild-type mice. Despite robust expression of MMP13 in the early phase of wound healing, wild-type and mmp13 knockout animals did not differ in their efficiency of re-epithelialization, inflammatory response, granulation tissue formation, angiogenesis, and restoration of basement membrane. Yet, among other MMPs also expressed during wound healing, MMP8 was found to be enhanced in wounds of MMP13-deficient mice. In summary, skin homeostasis and also tissue remodelling processes like embryonic skin development and cutaneous wound healing are independent of MMP13 either owing to MMP13 dispensability or owing to functional substitution by other collagenolytic proteinases such as MMP8.
Collapse
Affiliation(s)
- Bettina Hartenstein
- Division of Signal Transduction and Growth Control (A100), Deutsches Krebsforschungszentrum Heidelberg (DKFZ), Heidelberg, Germany
| | - Bernd Thilo Dittrich
- Division of Signal Transduction and Growth Control (A100), Deutsches Krebsforschungszentrum Heidelberg (DKFZ), Heidelberg, Germany
| | - Dominique Stickens
- Department of Anatomy, University of California, San Francisco, California, USA
| | - Babette Heyer
- Department of Anatomy, University of California, San Francisco, California, USA
| | - Thiennu H. Vu
- Department of Medicine and Lung Biology Center, University of California, San Francisco, California, USA
| | - Sibylle Teurich
- Division of Signal Transduction and Growth Control (A100), Deutsches Krebsforschungszentrum Heidelberg (DKFZ), Heidelberg, Germany
| | - Marina Schorpp-Kistner
- Division of Signal Transduction and Growth Control (A100), Deutsches Krebsforschungszentrum Heidelberg (DKFZ), Heidelberg, Germany
| | - Zena Werb
- Department of Anatomy, University of California, San Francisco, California, USA
| | - Peter Angel
- Division of Signal Transduction and Growth Control (A100), Deutsches Krebsforschungszentrum Heidelberg (DKFZ), Heidelberg, Germany
| |
Collapse
|
35
|
Mott JD, Werb Z. Regulation of matrix biology by matrix metalloproteinases. Curr Opin Cell Biol 2005; 16:558-64. [PMID: 15363807 PMCID: PMC2775446 DOI: 10.1016/j.ceb.2004.07.010] [Citation(s) in RCA: 784] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2004] [Accepted: 07/19/2004] [Indexed: 01/12/2023]
Abstract
Matrix metalloproteinases (MMPs) are endopeptidases that contribute to growth, development and wound healing as well as to pathologies such as arthritis and cancer. Until recently, it has been thought that MMPs participate in these processes simply by degrading extracellular matrix (ECM) molecules. However, it is now clear that MMP activity is much more directed and causes the release of cryptic information from the ECM. By precisely cleaving large insoluble ECM components and ECM-associated molecules, MMPs liberate bioactive fragments and growth factors and change ECM architecture, all of which influence cellular behavior. Thus, MMPs have become a focal point for understanding matrix biology.
Collapse
Affiliation(s)
- Joni D Mott
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA.
| | | |
Collapse
|
36
|
Hyytiäinen M, Penttinen C, Keski-Oja J. Latent TGF-beta binding proteins: extracellular matrix association and roles in TGF-beta activation. Crit Rev Clin Lab Sci 2004; 41:233-64. [PMID: 15307633 DOI: 10.1080/10408360490460933] [Citation(s) in RCA: 243] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Transforming growth factor betas (TGF-betas) are multifunctional and pleiotropic growth factors. Their major effects include inhibition of cell proliferation and enhancement of extracellular matrix production. TGF-betas are secreted from cells as latent complexes, consisting of mature dimeric growth factor, the latency-associated propeptide (LAP), and a distinct gene product, latent TGF-beta binding protein LTBP. The secreted complex is targeted to specific locations in the extracellular matrix by the appropriate LTBP. The latent complex needs subsequently to be activated. Most studies describing biological effects of TGF-beta have been carried out in cell cultures using high concentrations of active, soluble TGF-beta, where appropriate targeting of the growth factor is missing. However, TGF-beta is produced and secreted in vivo as a latent complex in a specific and targeted manner. Various experimental approaches have convincingly shown the importance of the activation of latent TGF-beta, as well as the importance of LTBPs as targeting molecules of the effects of TGF-beta. Essential steps in the activation appear to be cellular recognition of extracellular matrix-associated LTBPs and subsequent recognition of the associated latent TGF-beta. Cell recognition by specific molecules like integrins and proteolytic events involving plasminogen activation evidently play multifaceted roles in the regulation of TGF-beta activation.
