1
|
Sakurai T, Ishii M, Miyata H, Ikeda N, Suehiro F, Komabashiri N, Oura Y, Nishimura M. Effect of CD10-positive cells on osteogenic differentiation of human maxillary/mandibular bone marrow-derived mesenchymal stem cells. Arch Oral Biol 2025; 170:106135. [PMID: 39591929 DOI: 10.1016/j.archoralbio.2024.106135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
OBJECTIVE This study was aimed at investigating the effect of CD10-positive cells within the maxillary/mandibular bone marrow-derived mesenchymal stem cells (MBMSCs) on osteogenic differentiation of MBMSCs. DESIGN CD10 expression in iliac bone marrow-derived MSCs (IBMSCs), MBMSCs, and gingival fibroblasts was measured using flow cytometry. The osteogenic potential of 19 MBMSC lines was evaluated, and based on it, they were classified into osteogenic-High and osteogenic-Low groups. The percentage of CD10-positive cells in each group was compared. Effect of coculturing gingival fibroblasts and CD10-positive cells on the osteogenic potential of MBMSCs was also assessed. Expression of tissue inhibitor of metalloprotease-1 (TIMP-1) in osteogenic-High and osteogenic-Low MBMSCs was measured using quantitative real-time polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay. The molecular mechanisms underlying the regulation of osteogenic differentiation in MBMSCs were investigated. RESULTS CD10 was not expressed in IBMSCs, but was highly expressed in fibroblasts. In MBMSCs, the CD10-positivity rate varied considerably between cells. MBMSCs with a high-CD10 positivity rate showed low osteogenic potential. Coculture with fibroblasts or CD10-positive cells reduced the osteogenic potential of MBMSCs. TIMP-1 was highly expressed in CD10-positive cells, and osteogenic-Low MBMSCs showed significantly higher TIMP-1 expression compared with osteogenic-High MBMSCs. β-catenin signaling was suppressed in osteogenic-Low MBMSCs. CONCLUSION This study revealed that TIMP-1 secreted from CD10-positive cells may be involved in the suppression of the osteogenic potential of MBMSCs by contamination with CD10-positive cells. This finding provides important insights for developing bone regeneration therapies using MBMSCs.
Collapse
Affiliation(s)
- Tomoaki Sakurai
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate school of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Masakazu Ishii
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate school of Medical and Dental Science, Kagoshima 890-8544, Japan.
| | - Haruka Miyata
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate school of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Nao Ikeda
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate school of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Fumio Suehiro
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate school of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Naohiro Komabashiri
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate school of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Yurika Oura
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate school of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Masahiro Nishimura
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate school of Medical and Dental Science, Kagoshima 890-8544, Japan
| |
Collapse
|
2
|
Lambert J, Oc S, Worssam MD, Häußler D, Solomon CU, Figg NL, Baxter R, Imaz M, Taylor JCK, Foote K, Finigan A, Mahbubani KT, Webb TR, Ye S, Bennett MR, Krüger A, Spivakov M, Jørgensen HF. Network-based prioritization and validation of regulators of vascular smooth muscle cell proliferation in disease. NATURE CARDIOVASCULAR RESEARCH 2024; 3:714-733. [PMID: 38898928 PMCID: PMC11182749 DOI: 10.1038/s44161-024-00474-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 04/18/2024] [Indexed: 06/21/2024]
Abstract
Aberrant vascular smooth muscle cell (VSMC) homeostasis and proliferation characterize vascular diseases causing heart attack and stroke. Here we elucidate molecular determinants governing VSMC proliferation by reconstructing gene regulatory networks from single-cell transcriptomics and epigenetic profiling. We detect widespread activation of enhancers at disease-relevant loci in proliferation-predisposed VSMCs. We compared gene regulatory network rewiring between injury-responsive and nonresponsive VSMCs, which suggested shared transcription factors but differing target loci between VSMC states. Through in silico perturbation analysis, we identified and prioritized previously unrecognized regulators of proliferation, including RUNX1 and TIMP1. Moreover, we showed that the pioneer transcription factor RUNX1 increased VSMC responsiveness and that TIMP1 feeds back to promote VSMC proliferation through CD74-mediated STAT3 signaling. Both RUNX1 and the TIMP1-CD74 axis were expressed in human VSMCs, showing low levels in normal arteries and increased expression in disease, suggesting clinical relevance and potential as vascular disease targets.
Collapse
MESH Headings
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Humans
- Cell Proliferation/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Gene Regulatory Networks
- Tissue Inhibitor of Metalloproteinase-1/metabolism
- Tissue Inhibitor of Metalloproteinase-1/genetics
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/genetics
- Signal Transduction/genetics
- Cells, Cultured
- Single-Cell Analysis
- Epigenesis, Genetic
- Transcriptome
- Animals
- Core Binding Factor Alpha 2 Subunit
Collapse
Affiliation(s)
- Jordi Lambert
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Sebnem Oc
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Functional Gene Control Group, MRC Laboratory of Medical Sciences, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK
| | - Matthew D. Worssam
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Present Address: Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Daniel Häußler
- TUM School of Medicine and Health, Institute of Experimental Oncology and Therapy Research, Technical University of Munich, Munich, Germany
| | - Charles U. Solomon
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
| | - Nichola L. Figg
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Ruby Baxter
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Maria Imaz
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - James C. K. Taylor
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Kirsty Foote
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Alison Finigan
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Krishnaa T. Mahbubani
- Collaborative Biorepository for Translational Medicine, Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Tom R. Webb
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
| | - Shu Ye
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
- Shantou University Medical College, Shantou, China
- Cardiovascular and Metabolic Disease Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - Martin R. Bennett
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Achim Krüger
- TUM School of Medicine and Health, Institute of Experimental Oncology and Therapy Research, Technical University of Munich, Munich, Germany
| | - Mikhail Spivakov
- Functional Gene Control Group, MRC Laboratory of Medical Sciences, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK
| | - Helle F. Jørgensen
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
3
|
Ebert S, Zang L, Ismail N, Otabil M, Fröhlich A, Egea V, Ács S, Hoeberg M, Berres ML, Weber C, Moreira JMA, Ries C, Bernhagen J, El Bounkari O. Tissue Inhibitor of Metalloproteinases-1 Interacts with CD74 to Promote AKT Signaling, Monocyte Recruitment Responses, and Vascular Smooth Muscle Cell Proliferation. Cells 2023; 12:1899. [PMID: 37508563 PMCID: PMC10378328 DOI: 10.3390/cells12141899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Tissue inhibitor of metalloproteinases-1 (TIMP-1), an important regulator of matrix metalloproteinases (MMPs), has recently been shown to interact with CD74, a receptor for macrophage migration inhibitory factor (MIF). However, the biological effects mediated by TIMP-1 through CD74 remain largely unexplored. Using sequence alignment and in silico protein-protein docking analysis, we demonstrated that TIMP-1 shares residues with both MIF and MIF-2, crucial for CD74 binding, but not for CXCR4. Subcellular colocalization, immunoprecipitation, and internalization experiments supported these findings, demonstrating that TIMP-1 interacts with surface-expressed CD74, resulting in its internalization in a dose-dependent manner, as well as with a soluble CD74 ectodomain fragment (sCD74). This prompted us to study the effects of the TIMP-1-CD74 axis on monocytes and vascular smooth muscle cells (VSCMs) to assess its impact on vascular inflammation. A phospho-kinase array revealed the activation of serine/threonine kinases by TIMP-1 in THP-1 pre-monocytes, in particular AKT. Similarly, TIMP-1 dose-dependently triggered the phosphorylation of AKT and ERK1/2 in primary human monocytes. Importantly, Transwell migration, 3D-based Chemotaxis, and flow adhesion assays demonstrated that TIMP-1 engagement of CD74 strongly promotes the recruitment response of primary human monocytes, while live cell imaging studies revealed a profound activating effect on VSMC proliferation. Finally, re-analysis of scRNA-seq data highlighted the expression patterns of TIMP-1 and CD74 in human atherosclerotic lesions, thus, together with our experimental data, indicating a role for the TIMP-1-CD74 axis in vascular inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Simon Ebert
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Lan Zang
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Noor Ismail
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Michael Otabil
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Adrian Fröhlich
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Virginia Egea
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Susann Ács
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Mikkel Hoeberg
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Marie-Luise Berres
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Munich Heart Alliance, 80802 Munich, Germany
| | - José M A Moreira
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christian Ries
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Munich Heart Alliance, 80802 Munich, Germany
| | - Omar El Bounkari
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| |
Collapse
|
4
|
Cut loose TIMP-1: an emerging cytokine in inflammation. Trends Cell Biol 2022; 33:413-426. [PMID: 36163148 DOI: 10.1016/j.tcb.2022.08.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022]
Abstract
Appreciation of the entire biological impact of an individual protein can be hampered by its original naming based on one function only. Tissue inhibitor of metalloproteinases-1 (TIMP-1), mostly known for its eponymous function to inhibit metalloproteinases, exhibits only a fraction of its cellular effects via this feature. Recently, TIMP-1 emerged as a potent cytokine acting via various cell-surface receptors, explaining a so-far under-appreciated role of TIMP-1-mediated signaling on immune cells. This, at least partly, resolved why elevated blood levels of TIMP-1 correlate with progression of numerous inflammatory diseases. Here, we emphasize the necessity of unbiased name-independent recognition of structure-function relationships to properly appreciate the biological potential of TIMP-1 and other cytokines in complex physiological processes such as inflammation.