Collapse
Affiliation(s)
- Marko Hyytiäinen
- Department of Virology, Haartman Institute and Helsinki University Hospital, University of Helsinki, Finland
| | | | | |
Collapse
|
37
|
|
38
|
Hayami T, Pickarski M, Wesolowski GA, McLane J, Bone A, Destefano J, Rodan GA, Duong LT. The role of subchondral bone remodeling in osteoarthritis: reduction of cartilage degeneration and prevention of osteophyte formation by alendronate in the rat anterior cruciate ligament transection model. ACTA ACUST UNITED AC 2004; 50:1193-206. [PMID: 15077302 DOI: 10.1002/art.20124] [Citation(s) in RCA: 435] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE It has been suggested that subchondral bone remodeling plays a role in the progression of osteoarthritis (OA). To test this hypothesis, we characterized the changes in the rat anterior cruciate ligament transection (ACLT) model of OA and evaluated the effects of alendronate (ALN), a potent inhibitor of bone resorption, on cartilage degradation and on osteophyte formation. METHODS Male Sprague-Dawley rats underwent ACLT or sham operation of the right knee. Animals were then treated with ALN (0.03 and 0.24 microg/kg/week subcutaneously) and necropsied at 2 or 10 weeks postsurgery. OA changes were evaluated. Subchondral bone volume and osteophyte area were measured by histomorphometric analysis. Coimmunostaining for transforming growth factor beta (TGF beta), matrix metalloproteinase 9 (MMP-9), and MMP-13 was performed to investigate the effect of ALN on local activation of TGF beta. RESULTS ALN was chondroprotective at both dosages, as determined by histologic criteria and collagen degradation markers. ALN suppressed subchondral bone resorption, which was markedly increased 2 weeks postsurgery, and prevented the subsequent increase in bone formation 10 weeks postsurgery, in the untreated tibial plateau of ACLT joints. Furthermore, ALN reduced the incidence and area of osteophytes in a dose-dependent manner. ALN also inhibited vascular invasion into the calcified cartilage in rats with OA and blocked osteoclast recruitment to subchondral bone and osteophytes. ALN treatment reduced the local release of active TGF beta, possibly via inhibition of MMP-13 expression in articular cartilage and MMP-9 expression in subchondral bone. CONCLUSION Subchondral bone remodeling plays an important role in the pathogenesis of OA. ALN or other inhibitors of bone resorption could potentially be used as disease-modifying agents in the treatment of OA.
Collapse
Affiliation(s)
- Tadashi Hayami
- Merck Research Laboratories, West Point, Pennsylvania 19486, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Karsdal MA, Andersen TA, Bonewald L, Christiansen C. Matrix metalloproteinases (MMPs) safeguard osteoblasts from apoptosis during transdifferentiation into osteocytes: MT1-MMP maintains osteocyte viability. DNA Cell Biol 2004; 23:155-65. [PMID: 15068585 DOI: 10.1089/104454904322964751] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Osteoblasts undergo apoptosis or differentiate into either osteocytes or bone-lining cells after termination of bone matrix synthesis. In this study, we investigated the role of matrix metalloproteinases (MMPs) in differentiation of osteoblasts, bone formation, transdifferentiation into osteocytes, and osteocyte apoptosis. This was accomplished by using calvarial sections from the MT1-MMP-deficient mouse and by culture of the mouse osteoblast cell line MC3T3-E1 and primary mouse calvarial osteoblasts. We found that a synthetic matrix metalloprotease inhibitor, GM6001, strongly inhibited bone formation in vitro of both primary osteoblasts and MC3T3 cells by approximately 75%. To further investigate at which level of osteoblast differentiation MMP inhibition was attenuating osteoblast function, we found that neither preosteoblast nor mature osteoblast activity was affected. In contrast, cell survival of osteoblasts forced to transdifferentiate into osteocytes in 3D type I collagen gels were inhibited by more than 50% when exposed to 10 microM GM6001 and to Tissue Inhibitor of Metalloproteinase-2 (TIMP-2), a natural MT1-MMP inhibitor. This shows the importance of MMPs in safeguarding osteoblasts from apoptosis when transdifferentiating into osteocytes. By examination of osteoblasts and osteocytes embedded in calvarial bone in the MT1-MMP deficient mice, we found that MT1-MMP deficient mice had 10-fold higher levels of apoptotic osteocytes than wild-type controls. We have previously shown that MT1-MMP activates latent Transforming Growth Factorbeta (TGF-beta). These findings strongly suggest that MT1-MMP-activated TGF-beta maintains osteoblast survival during transdifferentiation into osteocytes, and maintains mature osteocyte viability. Thus, the interrelationship of MMPs and TGF-beta may play an important role in bone formation and maintenance.
Collapse
Affiliation(s)
- M A Karsdal
- Nordic Bioscience A/S, CCBR, Herlev/Ballerup, DK-2730 Herlev, Denmark.
| | | | | | | |
Collapse
|
40
|
Chen YJ, Wurtz T, Wang CJ, Kuo YR, Yang KD, Huang HC, Wang FS. Recruitment of mesenchymal stem cells and expression of TGF-beta 1 and VEGF in the early stage of shock wave-promoted bone regeneration of segmental defect in rats. J Orthop Res 2004; 22:526-34. [PMID: 15099631 DOI: 10.1016/j.orthres.2003.10.005] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2003] [Accepted: 10/03/2003] [Indexed: 02/04/2023]
Abstract
Extracorporeal shock wave (ESW) treatment has recently been established as a method to enhance bone repair. Here, we reported that ESW-promoted healing of segmental defect via stimulation of mesenchymal stem cell recruitment and differentiation into bone forming cells. Rats with a segmental femoral defect were exposed to a single ESW treatment (0.16 mJ/mm(2), 1 Hz, 500 impulses). Cell morphology and histological changes in the defect region were assessed 3, 7, 14, and 28 days post-treatment. Presence of mesenchymal stem cell was assayed by immuno-staining for RP59, a recently discovered marker, and also production of TGF-beta 1 and VEGF was monitored. ESW treatment increased total cell density and the proportion of RP59 positive cells in the defect region. High numbers of round- and cuboidal-shaped cells strongly expressing RP59 were initially found. Later, the predominant cell type was spindle-shaped fibroblastic cells, subsequently, aggregates of osteogenic and chondrogenic cells were observed. Histological observation suggested that bone marrow stem cells were progressively differentiated into osteoblasts and chondrocytes. RP59 staining was initially intense and decreased with the appearance of expression depended on the differentiation states of osteogenic and chondrogenic cells during the regeneration phase. Mature chondrocytes and osteoblasts exhibited only slight RP59 immuno-reactivity. Expression of TGF-beta 1 and VEGF-A mRNA in the defect tissues was also significantly increased (P<0.05) after ESW treatment as determined by RT-PCR. Intensive TGF-beta 1 immuno-reactivity was induced immediately, whereas a lag period was observed for VEGF-A. Chondrocytes and osteoblasts at the junction of ossified cartilage clearly exhibited VEGF-A expression. Our findings suggest that recruitment of meseoblasts at the junction of ossified cartilage clearly exhibited mesenchymal stem cells is a critical step in bone reparation that is enhanced by ESW treatment. TGF-beta 1 and VEGF-A are proposed to play a chemotactic and mitogenic role in recruitment and differentiation of mesenchymal stem cells.
Collapse
Affiliation(s)
- Yeung-Jen Chen
- Department of Orthopedic Surgery, Chang Gung University, Linkou, Taiwan
| | | | | | | | | | | | | |
Collapse
|
41
|
Olney RC, Wang J, Sylvester JE, Mougey EB. Growth factor regulation of human growth plate chondrocyte proliferation in vitro. Biochem Biophys Res Commun 2004; 317:1171-82. [PMID: 15094393 DOI: 10.1016/j.bbrc.2004.03.170] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Indexed: 11/25/2022]
Abstract
Linear growth occurs as the result of growth plate chondrocytes undergoing proliferative and hypertrophic phases. Paracrine feedback loops that regulate the entry of chondrocytes into the hypertrophic phase have been shown and similar pathways likely exist for the proliferative phase. Human long-bone growth plate chondrocytes were cultured in vitro. The proliferative effects of a variety of factors were determined by [3H]thymidine uptake and the gene expression profile of these cells was determined by DNA microarray analysis. Serum, insulin-like growth factor (IGF)-I and -II, transforming growth factor-beta (TGF-beta, fibroblast growth factor (FGF)-1, -2, and -18, and platelet-derived growth factor (PDGF)-BB were potent stimulators of proliferation. FGF-10, testosterone, and bone morphogenetic proteins (BMP)-2, -4, and -6 inhibited proliferation. Microarray analysis showed that the genes for multiple members of the IGF-I, TGF-beta, FGF, and BMP pathways were expressed, suggesting the presence of autocrine/paracrine pathways that regulate the proliferative phase of growth plate-mediated growth.