Collapse
|
5
|
Omran F, Kyrou I, Osman F, Lim VG, Randeva HS, Chatha K. Cardiovascular Biomarkers: Lessons of the Past and Prospects for the Future. Int J Mol Sci 2022; 23:5680. [PMID: 35628490 PMCID: PMC9143441 DOI: 10.3390/ijms23105680] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are a major healthcare burden on the population worldwide. Early detection of this disease is important in prevention and treatment to minimise morbidity and mortality. Biomarkers are a critical tool to either diagnose, screen, or provide prognostic information for pathological conditions. This review discusses the historical cardiac biomarkers used to detect these conditions, discussing their application and their limitations. Identification of new biomarkers have since replaced these and are now in use in routine clinical practice, but still do not detect all disease. Future cardiac biomarkers are showing promise in early studies, but further studies are required to show their value in improving detection of CVD above the current biomarkers. Additionally, the analytical platforms that would allow them to be adopted in healthcare are yet to be established. There is also the need to identify whether these biomarkers can be used for diagnostic, prognostic, or screening purposes, which will impact their implementation in routine clinical practice.
Collapse
Affiliation(s)
- Farah Omran
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; (F.O.); (I.K.); (F.O.); (V.G.L.); (H.S.R.)
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Clinical Sciences Research Laboratories, University Hospitals Coventry and Warwickshire, Coventry CV2 2DX, UK
| | - Ioannis Kyrou
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; (F.O.); (I.K.); (F.O.); (V.G.L.); (H.S.R.)
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Centre of Applied Biological & Exercise Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
| | - Faizel Osman
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; (F.O.); (I.K.); (F.O.); (V.G.L.); (H.S.R.)
- Department of Cardiology, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
| | - Ven Gee Lim
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; (F.O.); (I.K.); (F.O.); (V.G.L.); (H.S.R.)
- Department of Cardiology, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
| | - Harpal Singh Randeva
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; (F.O.); (I.K.); (F.O.); (V.G.L.); (H.S.R.)
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Clinical Sciences Research Laboratories, University Hospitals Coventry and Warwickshire, Coventry CV2 2DX, UK
| | - Kamaljit Chatha
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; (F.O.); (I.K.); (F.O.); (V.G.L.); (H.S.R.)
- Biochemistry and Immunology Department, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
| |
Collapse
|
6
|
Ónody A, Veres-Székely A, Pap D, Rokonay R, Szebeni B, Sziksz E, Oswald F, Veres G, Cseh Á, Szabó AJ, Vannay Á. Interleukin-24 regulates mucosal remodeling in inflammatory bowel diseases. J Transl Med 2021; 19:237. [PMID: 34078403 PMCID: PMC8173892 DOI: 10.1186/s12967-021-02890-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/17/2021] [Indexed: 12/30/2022] Open
Abstract
Background Recently, increased interleukin (IL)-24 expression has been demonstrated in the colon biopsies of adult patients with inflammatory bowel disease (IBD). However, the role of IL-24 in the pathomechanism of IBD is still largely unknown. Methods Presence of IL-24 was determined in the samples of children with IBD and in the colon of dextran sodium sulfate (DSS) treated mice. Effect of inflammatory factors on IL24 expression was determined in peripheral blood (PBMCs) and lamina propria mononuclear cells (LPMCs). Also, the impact of IL-24 was investigated on HT-29 epithelial cells and CCD-18Co colon fibroblasts. Expression of tissue remodeling related genes was investigated in the colon of wild type (WT) mice locally treated with IL-24 and in the colon of DSS treated WT and Il20rb knock out (KO) mice. Results Increased amount of IL-24 was demonstrated in the serum and colon samples of children with IBD and DSS treated mice compared to that of controls. IL-1β, LPS or H2O2 treatment increased the expression of IL24 in PBMCs and LPMCs. IL-24 treatment resulted in increased amount of TGF-β and PDGF-B in HT-29 cells and enhanced the expression of extracellular matrix (ECM)-related genes and the motility of CCD-18Co cells. Similarly, local IL-24 treatment increased the colonic Tgfb1 and Pdgfb expression of WT mice. Moreover, expression of pro-fibrotic Tgfb1 and Pdgfb were lower in the colon of DSS treated Il20rb KO compared to that of WT mice. The disease activity index of colitis was less severe in DSS treated Il20rb KO compared to WT mice. Conclusion Our study suggest that IL-24 may play a significant role in the mucosal remodeling of patients with IBD by promoting pro-fibrotic processes. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02890-7.
Collapse
Affiliation(s)
- Anna Ónody
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Apor Veres-Székely
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary.,ELKH-SE Pediatrics and Nephrology Research Group, 53-54 Bókay J. Street, Budapest, H-1083, Hungary
| | - Domonkos Pap
- ELKH-SE Pediatrics and Nephrology Research Group, 53-54 Bókay J. Street, Budapest, H-1083, Hungary
| | - Réka Rokonay
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Beáta Szebeni
- ELKH-SE Pediatrics and Nephrology Research Group, 53-54 Bókay J. Street, Budapest, H-1083, Hungary
| | - Erna Sziksz
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Franz Oswald
- Department of Internal Medicine I, University Medical Center, Ulm, Germany
| | - Gábor Veres
- Pediatric Institute-Clinic, University of Debrecen, Debrecen, Hungary
| | - Áron Cseh
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Attila J Szabó
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary.,ELKH-SE Pediatrics and Nephrology Research Group, 53-54 Bókay J. Street, Budapest, H-1083, Hungary
| | - Ádám Vannay
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary. .,ELKH-SE Pediatrics and Nephrology Research Group, 53-54 Bókay J. Street, Budapest, H-1083, Hungary.
| |
Collapse
|
7
|
Interleukin-11 is important for vascular smooth muscle phenotypic switching and aortic inflammation, fibrosis and remodeling in mouse models. Sci Rep 2020; 10:17853. [PMID: 33082445 PMCID: PMC7576123 DOI: 10.1038/s41598-020-74944-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
Transforming growth factor beta-1 (TGFβ1) is a major driver of vascular smooth muscle cell (VSMC) phenotypic switching, an important pathobiology in arterial disease.
We performed RNA-sequencing of TGFβ1-stimulated human aortic or arterial VSMCs which revealed large and consistent upregulation of Interleukin 11 (IL11). IL11 has an unknown function in VSMCs, which highly express the IL11 receptor alpha, suggestive of an autocrine loop. In vitro, IL11 activated ERK signaling, but inhibited STAT3 activity, and caused VSMC phenotypic switching to a similar extent as TGFβ1 or angiotensin II (ANGII) stimulation. Genetic or therapeutic inhibition of IL11 signaling reduced TGFβ1- or ANGII-induced VSMC phenotypic switching, placing IL11 activity downstream of these factors. Aortas of mice with Myh11-driven IL11 expression were remodeled and had reduced contractile but increased matrix and inflammatory genes expression. In two models of arterial pressure loading, IL11 was upregulated in the aorta and neutralizing IL11 antibodies reduced remodeling along with matrix and pro-inflammatory gene expression. These data show that IL11 plays an important role in VSMC phenotype switching, vascular inflammation and aortic pathobiology.
Collapse
|
8
|
Nordgaard C, Doll S, Matos ALDSA, Høeberg M, Kazi JU, Friis S, Stenvang J, Rönnstrand L, Mann M, Manuel Afonso Moreira J. Metallopeptidase inhibitor 1 (TIMP-1) promotes receptor tyrosine kinase c-Kit signaling in colorectal cancer. Mol Oncol 2019; 13:2646-2662. [PMID: 31545548 PMCID: PMC6887592 DOI: 10.1002/1878-0261.12575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/13/2019] [Accepted: 09/20/2019] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent cancer worldwide causing an estimated 700 000 deaths annually. Different types of treatment are available for patients with advanced metastatic colorectal cancer, including targeted biological agents, such as cetuximab, a monoclonal antibody that targets EGFR. We have previously reported a study indicating multiple levels of interaction between metallopeptidase inhibitor 1 (TIMP‐1) and the epidermal growth factor (EGF) signaling axis, which could explain how TIMP‐1 levels can affect the antitumor effects of EGFR inhibitors. We also reported an association between TIMP‐1‐mediated cell invasive behavior and KRAS status. To gain insight into the molecular mechanisms underlying the effects of TIMP‐1 in CRC, we examined by transcriptomics, proteomics, and kinase activity profiling a matched pair of isogenic human CRC isogenic DLD‐1 CRC cell clones, bearing either an hemizygous KRAS wild‐type allele or KRAS G13D mutant allele, exposed, or not, to TIMP‐1. Omics analysis of the two cell lines identified the receptor tyrosine kinase c‐Kit, a proto‐oncogene that can modulate cell proliferation and invasion in CRC, as a target for TIMP‐1. We found that exposure of DLD‐1 CRC cells to exogenously added TIMP‐1 promoted phosphorylation of c‐Kit, indicative of a stimulatory effect of TIMP‐1 on the c‐Kit signaling axis. In addition, TIMP‐1 inhibited c‐Kit shedding in CRC cells grown in the presence of exogenous TIMP‐1. Given the regulatory roles that c‐Kit plays in cell proliferation and migration, and the realization that c‐Kit is an important oncogene in CRC, it is likely that some of the biological effects of TIMP‐1 overexpression in CRC may be exerted through its effect on c‐Kit signaling.