Collapse
Affiliation(s)
- Robert C Olney
- Cellular and Molecular Medicine Laboratory, The Nemours Children's Clinic, 807 Children's Way, Jacksonville, FL 32207, USA.
| | | | | | | |
Collapse
|
42
|
Nurminskaya M, Magee C, Faverman L, Linsenmayer TF. Chondrocyte-derived transglutaminase promotes maturation of preosteoblasts in periosteal bone. Dev Biol 2003; 263:139-52. [PMID: 14568552 DOI: 10.1016/s0012-1606(03)00445-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
During endochondral development, elongation of the bone collar occurs coordinately with growth of the underlying cartilaginous growth plate. Transglutaminases (TGases) are upregulated in hypertrophic chondrocytes, and correlative evidence suggests a relationship between these enzymes and mineralization. To examine whether TGases are involved in regulating mineralization/osteogenesis during bone development, we devised a coculture system in which one cellular component (characterized as preosteoblastic) is derived from the nonmineralized region of the bone, and the other cellular component is hypertrophic chondrocytes. In these cocultures, mineralization is extensive, with the preosteoblasts producing the mineralized matrix, and the chondrocytes regulating this process. Secreted regulators are involved, as conditioned medium from chondrocytes induces mineralization in preosteoblasts, but not vice versa. One factor is TGase. In the cocultures, inhibition of TGase reduces mineralization, and addition of the enzyme enhances it. Exogenous TGase also induces markers of osteoblastic differentiation (i.e., bone sialoprotein and osteocalcin) in the preosteoblasts, suggesting their differentiation into osteoblasts. Two possible signaling pathways may be affected by TGase and result in increased mineralization (i.e., TGF-beta and protein kinase A pathways). Addition of exogenous TGF-beta2 to the cocultures increases mineralization; though, when mineralization is induced by TGase, there is no detectible elevation of TGF-beta, suggesting that these two factors stimulate osteogenesis by different pathways. However, an interrelationship seems to exist between TGase and PKA-dependent signaling. When mineralization of the cocultures is stimulated through the addition of TGase, a concomitant reduction (50%) in PKA activity occurs. Consistent with this observation, addition of an activator of PKA (cyclic AMP) to the cultures inhibits matrix mineralization, while known inhibitors of PKA (H-89 and a peptide inhibitor) cause an increase in mineralization. Thus, at least one mechanism of TGase stimulation probably involves inhibition of the PKA-mediated signaling.
Collapse
Affiliation(s)
- Maria Nurminskaya
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA 02111, USA.
| | | | | | | |
Collapse
|
43
|
Abstract
Manipulation of the mouse genome through mis-expressing, knocking out, and introducing mutations into genes of interest has provided important insights into the genetic pathways responsible for human skeletal development. These pathways contribute to the sequential phases of skeletal morphogenesis that include patterning, condensation, and overt organogenesis of the membranous and endochondral embryonic skeletons and to subsequent linear growth. Disturbances in these pathways account for many developmental syndromes and disorders of the human skeleton. Recurrent themes include establishment of interlocking regulatory circuits involving growth factors, receptors, signalling pathways, and transcription factors that control cellular programmes such as migration, adhesion, proliferation, differentiation, and apoptosis, and use of common molecules for different purposes. Technical advances suggest that genetic engineering in mice will continue to be highly instructive in the field of skeletal biology.