Collapse
Affiliation(s)
- Cathrine Nordgaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Sophia Doll
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Denmark
| | | | - Mikkel Høeberg
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Julhash Uddin Kazi
- Division of Translational Cancer Research and Lund Stem Cell Center, Lund University, Sweden
| | - Stine Friis
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jan Stenvang
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lars Rönnstrand
- Division of Translational Cancer Research and Lund Stem Cell Center, Lund University, Sweden.,Division of Oncology, Skåne University Hospital, Lund, Sweden
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - José Manuel Afonso Moreira
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
9
|
Zhong C, Wang G, Xu T, Zhu Z, Guo D, Zheng X, Wang A, Bu X, Peng H, Chen J, Xu T, Peng Y, Li Q, Ju Z, Geng D, He J, Zhang Y. Tissue inhibitor metalloproteinase-1 and clinical outcomes after acute ischemic stroke. Neurology 2019; 93:e1675-e1685. [PMID: 31551260 DOI: 10.1212/wnl.0000000000008389] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/23/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To prospectively investigate the relationships between serum tissue inhibitor metalloproteinase-1 (TIMP-1) and clinical outcomes in patients with acute ischemic stroke. METHODS We derived data from the China Antihypertensive Trial in Acute Ischemic Stroke. Baseline serum TIMP-1 concentrations were measured in 3,342 participants. The primary outcome was the combination of death and major disability (modified Rankin Scale score ≥3) at 3 months after ischemic stroke, and secondary outcomes included major disability, death, and vascular events. RESULTS A total of 843 participants (25.2%) experienced major disability or died within 3 months. After adjustment for age, sex, admission NIH Stroke Scale score, and other important covariates, odds ratios or hazard ratios (95% confidence intervals) of 1-SD (0.17 ng/mL) higher log-TIMP-1 were 1.17 (1.06-1.29) for the primary outcome, 1.13 (1.02-1.25) for major disability, 1.49 (1.19-1.87) for death, and 1.34 (1.11-1.62) for the composite outcome of death and vascular events. The addition of serum TIMP-1 to conventional risk factors model significantly improved risk prediction of the primary outcome (net reclassification index 9.0%, p = 0.02; integrated discrimination improvement 0.2%, p = 0.03). Participants with both higher TIMP-1 and matrix metalloproteinase-9 levels simultaneously had the highest risk of all study outcomes. CONCLUSIONS Higher TIMP-1 levels were associated with increased risk of mortality and major disability after acute ischemic stroke. Our findings provided evidence supporting the important prognostic role of extracellular matrix biomarkers after acute ischemic stroke.
Collapse
Affiliation(s)
- Chongke Zhong
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Guangli Wang
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Tan Xu
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Zhengbao Zhu
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Daoxia Guo
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Xiaowei Zheng
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Aili Wang
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Xiaoqing Bu
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Hao Peng
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Jing Chen
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Tian Xu
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Yanbo Peng
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Qunwei Li
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Zhong Ju
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Deqin Geng
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China
| | - Jiang He
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China.
| | - Yonghong Zhang
- From the Department of Epidemiology (C.Z., G.W., Tan Xu, Z.Z., D. Guo, X.Z., A.W., X.B., H.P., Tian Xu, Y.Z.), School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology (C.Z., J.C., J.H.), Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Neurology (Y.P.), Affiliated Hospital of North China University of Science and Technology, Tangshan; Department of Epidemiology (Q.L.), School of Public Health, Taishan Medical College, Taian; Department of Neurology (Z.J.), Kerqin District First People's Hospital of Tongliao City; Department of Neurology (D. Geng), Affiliated Hospital of Xuzhou Medical College, China; Department of Medicine (J.C., J.H.), Tulane University School of Medicine, New Orleans, LA; and Department of Neurology (Tian Xu), Affiliated Hospital of Nantong University, China.
| |
Collapse
|
10
|
Sendon-Lago J, Seoane S, Martinez-Ordoñez A, Eiro N, Saa J, Vizoso FJ, Gonzalez F, Perez-Fernandez R, Bermudez MA. Corneal regeneration by conditioned medium of human uterine cervical stem cells is mediated by TIMP-1 and TIMP-2. Exp Eye Res 2019; 180:110-121. [PMID: 30557571 DOI: 10.1016/j.exer.2018.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/22/2018] [Accepted: 12/11/2018] [Indexed: 01/15/2023]
Abstract
The aim of the present study was to evaluate the effect and the mechanism of action of the conditioned medium from human uterine cervical stem cells (CM-hUCESC) on corneal wound healing in a rabbit dry eye model. To do this, dry eye and corneal epithelial injuries were induced in rabbits by topical administration of atropine sulfate and NaOH. Hematoxylin-Eosin (H&E) and Ki-67 immunostaining were carried out to evaluate corneal damage and cell proliferation, and real-time PCR was used to evaluate proinflammatory cytokines in the cornea. In addition, in order to investigate possible factors involved in corneal regeneration, primary cultures of rat corneal epithelial cells (rCECs) were used to evaluate cell migration, proliferation, and apoptosis before and after immunoprecipitation of specific factors from the CM-hUCESC. Results showed that CM-hUCESC treatment significantly improved epithelial regeneration in rabbits with dry eye induced by atropine and reduced corneal pro-inflammatory TNF-α, MCP-1, MIP-1α and IL-6 cytokines. In addition, metalloproteinase inhibitors TIMP-1 and TIMP-2, which are present at high levels in CM-hUCESC, mediated corneal regenerative effects by both inducing corneal epithelial cell proliferation and inhibiting apoptosis. In summary, CM-hUCESC induces faster corneal regeneration in a rabbit model of dry eye induced by atropine than conventional treatments, being TIMP-1 and TIMP-2 mediators in this process. The results indicate that an alternative CM-based treatment for some corneal conditions is achievable, although future studies would be necessary to investigate other factors involved in the multiple observed effects of CM-hUCESC.
Collapse
Affiliation(s)
- Juan Sendon-Lago
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Samuel Seoane
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Anxo Martinez-Ordoñez
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Noemi Eiro
- Research Unit, Hospital Fundacion de Jove, Gijón, Spain.
| | - Jorge Saa
- Research Unit, Hospital Fundacion de Jove, Gijón, Spain; Service of Ophthalmology, Fundación Hospital de Jove, Gijón, Spain.
| | | | - Francisco Gonzalez
- Department of Surgery and CIMUS, University of Santiago de Compostela, Spain; Service of Ophthalmology and IDIS, Complejo Hospitalario Universitario de Santiago de Compostela, Spain.
| | - Roman Perez-Fernandez
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Maria A Bermudez
- Department of Biology, Faculty of Science, University of A Coruña, Spain.
| |
Collapse
|
11
|
Corbitt H, Gutierrez J, Silberbach M, Maslen CL. The genetic basis of Turner syndrome aortopathy. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2019; 181:117-125. [PMID: 30770620 DOI: 10.1002/ajmg.c.31686] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 01/15/2023]
Abstract
Our goal is to identify the genetic underpinnings of bicuspid aortic valve and aortopathy in Turner syndrome. We performed whole exome sequencing on 188 Turner syndrome study subjects from the GenTAC registry. A gene-based burden test, SKAT-O, was used to evaluate the data using bicuspid aortic valve (BAV) and aortic dimension z-scores as covariates. This revealed that TIMP3 was associated with BAV and increased aortic dimensions at exome-wide significance. It had been previously shown that genes on chromosome Xp contribute to aortopathy when hemizygous. Our analysis of Xp genes revealed that hemizygosity for TIMP1, a functionally redundant paralogue of TIMP3, increased the odds of having BAV aortopathy compared to individuals with more than one TIMP1 copy. The combinatorial effect of a single copy of TIMP1 and TIMP3 risk alleles synergistically increased the risk for BAV aortopathy to nearly 13-fold. TIMP1 and TIMP3 are tissue inhibitors of matrix metalloproteinases (TIMPs) which are involved in development of the aortic valve and protection from thoracic aneurysms. We propose that the combination of TIMP1 haploinsufficiency and deleterious variants in TIMP3 significantly increases the risk of BAV aortopathy in Turner syndrome, and suggest that TIMP1 hemizygosity may play a role in euploid male aortic disease.
Collapse
Affiliation(s)
- Holly Corbitt
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
| | - Jacob Gutierrez
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Michael Silberbach
- Department of Pediatric Cardiology, Oregon Health & Science University, Portland, Oregon
| | - Cheryl L Maslen
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
12
|
Abstract
Jawed vertebrates (Gnathostomes) have 4 tissue inhibitors of metalloproteinases (TIMPs), multifunctional proteins that all inhibit members of the large matrix metalloproteinase (MMP) family but differ in their other roles, including the regulation of pro-MMP activation, cell growth, apoptosis and angiogenesis, and the structure of extracellular matrices (ECMs). Molecular phylogeny analyses indicate that vertebrate TIMP genes arose from an invertebrate ancestor through 3 successive duplications, possibly including 2 whole genome duplications, during early vertebrate phylogeny. TIMPs from invertebrates also inhibit metalloproteinases, bind to pro-MMPs, and contribute to ECM structures but are not orthologs of any particular vertebrate TIMP. The most ancient vertebrate superclass, the Agnatha (jawless fish), seems to provide a snapshot of a stage in TIMP evolution preceding the third gene duplication. This review examines the structures of TIMPs from different vertebrate orders using information relating to the structural basis of their various functions. Provisional conclusions are that during their evolutionary divergence, various TIMPs lost inhibitory activity toward some metalloproteinases, specialized in effects on different pro-MMPs, and developed new interactions with discrete targets (including integrins and receptors), while recapitulating a role in ECM structure. The analysis is limited by the sparse information available regarding the functional properties of nonmammalian TIMPs.-Brew, K. Reflections on the evolution of the vertebrate tissue inhibitors of metalloproteinases.
Collapse
Affiliation(s)
- Keith Brew
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| |
Collapse
|
13
|
Tarpgaard LS, Ørum-Madsen MS, Christensen IJ, Nordgaard C, Noer J, Guren TK, Glimelius B, Sorbye H, Ikdahl T, Kure EH, Tveit KM, Nielsen HJ, Pfeiffer P, Brünner N, Moreira JMA. TIMP-1 is under regulation of the EGF signaling axis and promotes an aggressive phenotype in KRAS-mutated colorectal cancer cells: a potential novel approach to the treatment of metastatic colorectal cancer. Oncotarget 2018; 7:59441-59457. [PMID: 27509063 PMCID: PMC5312323 DOI: 10.18632/oncotarget.11118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/17/2016] [Indexed: 01/05/2023] Open
Abstract
It is now widely accepted that therapeutic antibodies targeting epidermal growth factor receptor (EGFR) can have efficacy in KRAS wild-type advanced colorectal cancer (CRC) patients. What remains to be ascertained is whether a subgroup of KRAS-mutated CRC patients might not also derive benefit from EGFR inhibitors. Metalloproteinase inhibitor 1 (TIMP-1) is a pleiotropic factor predictive of survival outcome of CRC patients. Levels of TIMP-1 were measured in pre-treatment plasma samples (n = 426) of metastatic CRC patients randomized to Nordic FLOX (5-fluorouracil and oxaliplatin) +/− cetuximab (NORDIC VII study). Multivariate analysis demonstrated a significant interaction between plasma TIMP-1 protein levels, KRAS status and treatment with patients bearing KRAS mutated tumors and high TIMP-1 plasma level (> 3rd quartile) showing a significantly longer overall survival if treated with cetuximab (HR, 0.48; 95% CI, 0.25 to 0.93). To gain mechanistic insights into this association we analyzed a set of five different CRC cell lines. We show here that EGFR signaling induces TIMP-1 expression in CRC cells, and that TIMP-1 promotes a more aggressive behavior, specifically in KRAS mutated cells. The two sets of data, clinical and in vitro, are complementary and support each other, lending strength to our contention that TIMP- 1 plasma levels can identify a subset of patients with KRAS-mutated metastatic CRC that will have benefit from EGFR-inhibition therapy.
Collapse
Affiliation(s)
- Line S Tarpgaard
- Department of Oncology, Odense University Hospital, Odense, Denmark and University of Southern Denmark, Odense, Denmark
| | - Maj Sofie Ørum-Madsen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ib J Christensen
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark and Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Cathrine Nordgaard
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Julie Noer
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tormod K Guren
- Department of Oncology and K. G. Jebsen Centre for Colorectal Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bengt Glimelius
- Departments of Radiology, Oncology and Radiation Science, Uppsala University, Uppsala and Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Tone Ikdahl
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Elin H Kure
- Department of Genetics, Oslo University Hospital, Oslo, Norway
| | - Kjell M Tveit
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Hans J Nielsen
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark and University of Southern Denmark, Odense, Denmark
| | - Nils Brünner
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - José M A Moreira
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
McCarthy CP, van Kimmenade RR, Gaggin HK, Simon ML, Ibrahim NE, Gandhi P, Kelly N, Motiwala SR, Belcher AM, Harisiades J, Magaret CA, Rhyne RF, Januzzi JL. Usefulness of Multiple Biomarkers for Predicting Incident Major Adverse Cardiac Events in Patients Who Underwent Diagnostic Coronary Angiography (from the Catheter Sampled Blood Archive in Cardiovascular Diseases [CASABLANCA] Study). Am J Cardiol 2017; 120:25-32. [PMID: 28487034 DOI: 10.1016/j.amjcard.2017.03.265] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 01/12/2023]
Abstract
We sought to develop a multiple biomarker approach for prediction of incident major adverse cardiac events (MACE; composite of cardiovascular death, myocardial infarction, and stroke) in patients referred for coronary angiography. In a 649-participant training cohort, predictors of MACE within 1 year were identified using least-angle regression; over 50 clinical variables and 109 biomarkers were analyzed. Predictive models were generated using least absolute shrinkage and selection operator with logistic regression. A score derived from the final model was developed and evaluated with a 278-patient validation set during a median of 3.6 years follow-up. The scoring system consisted of N-terminal pro B-type natriuretic peptide (NT-proBNP), kidney injury molecule-1, osteopontin, and tissue inhibitor of metalloproteinase-1; no clinical variables were retained in the predictive model. In the validation cohort, each biomarker improved model discrimination or calibration for MACE; the final model had an area under the curve (AUC) of 0.79 (p <0.001), higher than AUC for clinical variables alone (0.75). In net reclassification improvement analyses, addition of other markers to NT-proBNP resulted in significant improvement (net reclassification improvement 0.45; p = 0.008). At the optimal score cutoff, we found 64% sensitivity, 76% specificity, 28% positive predictive value, and 93% negative predictive value for 1-year MACE. Time-to-first MACE was shorter in those with an elevated score (p <0.001); such risk extended to at least to 4 years. In conclusion, in a cohort of patients who underwent coronary angiography, we describe a novel multiple biomarker score for incident MACE within 1 year (NCT00842868).
Collapse
|
15
|
Voutilainen SH, Kosola SK, Tervahartiala TI, Sorsa TA, Jalanko HJ, Pakarinen MP. Liver and serum expression of matrix metalloproteinases in asymptomatic pediatric liver transplant recipients. Transpl Int 2016; 30:124-133. [DOI: 10.1111/tri.12879] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 07/29/2016] [Accepted: 10/19/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Silja H. Voutilainen
- Pediatric Surgery and Pediatric Transplantation Surgery; Pediatric Liver and Gut Research Group; Children's Hospital; Helsinki University Central Hospital and University of Helsinki; Helsinki Finland
| | - Silja K. Kosola
- Pediatric Surgery and Pediatric Transplantation Surgery; Pediatric Liver and Gut Research Group; Children's Hospital; Helsinki University Central Hospital and University of Helsinki; Helsinki Finland
| | - Taina I. Tervahartiala
- Department of Oral and Maxillofacial Diseases; Institute of Dentistry; University of Helsinki and Helsinki University Central Hospital; Helsinki Finland
| | - Timo A. Sorsa
- Department of Oral and Maxillofacial Diseases; Institute of Dentistry; University of Helsinki and Helsinki University Central Hospital; Helsinki Finland
| | - Hannu J. Jalanko
- Pediatric Nephrology and Transplantation; Children's Hospital; Helsinki University Central Hospital and University of Helsinki; Helsinki Finland
| | - Mikko P. Pakarinen
- Pediatric Surgery and Pediatric Transplantation Surgery; Pediatric Liver and Gut Research Group; Children's Hospital; Helsinki University Central Hospital and University of Helsinki; Helsinki Finland
| |
Collapse
|
16
|
Alfakry H, Malle E, Koyani CN, Pussinen PJ, Sorsa T. Neutrophil proteolytic activation cascades: a possible mechanistic link between chronic periodontitis and coronary heart disease. Innate Immun 2016; 22:85-99. [PMID: 26608308 DOI: 10.1177/1753425915617521] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/13/2015] [Indexed: 12/30/2022] Open
Abstract
Cardiovascular diseases are chronic inflammatory diseases that affect a large segment of society. Coronary heart disease (CHD), the most common cardiovascular disease, progresses over several years and affects millions of people worldwide. Chronic infections may contribute to the systemic inflammation and enhance the risk for CHD. Periodontitis is one of the most common chronic infections that affects up to 50% of the adult population. Under inflammatory conditions the activation of endogenous degradation pathways mediated by immune responses leads to the release of destructive cellular molecules from both resident and immigrant cells. Matrix metalloproteinases (MMPs) and their regulators can activate each other and play an important role in immune response via degrading extracellular matrix components and modulating cytokines and chemokines. The action of MMPs is required for immigrant cell recruitment at the site of inflammation. Stimulated neutrophils represent the major pathogen-fighting immune cells that upregulate expression of several proteinases and oxidative enzymes, which can degrade extracellular matrix components (e.g. MMP-8, MMP-9 and neutrophil elastase). The activity of MMPs is regulated by endogenous inhibitors and/or candidate MMPs (e.g. MMP-7). The balance between MMPs and their inhibitors is thought to mirror the proteolytic burden. Thus, neutrophil-derived biomarkers, including myeloperoxidase, may activate proteolytic destructive cascades that are involved in subsequent immune-pathological events associated with both periodontitis and CHD. Here, we review the existing studies on the contribution of MMPs and their regulators to the infection-related pathology. Also, we discuss the possible proteolytic involvement and role of neutrophil-derived enzymes as an etiological link between chronic periodontitis and CHD.
Collapse
Affiliation(s)
- Hatem Alfakry
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ernst Malle
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Chintan N Koyani
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Pirkko J Pussinen
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Timo Sorsa
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
17
|
The tissue inhibitor of metalloproteinases 1 increases the clonogenic efficiency of human hematopoietic progenitor cells through CD63/PI3K/Akt signaling. Exp Hematol 2015. [DOI: 10.1016/j.exphem.2015.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
18
|
Lindsey ML, Yabluchanskiy A, Ma Y. Tissue Inhibitor of Metalloproteinase-1: Actions beyond Matrix Metalloproteinase Inhibition. Cardiology 2015; 132:147-50. [PMID: 26279068 DOI: 10.1159/000433419] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 05/19/2015] [Indexed: 01/05/2023]
Affiliation(s)
- Merry L Lindsey
- San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Miss., USA
| | | | | |
Collapse
|
19
|
TIMP-1 overexpression in lung carcinoma enhances tumor kinetics and angiogenesis in brain metastasis. J Neuropathol Exp Neurol 2015; 74:293-304. [PMID: 25756591 DOI: 10.1097/nen.0000000000000175] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Tissue inhibitors of matrix metalloproteinase (TIMP) orchestrate many biologic activities, including inhibition of matrix metalloproteinase activity, activation of pro-matrix metalloproteinases, and regulation of cell proliferation, angiogenesis, and apoptosis induction. Tissue inhibitors of matrix metalloproteinase can play a protective role during tumor invasion and metastasis, but elevated TIMP messenger RNA levels have also been associated with aggressive cancers and poor clinical outcome. We examined the potential roles of TIMP-1 in H2009 lung adenocarcinoma cells and in cells transfected with a human TIMP-1-overexpressing vector (HB-6 and HB-1). Tumors resulting from the implantation of parental cell lines and transfected HB-1 cells into the brains of nude mice had a typical carcinoma profile, but human TIMP-1-overexpressing tumors showed enhanced tumor kinetics and focally more infiltrative features; vessel density assessed with anti-CD31 immunohistochemistry was also greater within HB-1 tumor implants. Similar effects on HB-6 and HB-1 cells versus parental cell lines and empty vector clones were observed in endothelial cell assays. Anchorage-independent growth and invasion through Matrigel were also increased in TIMP-1-overexpressing cells. Together, these results indicate tumor-promoting functions of TIMP-1 through alterations in angiogenesis, increased tumorigenicity, and invasive behavior. Although matrix metalloproteinase inhibition has been the traditionally identified function of TIMP-1, matrix metalloproteinase-independent interactions may contribute to the growth of metastatic carcinomas in the brain.