Collapse
Affiliation(s)
- William A Horton
- Shriners Hospital for Children, Oregon Health and Science University, 3101 Sam Jackson Park Road, Portland, OR 97239-3009, USA.
| |
Collapse
|
44
|
Karsdal MA, Larsen L, Engsig MT, Lou H, Ferreras M, Lochter A, Delaissé JM, Foged NT. Matrix metalloproteinase-dependent activation of latent transforming growth factor-beta controls the conversion of osteoblasts into osteocytes by blocking osteoblast apoptosis. J Biol Chem 2002; 277:44061-7. [PMID: 12226090 DOI: 10.1074/jbc.m207205200] [Citation(s) in RCA: 188] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Upon termination of bone matrix synthesis, osteoblasts either undergo apoptosis or differentiate into osteocytes or bone lining cells. In this study, we investigated the role of matrix metalloproteinases (MMPs) and growth factors in the differentiation of osteoblasts into osteocytes and in osteoblast apoptosis. The mouse osteoblast cell line MC3T3-E1 and primary mouse calvarial osteoblasts were either grown on two-dimensional (2-D) collagen-coated surfaces, where they morphologically resemble flattened, cuboidal bone lining cells, or embedded in three-dimensional (3-D) collagen gels, where they resemble dendritic osteocytes constituting a network of cells. When MC3T3-E1 osteoblasts were grown in a 3-D matrix in the presence of an MMP inhibitor (GM6001), the cell number was dose-dependently reduced by approximately 50%, whereas no effect was observed on a 2-D substratum. In contrast, the murine mature osteocyte cell line, MLO-Y4, was unaffected by GM6001 under all culture conditions. According to TUNEL assay, the osteoblast apoptosis was increased 2.5-fold by 10 microm GM6001. To investigate the mechanism by which MMPs mediate the survival of osteoblasts, we examined the effect of GM6001 on MC3T3-E1 osteoblasts in the presence of extracellular matrix components and growth factors, including tenascin, fibronectin, laminin, collagenase-cleaved collagen, gelatin, parathyroid hormone, basic fibroblast growth factor, vascular epidermal growth factor, insulin-like growth factor, interleukin-1, and latent and active transforming growth factor-beta (TGF-beta). Only active TGF-beta, but not latent TGF-beta or other agents tested, restored cell number and apoptosis to control levels. Furthermore, we found that the membrane type MMP, MT1-MMP, which is produced by osteoblasts, could activate latent TGF-beta and that antibodies neutralizing endogenous TGF-beta led to a similar decrease in cell number as GM6001. Whereas inhibitors of other protease families did not induce osteoblast apoptosis, an inhibitor of the p44/42 mitogen-activated protein kinase showed the same but non-synergetic effect as GM6001. These findings suggest that MMP-activated TGF-beta maintains osteoblast survival during trans-differentiation into osteocytes by a p44/42-dependent pathway.
Collapse
Affiliation(s)
- Morten A Karsdal
- Nordic Bioscience A/S, Center for Clinical and Basic Research, Herlev/Ballerup, Herlev DK-2730, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Brandau O, Aszódi A, Hunziker EB, Neame PJ, Vestweber D, Fässler R. Chondromodulin I is dispensable during enchondral ossification and eye development. Mol Cell Biol 2002; 22:6627-35. [PMID: 12192060 PMCID: PMC135637 DOI: 10.1128/mcb.22.18.6627-6635.2002] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chondromodulin I (chm-I), a type II transmembrane protein, is highly expressed in the avascular zones of cartilage but is downregulated in the hypertrophic region, which is invaded by blood vessels during enchondral ossification. In vitro and in vivo assays with the purified protein have shown chondrocyte-modulating and angiogenesis-inhibiting functions. To investigate chm-I function in vivo, we generated transgenic mice lacking chm-I mRNA and protein. Null mice are viable and fertile and show no morphological changes. No abnormalities in vascular invasion and cartilage development were detectable. No evidence was found for a compensating function of tendin, a recently published homologue highly expressed in tendons and also, at low levels, in cartilage. Furthermore, no differences in the expression of other angiogenic or antiangiogenic factors such as transforming growth factor beta1 (TGF-beta1), TGF-beta2, TGF-beta3, fibroblast growth factor 2, and vascular endothelial growth factor were found. The surprising lack of phenotype in the chm-I-deficient mice suggests either a different function for chm-I in vivo than has been proposed or compensatory changes in uninvestigated angiogenic or angiogenesis-inhibiting factors. Further analysis using double-knockout technology will be necessary to analyze the function of chm-I in the complex process of enchondral ossification.
Collapse
Affiliation(s)
- Oliver Brandau
- Department of Experimental Pathology, Lund University, Sweden.
| | | | | | | | | | | |
Collapse
|