Collapse
|
20
|
Ries C. Cytokine functions of TIMP-1. Cell Mol Life Sci 2014; 71:659-72. [PMID: 23982756 PMCID: PMC11113289 DOI: 10.1007/s00018-013-1457-3] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/09/2013] [Accepted: 08/12/2013] [Indexed: 12/13/2022]
Abstract
The tissue inhibitors of metalloproteinases (TIMPs) are well recognized for their role in extracellular matrix remodeling by controlling the activity of matrix metalloproteinases (MMPs). Independent of MMP inhibition, TIMPs act as signaling molecules with cytokine-like activities thereby influencing various biological processes including cell growth, apoptosis, differentiation, angiogenesis, and oncogenesis. Recent studies on TIMP-1's cytokine functions have identified complex regulatory networks involving a specific surface receptor and subsequent signaling pathways including miRNA-mediated posttranscriptional regulation of gene expression that ultimately control the fate and behavior of the cells. The present review summarizes the current knowledge on TIMP-1 as a cytokine modulator of cell functions, outlines recent progress in defining molecular pathways that transmit TIMP-1 signals from the cell periphery into the nucleus, and discusses TIMP-1's role as a cytokine in the pathophysiology of cancer and other human diseases.
Collapse
Affiliation(s)
- Christian Ries
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University of Munich, Pettenkoferstrasse 9b, 80336, Munich, Germany,
| |
Collapse
|
21
|
Abrial M, Da Silva CC, Pillot B, Augeul L, Ivanes F, Teixeira G, Cartier R, Angoulvant D, Ovize M, Ferrera R. Cardiac fibroblasts protect cardiomyocytes against lethal ischemia-reperfusion injury. J Mol Cell Cardiol 2014; 68:56-65. [PMID: 24440456 DOI: 10.1016/j.yjmcc.2014.01.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 01/07/2014] [Accepted: 01/09/2014] [Indexed: 02/07/2023]
Abstract
Roles of cardiac fibroblasts (CFs) in the regulation of myocardial structure and function have been emphasized in the last decade. Their implications in pathophysiological aspects of chronic heart diseases such as myocardial remodeling and fibrosis are now well established; however their contribution to the acute phase of ischemia-reperfusion injury still remains elusive. We hypothesized that CF may contribute to cardiomyocyte (CM) protection against ischemia-reperfusion injuries. Experiments performed on isolated neonatal rat CF and CM demonstrated that the presence of CF in co-cultures increases CM viability (58 ± 2% versus 30 ± 2% in control) against hypoxia-reoxygenation injury, in a paracrine manner. It was confirmed by a similar effect of hypoxic CF secretome alone on CM viability (51 ± 9% versus 31 ± 4% in untreated cells). These findings were corroborated by in vivo experiments in a mice model of myocardial infarction in which a 25% infarct size reduction was observed in CF secretome treated mice compared to control. Tissue inhibitor of metalloproteinases-1 (TIMPs-1) alone, abundantly detected in CF secretome, was able to decrease CM cell death (35%) and experiments with pharmacological inhibitors of PI3K/Akt and ERK1/2 pathways provided more evidence that this paracrine protection is partly mediated by these signaling pathways. In vivo experiments strengthened that TIMP-1 alone was able to decrease infarct size (37%) and were validated by depletion experiments demonstrating that CF secretome cardioprotection was abolished by TIMP-1 depletion. Our data demonstrated for the first time that CFs participate in cardioprotection during the acute phase of ischemia-reperfusion via a paracrine pathway involving TIMP-1.
Collapse
Affiliation(s)
- Maryline Abrial
- INSERM U1060, CarMeN Laboratory, Université Lyon 1, F-69373 Lyon, France.
| | | | - Bruno Pillot
- INSERM U1060, CarMeN Laboratory, Université Lyon 1, F-69373 Lyon, France
| | - Lionel Augeul
- INSERM U1060, CarMeN Laboratory, Université Lyon 1, F-69373 Lyon, France
| | - Fabrice Ivanes
- INSERM U1060, CarMeN Laboratory, Université Lyon 1, F-69373 Lyon, France; Université François Rabelais EA 4245, CHRU Tours, Hôpital Trousseau, Service de Cardiologie, F-37044 Tours, France
| | - Geoffrey Teixeira
- INSERM U1060, CarMeN Laboratory, Université Lyon 1, F-69373 Lyon, France
| | - Régine Cartier
- Hospices Civils de Lyon, Hôpital Louis Pradel, Service d'Explorations Fonctionnelles Cardiovasculaires & CIC de Lyon, F-69394 Lyon, France
| | - Denis Angoulvant
- Université François Rabelais EA 4245, CHRU Tours, Hôpital Trousseau, Service de Cardiologie, F-37044 Tours, France
| | - Michel Ovize
- INSERM U1060, CarMeN Laboratory, Université Lyon 1, F-69373 Lyon, France; Hospices Civils de Lyon, Hôpital Louis Pradel, Service d'Explorations Fonctionnelles Cardiovasculaires & CIC de Lyon, F-69394 Lyon, France
| | - René Ferrera
- INSERM U1060, CarMeN Laboratory, Université Lyon 1, F-69373 Lyon, France
| |
Collapse
|
22
|
The balance of serum matrix metalloproteinase-8 and its tissue inhibitor in acute coronary syndrome and its recurrence. Int J Cardiol 2013; 167:362-8. [DOI: 10.1016/j.ijcard.2011.12.095] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 12/19/2011] [Accepted: 12/24/2011] [Indexed: 11/24/2022]
|
23
|
Tsai HP, Chen SC, Chien HT, Jan YY, Chao TC, Chen MF, Hsieh LL. Relationships between serum HER2 ECD, TIMP-1 and clinical outcomes in Taiwanese breast cancer. World J Surg Oncol 2012; 10:42. [PMID: 22339939 PMCID: PMC3312842 DOI: 10.1186/1477-7819-10-42] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 02/17/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Serum levels of the extracellular domain of HER2/neu (HER2 ECD) have been demonstrated to be associated with clinical outcomes. A disintegrin and metalloproteinase-10, a sheddase of HER2/neu, can drive cancer progression and its activity is inhibited by tissue inhibitor of metalloproteinase-1 (TIMP-1). However, elevated TIMP-1 expression has been associated with a poor prognosis of breast cancer. Therefore, this study was performed to explore the relationships between serum HER2 ECD, TIMP-1 and clinical outcomes. METHODS One hundred and eighty-five female breast cancer patients, who received curative mastectomy without neo-adjuvant chemotherapy at Chang-Gung Memorial Hospital, were recruited with informed consent for this study. Pre-operative serum levels of HER2 ECD and TIMP-1 were measured using an enzyme-linked immunosorbent assay. RESULTS Twenty-three cases (12.4%) were classified HER2 ECD positive. HER2 ECD positivity was significantly associated with age, lymph node involvement, histological grade, estrogen receptor status, progesterone receptor status, tissue HER2/neu overexpression, and disease-free survival (DFS). In an age, stage, ER and HER2/neu status matched subgroup (N = 41), the serum level of TIMP-1 was significantly associated with HER2 ECD positivity and DFS. CONCLUSIONS A high serum TIMP-1 was significantly associated with HER2 ECD positivity and a poorer DFS among Taiwanese primary breast cancer patients with HER2 overexpression.
Collapse
Affiliation(s)
- Hsiu-Pei Tsai
- Graduate Institute of Clinical Medical Sciences,Chang Gung University, Tao-Yuan, Taiwan
| | | | | | | | | | | | | |
Collapse
|
24
|
Lu Y, Liu S, Zhang S, Cai G, Jiang H, Su H, Li X, Hong Q, Zhang X, Chen X. Tissue inhibitor of metalloproteinase-1 promotes NIH3T3 fibroblast proliferation by activating p-Akt and cell cycle progression. Mol Cells 2011; 31:225-30. [PMID: 21350939 PMCID: PMC3932703 DOI: 10.1007/s10059-011-0023-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 11/09/2010] [Accepted: 11/24/2010] [Indexed: 11/29/2022] Open
Abstract
Tissue inhibitor of metalloproteinase-1 (TIMP-1) plays various roles in cell growth in different cell types. However, few studies have focused on TIMP-1's effect on fibroblast cells. In this study, we investigated the effects of TIMP-1 overexpression on NIH3T3 fibroblast proliferation and potential transduction signaling pathways involved. Overexpression of TIMP-1, by transfection of the pLenti6/V5-DESTTIMP-1 plasmid, significantly promoted NIH3T3 proliferation as determined by the BrdU array. Neither 5 nor 15 nM GM6001 (matrix metalloproteinase system inhibitor) affected NIH3T3 proliferation, but 45 nM GM6001 inhibited proliferation. TIMP-1 overexpression activated the p-Akt pathway, but not the p-ERK or p-p38 pathway. In TIMP-1-transfected cells, cyclinD1 was upregulated and p21CIP1 and p27(KIP1) were downregulated, which promoted cell entry into the S and G2/M phases. The PI3-K inhibitor LY294002 abolished the TIMP-1-induced effects. Overexpression of intracellular TIMP-1 stimulated NIH3T3 fibroblast proliferation in a matrix metalloproteinase (MMP)-independent manner by activating the p-Akt pathway and related cell cycle progression.
Collapse
Affiliation(s)
- Yang Lu
- Department of Nephrology, Kidney Center and Key Lab of the People's Liberation Army (PLA), General Hospital of PLA, Beijing, China
| | - Shuxin Liu
- Dalian Municipal Central Hospital, Dalian, China
| | - Shujia Zhang
- Department of Nephrology, Kidney Center and Key Lab of the People's Liberation Army (PLA), General Hospital of PLA, Beijing, China
- Department of Nephrology, 1st Hospital of Haerbin, China
| | - Guangyan Cai
- Department of Nephrology, Kidney Center and Key Lab of the People's Liberation Army (PLA), General Hospital of PLA, Beijing, China
| | - Hongwei Jiang
- Department of Endocrinology and Metabolism, 1st Affiliated Hospital, Henan University of Science and Technology, Luoyang, China
| | - Huabin Su
- Department of Nephrology, Kidney Center and Key Lab of the People's Liberation Army (PLA), General Hospital of PLA, Beijing, China
| | - Xiaofan Li
- Department of Nephrology, Kidney Center and Key Lab of the People's Liberation Army (PLA), General Hospital of PLA, Beijing, China
| | - Quan Hong
- Department of Nephrology, Kidney Center and Key Lab of the People's Liberation Army (PLA), General Hospital of PLA, Beijing, China
| | - Xueguang Zhang
- Department of Nephrology, Kidney Center and Key Lab of the People's Liberation Army (PLA), General Hospital of PLA, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, Kidney Center and Key Lab of the People's Liberation Army (PLA), General Hospital of PLA, Beijing, China
| |
Collapse
|
25
|
Pradhan-Palikhe P, Vikatmaa P, Lajunen T, Palikhe A, Lepäntalo M, Tervahartiala T, Salo T, Saikku P, Leinonen M, Pussinen PJ, Sorsa T. Elevated MMP-8 and Decreased Myeloperoxidase Concentrations Associate Significantly with the Risk for Peripheral Atherosclerosis Disease and Abdominal Aortic Aneurysm1. Scand J Immunol 2010; 72:150-7. [DOI: 10.1111/j.1365-3083.2010.02418.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
26
|
Gaubatz JW, Ballantyne CM, Wasserman BA, He M, Chambless LE, Boerwinkle E, Hoogeveen RC. Association of circulating matrix metalloproteinases with carotid artery characteristics: the Atherosclerosis Risk in Communities Carotid MRI Study. Arterioscler Thromb Vasc Biol 2010; 30:1034-42. [PMID: 20167662 DOI: 10.1161/atvbaha.109.195370] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To examine the relationship of plasma levels of matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinase 1 (TIMP-1) with carotid artery characteristics measured by MRI in a cross-sectional investigation among Atherosclerosis Risk in Communities Carotid MRI Study participants. METHODS AND RESULTS A stratified random sample was recruited based on intima-media thickness from a previous ultrasonographic examination. A high-resolution gadolinium-enhanced MRI examination of the carotid artery was performed from 2004 to 2005 on 1901 Atherosclerosis Risk in Communities cohort participants. Multiple carotid wall characteristics, including wall thickness, lumen area, calcium area, lipid core, and fibrous cap measures, were evaluated for associations with plasma MMPs 1, 2, 3, 7, 8, and 9 and TIMP-1. Plasma MMPs 1, 3, and 7 were significantly higher among participants in the high intima-media thickness group compared with those in the low intima-media thickness group. The normalized wall index was independently associated with MMPs 3 and 7 and TIMP-1. MMP-7 was positively associated with carotid calcification. The mean fibrous cap thickness was significantly higher in individuals with elevated TIMP-1 levels. In addition, TIMP-1 was positively associated with measures of lipid core. CONCLUSION Circulating levels of specific MMPs and TIMP-1 were associated with carotid wall remodeling and structural changes related to plaque burden in elderly participants.
Collapse
Affiliation(s)
- John W Gaubatz
- Department of Medicine, Baylor College of Medicine, 6565 Fannin St, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Serum metalloproteinases MMP-2, MMP-9 and metalloproteinase tissue inhibitors TIMP-1 and TIMP-2 in patients on hemodialysis. Int Urol Nephrol 2009; 43:491-8. [PMID: 19953348 DOI: 10.1007/s11255-009-9683-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2008] [Accepted: 11/14/2009] [Indexed: 01/03/2023]
Abstract
BACKGROUND We assessed the effect of hemodialysis (HD) and chronic kidney disease (CKD) on the serum levels of metalloproteinase-2 (MMP-2), MMP-9 and metalloproteinase tissue inhibitors (TIMP-1) and TIMP-2. METHODS 18 patients on regular HD treatment with low-flux, cuprophane membrane, 15 non-dialyzed patients with CKD and 15 healthy controls were sampled. The serum MMP and TIMP concentrations were determined by ELISA assays. RESULTS MMP-9, TIMP-1, and TIMP-2 serum levels were significantly decreased in HD patients to 32.7 ± 20.1 ng/ml, 178.8 ± 73.0 ng/ml, and 103.4 ± 55.3 ng/ml compared with 482.3 ± 139.5, 367.6 ± 75.5 ng/ml, and 299.7 ± 63.2 ng/ml in patients with CKD and 594.6 ± 154.7 ng/ml, 354.5 ± 81.2 ng/ml, and 272.4 ± 91.8 ng/ml in healthy controls, respectively, (P < 0.001 vs. HD patients). MMP-2 was lower in patients with CKD: 405.6 ± 106.1 ng/ml compared with 516.9 ± 81.7 ng/ml in controls (P = 0.02). The MMP-2/TIMP-2 ratio was increased in HD patients compared with both patients with CKD and controls. In the course of an HD session, MMP-2 and TIMP-1 serum levels were significantly decreased from pre-HD 570.0 ± 256.5 and 178.8 ± 66.9 ng/ml to post-HD 492.6 ± 212.5 and 144.6 ± 44.2 ng/ml (P = 0.004 and 0.013, respectively). However, the MMP-9/TIMP-1 ratio increased from pre-HD 0.15 (2.19) (median, range) to 0.23 (0.33) after a HD session (P = 0.03). CRP was positively correlated with MMP-9 and MMP-9/TIMP-1 ratio in HD patients and patients with CKD (r = 0.67; P = 0.03). CONCLUSIONS The MMP-9/TIMP-1 ratio increased during HD sessions, although their absolute levels were lowered. This change may represent a chronic state of enhanced fibrosis in patients undergoing HD.
Collapse
|
28
|
Zhong H, Matsui O, Xu K, Ogi T, Sanada JI, Okamoto Y, Tabata Y, Takuwa Y. Gene transduction into aortic wall using plasmid-loaded cationized gelatin hydrogel-coated polyester stent graft. J Vasc Surg 2009; 50:1433-43. [DOI: 10.1016/j.jvs.2009.07.071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 07/13/2009] [Accepted: 07/13/2009] [Indexed: 01/28/2023]
|
29
|
Rysz J, Aronow WS, Stolarek RS, Hannam S, Mikhailidis DP, Banach M. Nephroprotective and clinical potential of statins in dialyzed patients. Expert Opin Ther Targets 2009; 13:541-50. [PMID: 19368496 DOI: 10.1517/14728220902882130] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
30
|
Møller Sørensen N, Vejgaard Sørensen I, Ørnbjerg Würtz S, Schrohl AS, Dowell B, Davis G, Jarle Christensen I, Nielsen HJ, Brünner N. Biology and potential clinical implications of tissue inhibitor of metalloproteinases-1 in colorectal cancer treatment. Scand J Gastroenterol 2008; 43:774-86. [PMID: 18584515 DOI: 10.1080/00365520701878163] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related death in the industrialized world. About half of "curatively" resected patients develop recurrent disease within the next 3-5 years despite the lack of clinical, histological and biochemical evidence of remaining overt disease after resection of the primary tumour. Availability of validated biological markers for early detection, selection for adjuvant therapy, prediction of treatment efficacy and monitoring of treatment efficacy would most probably increase survival. Tissue inhibitor of metalloproteinases-1 (TIMP-1) may be such a marker. TIMP-1 inhibits the proteolytic activity of metalloproteinases, which are centrally involved in tumour invasion and metastases. However, in clinical investigations high tumour tissue or plasma levels of TIMP-1 have shown a strong and independent association with a shorter survival time in CRC patients, suggesting that TIMP-1 could have a tumour-promoting function. Furthermore, measurement of plasma TIMP-1 has been shown to be useful for disease detection, with a high sensitivity and high specificity for early-stage colon cancer. This review describes some basic information on the current knowledge of the biology of TIMP-1 as well as the potential use of TIMP-1 as a biological marker in the management of CRC patients.
Collapse
Affiliation(s)
- Nanna Møller Sørensen
- Section of Biomedicine, Department of Veterinary Pathobiology, Faculty of Life Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Mateos-Cáceres PJ, López-Farré AJ, Morata PC, Ramos-Mozo P, Macaya C, Serrano FJ, Moñux G. Pravastatin increases the expression of the tissue inhibitor of matrix metalloproteinase-1 and the oncogeneBaxin human aortic abdominal aneurysms. Can J Physiol Pharmacol 2008; 86:431-7. [DOI: 10.1139/y08-060] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effect of pravastatin on matrix metalloproteinase-9 (MMP-9) and the level of tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2 was studied in explants of human abdominal aortic aneurysm (AAA) obtained from 13 patients. The effect of pravastatin on the apoptotic status of human AAA explants was also examined. Total MMP-9 content did not differ in human AAA explants incubated in vitro in the presence or absence of pravastatin (10−6mol/L) for 48 h. TIMP-1 levels were significantly increased in pravastatin-incubated AAA explants, but TIMP-2 production was not modified by pravastatin. Western blot experiments showed that, whereas Bax expression was increased in pravastatin-incubated AAA explants, the expression of Bcl-2 was not modified. On the other hand, the ratio of the expression of Bax to Bcl-2, an apoptotic index, was not modified by pravastatin. In the human AAA explants, the increase in Bax expression, but not the increase in TIMP-1 expression elicited by pravastatin, was reversed by l-mevalonate, a downstream HMG-CoA reductase metabolite, suggesting that the expression of Bax and TIMP-1 followed HMG-CoA reductase-dependent and -independent pathways, respectively. In conclusion, pravastatin increases both TIMP-1 and Bax expression in human AAA explants without changes in either MMP-9 activity or the apoptotic status.
Collapse
Affiliation(s)
- Petra J. Mateos-Cáceres
- Vascular Surgery Department, Cardiovascular Institute, Hospital Clínico San Carlos, C/Profesor Martín Lagos s/n, Madrid 28040, Spain
- Cardiovascular Research Unit, Cardiovascular Institute, Hospital Clínico San Carlos, Madrid 28040, Spain
| | - Antonio J. López-Farré
- Vascular Surgery Department, Cardiovascular Institute, Hospital Clínico San Carlos, C/Profesor Martín Lagos s/n, Madrid 28040, Spain
- Cardiovascular Research Unit, Cardiovascular Institute, Hospital Clínico San Carlos, Madrid 28040, Spain
| | - Pilar C. Morata
- Vascular Surgery Department, Cardiovascular Institute, Hospital Clínico San Carlos, C/Profesor Martín Lagos s/n, Madrid 28040, Spain
- Cardiovascular Research Unit, Cardiovascular Institute, Hospital Clínico San Carlos, Madrid 28040, Spain
| | - Priscila Ramos-Mozo
- Vascular Surgery Department, Cardiovascular Institute, Hospital Clínico San Carlos, C/Profesor Martín Lagos s/n, Madrid 28040, Spain
- Cardiovascular Research Unit, Cardiovascular Institute, Hospital Clínico San Carlos, Madrid 28040, Spain
| | - Carlos Macaya
- Vascular Surgery Department, Cardiovascular Institute, Hospital Clínico San Carlos, C/Profesor Martín Lagos s/n, Madrid 28040, Spain
- Cardiovascular Research Unit, Cardiovascular Institute, Hospital Clínico San Carlos, Madrid 28040, Spain
| | - Francisco J. Serrano
- Vascular Surgery Department, Cardiovascular Institute, Hospital Clínico San Carlos, C/Profesor Martín Lagos s/n, Madrid 28040, Spain
- Cardiovascular Research Unit, Cardiovascular Institute, Hospital Clínico San Carlos, Madrid 28040, Spain
| | - Guillermo Moñux
- Vascular Surgery Department, Cardiovascular Institute, Hospital Clínico San Carlos, C/Profesor Martín Lagos s/n, Madrid 28040, Spain
- Cardiovascular Research Unit, Cardiovascular Institute, Hospital Clínico San Carlos, Madrid 28040, Spain
| |
Collapse
|
32
|
Cortelezzi A, Colombo G, Pellegrini C, Silvestris I, Moronetti Mazzeo L, Bosari S, Lambertenghi Deliliers G, Fracchiolla NS. Bone marrow glycophorin-positive erythroid cells of myelodysplastic patients responding to high-dose rHuEPO therapy have a different gene expression pattern from those of nonresponders. Am J Hematol 2008; 83:531-9. [PMID: 18383321 DOI: 10.1002/ajh.21178] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The main clinical problems of low-risk patients with myelodysplastic syndromes (MDS), as defined by the International Prognostic Scoring System, are infections and the need for frequent transfusions due to ineffective myelopoiesis and peripheral blood cytopenia. Promising results in treating MDS-related anemia have been obtained using high-dose recombinant human erythropoietin (rhEPO). To evaluate the molecular basis of the response to rhEPO, we used commercially available macro-arrays to investigate gene expression profiles in the glycophorin-expressing (Gly+) bone marrow (BM) erythroid cells of five responders (ERs) and five non-responders (ENRs) to rhEPO treatment. The cells were separated by means of positive selection using an immunomagnetic procedure, after which flow cytometry showed that their purity was more than 97% in all cases. The array data were validated by means of real time RT-PCR. The results showed that the genes responsible for proliferation/differentiation and DNA repair/stability were repressed in the BM Gly+ erythroid cells of the ENRs, but almost normally expressed in the ERs. Furthermore, the expression of genes involved in signal transduction suggested that the activity of the MAPK signaling pathway is inhibited in ERs. The different gene expression profiles of ERs and ENRs may provide a basis for early gene testing as a means of predicting the response to rhEPO of MDS patients with low endogenous EPO levels.
Collapse
Affiliation(s)
- Agostino Cortelezzi
- Hematology-Bone Marrow Transplant Unit, Fondazione Ospedale Maggiore Maggiore Policlinico, Mangiagalli, Regina Elena IRCCS, Milan.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Stetler-Stevenson WG. The tumor microenvironment: regulation by MMP-independent effects of tissue inhibitor of metalloproteinases-2. Cancer Metastasis Rev 2008; 27:57-66. [PMID: 18058195 DOI: 10.1007/s10555-007-9105-8] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Proteolytic remodeling of the extracellular matrix is an important component of disease progression in many chronic disease states and is the initiating event in the formation of the tumor microenvironment in cancer. It is the balance of extracellular matrix degrading enzymes, the matrix metalloproteinases (MMPs) and their endogenous inhibitors that determine the extent of tissue remodeling. Unchecked MMP activity can result in significant tissue damage, facilitate disease progression and is associated with host responses to pathologic injury such as angiogenesis and inflammation. The tissue inhibitors of metalloproteinases (TIMPs) have been shown to regulate MMP activity. However, recent findings demonstrate that the tissue inhibitor of metalloproteinases-2 (TIMP-2) inhibits the mitogenic response of human microvascular endothelial cells to growth factors, such as VEGF-A and FGF-2 in vitro and angiogenesis in vivo. The mechanism of this effect is independent of metalloproteinase inhibition. Our lab is the first to demonstrate a cell-surface signaling receptor for a member of the TIMP family and suggest that TIMP-2 functions to regulate cellular responses to growth factors. These new findings are discussed in terms of a model of TIMP-2 regulation of cellular functions in the tumor microenvironment.
Collapse
|
34
|
Porter JF, Sharma S, Wilson DL, Kappil MA, Hart RP, Denhardt DT. Tissue inhibitor of metalloproteinases-1 stimulates gene expression in MDA-MB-435 human breast cancer cells by means of its ability to inhibit metalloproteinases. Breast Cancer Res Treat 2007; 94:185-93. [PMID: 16142437 DOI: 10.1007/s10549-005-7728-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Tissue inhibitor of metalloproteinases-1 (TIMP-1) is a widely expressed, secreted protein that functions primarily to inhibit members of a large family of metalloproteinases (MPs). Because of the ability of TIMP-1 to inhibit MPs, it functions in many of the same pathophysiological processes as these enzymes, e.g. wound healing, ovulation, angiogenesis, and cancer cell metastasis. TIMP-1 can also stimulate proliferation ([3H]thymidine incorporation) and cellular anabolic processes (Alamar Blue reduction). This stimulation has been shown to be dependent on the MP-inhibitory ability of TIMP-1 in the human breast cancer cell line MDA-MB-435 (Porter et al., Br J Cancer 90: 463, 2004). To shed light on the mechanism by which TIMP-1 stimulates cellular anabolic processes, an oligonucleotide microarray analysis was performed over a time course of TIMP-1 treatment of MDA-MB-435 cells. Fifteen genes whose mRNAs were differentially regulated were identified. Six (Importin-7, MGC10471, FOXC1, subunit p20 of Arp2/3 complex, mitochondrial ribosomal protein L32, and the serine/threonine kinase-4 (MST1)) of these genes were confirmed by quantitative real time PCR. These same mRNAs were shown to be regulated by the synthetic hydroxamate MP-inhibitor GM6001 but not by its inactive derivative GM6001*, suggesting that the differential regulation occurs through the MP-inhibitory ability of TIMP-1. These results suggest a complex action of TIMP-1 on cancer cells mediated by constitutively active cell surface metalloproteinases that release factors regulating cell signaling pathways; they may account for the paradoxical observation that elevated levels of TIMP-1 in tumors can correlate with an adverse prognosis.
Collapse
Affiliation(s)
- Joseph F Porter
- The Graduate Program in Microbiology and Molecular Genetics, Nelson Laboratories, Rutgers University, Piscataway, NJ 08854, USA
| | | | | | | | | | | |
Collapse
|
35
|
Nazari H, Takahashi A, Harada N, Mawatari K, Nakano M, Kishi K, Ebina Y, Nakaya Y. Angiotensin II inhibits insulin-induced actin stress fiber formation and glucose uptake via ERK1/2. THE JOURNAL OF MEDICAL INVESTIGATION 2007; 54:19-27. [PMID: 17380010 DOI: 10.2152/jmi.54.19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
There is crosstalk in intracellular signaling between Angiotensin II (Ang II) and insulin. We hypothesized that the underlying mechanism might be related to changes in cytoskeleton. In the presence of 100 nM of Ang II, insulin-induced glucose uptake was decreased and insulin-induced actin filament organization was inhibited. PKC inhibitors, including GF109203x and p38MAPK inhibitor (SB203580) neither improved insulin-induced actin reorganization nor glucose uptake. In contrast, the Ang II-induced inhibition of glucose uptake and actin filament disorganization was reversed by 10 micromol ERK 1/2 MAPK inhibitor (PD98059). Pretreatment of Ang II increased ERK1/2 phosphorylation and inhibited insulin-induced Akt phosphorylation. The effect of Ang II on ERK1/2 phosphorylation was blocked by Ang II type 1 receptor antagonists, RNH6270 and PD98059 but not by SB203580 or Guanosine-5'-O-(2-ThioDiphosphate), a G-protein inhibitor. We next tested the effect of broad-spectrum matrix metalloproteinase (MMP) inhibitor (GM6001) on Ang II-inhibition of insulin signaling pathway. GM6001 did not improve Ang II-induced actin filament disorganization and did not inhibit ERK1/2 phosphorylation. From these data in L6 myotube, we conclude that Ang II negatively regulates the insulin signal not through MMP signaling pathway but specifically through MMP-independent ERK1/2 activation pathway, providing an alternative molecular mechanism for angiotensin-induced insulin resistance.
Collapse
Affiliation(s)
- Hossein Nazari
- Department of Nutrition and Metabolism, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Bogaczewicz J, Jasielski P, Mosiewicz A, Trojanowski T, Suchozebrska-Jesionek D, Stryjecka-Zimmer M. [The role of matrix metalloproteinases and tissue inhibitors of metalloproteinases in invasion of tumours of neuroepithelial tissue]. Neurol Neurochir Pol 2007; 45:291-338. [PMID: 17103354 DOI: 10.1080/10408360801973244] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tumour invasion requires degradation of extracellular matrix components and migration of cells through degraded structures into surrounding tissues. Matrix metalloproteinases (MMP) constitute a family of zinc and calcium-dependent endopeptidases that play a key role in the breakdown of extracellular matrix, and in processing of cytokines, growth factors, chemokines and cell surface receptors. Their activity is regulated at the levels of transcription, activation and inhibition by tissue inhibitors of metalloproteinases (TIMP). Changes in expression of MMP and TIMP are implicated in tumour invasion, because they may contribute to both migration of tumour cells and angiogenesis. Alterations of MMP expression observed in brain tumours arouse interest in the development and evaluation of synthetic matrix metalloproteinase inhibitors as antitumour agents.
Collapse
Affiliation(s)
- Jarosław Bogaczewicz
- Katedra i Klinika Neurochirurgii i Neurochirurgii Dzieciêcej, Akademia Medyczna im. prof. Feliksa Skubiszewskiego, ul. Jaczewskiego 8, 20-954 Lublin.
| | | | | | | | | | | |
Collapse
|
37
|
Verstappen J, Von den Hoff JW. Tissue inhibitors of metalloproteinases (TIMPs): their biological functions and involvement in oral disease. J Dent Res 2007; 85:1074-84. [PMID: 17122157 DOI: 10.1177/154405910608501202] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Several families of enzymes are responsible for the degradation of extracellular matrix (ECM) proteins during the remodeling of tissues. An important family of such enzymes is that of the matrix metalloproteinases (MMPs). To control MMP-mediated ECM breakdown, tissue inhibitors of metalloproteinases (TIMPs) are able to inhibit MMP activity. A disturbed balance of MMPs and TIMPs is found in various pathologic conditions, such as cancer, rheumatoid arthritis, and periodontitis. The role of MMPs in pathology has been extensively described in the literature. The main focus of this review lies in the biological functions of TIMPs and their occurrence in disease, especially in the head and neck area. Their biological functions and their role in diseases like oral cancers and periodontitis, and in the development of cleft palate, will be discussed. Finally, the diagnostic and therapeutical opportunities of TIMPs will be evaluated.
Collapse
Affiliation(s)
- J Verstappen
- Department of Orthodontics and Oral Biology, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | | |
Collapse
|
38
|
Elmas E, Lang S, Dempfle CE, Kälsch T, Hannak D, Sueselbeck T, Wolpert C, Borggrefe M, Brueckmann M. High plasma levels of tissue inhibitor of metalloproteinase-1 (TIMP-1) and interleukin-8 (IL-8) characterize patients prone to ventricular fibrillation complicating myocardial infarction. ACTA ACUST UNITED AC 2007; 45:1360-5. [PMID: 17727318 DOI: 10.1515/cclm.2007.286] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractClin Chem Lab Med 2007;45:1360–5.
Collapse
Affiliation(s)
- Elif Elmas
- 1st Department of Medicine, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Karakiulakis G, Papakonstantinou E, Aletras AJ, Tamm M, Roth M. Cell type-specific effect of hypoxia and platelet-derived growth factor-BB on extracellular matrix turnover and its consequences for lung remodeling. J Biol Chem 2006; 282:908-15. [PMID: 17099219 DOI: 10.1074/jbc.m602178200] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hypoxia is associated with extracellular matrix remodeling in several inflammatory lung diseases, such as fibrosis, chronic obstructive pulmonary disease, and asthma. In a human cell culture model, we assessed whether extracellular matrix modification by hypoxia and platelet-derived growth factor (PDGF) involves the action of matrix metalloproteinases (MMPs) and thereby affects cell proliferation. Expression of MMP and its activity were assessed by zymography and enzyme-linked immunosorbent assay in human lung fibroblasts and pulmonary vascular smooth muscle cells (VSMCs), and synthesis of soluble collagen type I was assessed by enzyme-linked immunosorbent assay. In both cell types, hypoxia up-regulated the expression of MMP-1, -2, and -9 precursors without subsequent activation. MMP-13 was increased by hypoxia only in fibroblasts. PDGF-BB inhibited the synthesis and secretion of all hypoxia-dependent MMP via Erk1/2 mitogen-activated protein (MAP) kinase activation. Hypoxia and PDGF-BB induced synthesis of soluble collagen type I via Erk1/2 and p38 MAP kinase. Hypoxia-induced cell proliferation was blocked by antibodies to PDGF-BB or by inhibition of Erk1/2 but not by the inhibition of MMP or p38 MAP kinase in fibroblasts. In VSMCs, hypoxia-induced proliferation involved Erk1/2 and p38 MAP kinases and was further increased by fibroblast-conditioned medium or soluble collagen type I via Erk1/2. In conclusion, hypoxia controls tissue remodeling and proliferation in a cell type-specific manner. Furthermore, fibroblasts may affect proliferation of VSMC indirectly by inducing the synthesis of soluble collagen type I.
Collapse
MESH Headings
- Becaplermin
- Cells, Cultured
- Collagen Type I/metabolism
- Enzyme Precursors/metabolism
- Extracellular Matrix/drug effects
- Extracellular Matrix/metabolism
- Feedback, Physiological/drug effects
- Feedback, Physiological/physiology
- Fibroblasts/cytology
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Gelatinases/metabolism
- Humans
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Lung/cytology
- Lung/metabolism
- Matrix Metalloproteinase 13/metabolism
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 9/metabolism
- Metalloendopeptidases/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Platelet-Derived Growth Factor/pharmacology
- Proto-Oncogene Proteins c-sis
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Tissue Inhibitor of Metalloproteinase-1/metabolism
Collapse
Affiliation(s)
- George Karakiulakis
- Department of Pharmacology, School of Medicine, Aristotle University, GR-54124 Thessaloniki, Greece, and Pulmonary Cell Research and Pneumology, University Hospital Basel, CH-4031 Basel, Switzerland
| | | | | | | | | |
Collapse
|
40
|
Yan YP, Sailor KA, Vemuganti R, Dempsey RJ. Insulin-like growth factor-1 is an endogenous mediator of focal ischemia-induced neural progenitor proliferation. Eur J Neurosci 2006; 24:45-54. [PMID: 16882007 DOI: 10.1111/j.1460-9568.2006.04872.x] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The adult mammalian brain contains resident neural progenitors in the subgranular zone of the dentate gyrus (DG) and the subventricular zone (SVZ) of the lateral ventricles. The proliferation of neural progenitors increases after focal cerebral ischemia in both of these regions, but the mechanisms that promote ischemia-induced neural progenitor proliferation are not yet understood. We hypothesize that diffusible factors from the ischemic area play a role in this process as the DG is remote from the area of infarction. In this study, we observed that the peak of neural progenitor proliferation in the ipsilateral DG was between day 2 and day 4 of reperfusion after transient middle cerebral artery occlusion in adult spontaneously hypertensive rats. GeneChip and real-time PCR analysis showed a three- to 102-fold increase in the expression of 15 diffusible, mitogenic factors in the ischemic cortex at 3 days of reperfusion. Of these, insulin-like growth factor-1 (IGF-1) showed increased protein expression in the activated astrocytes in the ischemic penumbra. In addition, the progenitors in both the SVZ and DG showed IGF-1 receptor expression. Inhibiting IGF-1 activity by introcerebroventricular infusion of IGF-1 antibody significantly prevented the ischemia-induced neural progenitor proliferation. These results indicate that IGF-1 formed in the ischemic penumbra might be one of the diffusible factors that mediate post-ischemic neural progenitor proliferation.
Collapse
Affiliation(s)
- Yi-Ping Yan
- Department of Neurological Surgery, University of Wisconsin-Madison, WI 53792, USA
| | | | | | | |
Collapse
|
41
|
Homo-Delarche F, Calderari S, Irminger JC, Gangnerau MN, Coulaud J, Rickenbach K, Dolz M, Halban P, Portha B, Serradas P. Islet inflammation and fibrosis in a spontaneous model of type 2 diabetes, the GK rat. Diabetes 2006; 55:1625-33. [PMID: 16731824 DOI: 10.2337/db05-1526] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The molecular pathways leading to islet fibrosis in diabetes are unknown. Therefore, we studied gene expression in islets of 4-month-old Goto-Kakizaki (GK) and Wistar control rats. Of 71 genes found to be overexpressed in GK islets, 24% belong to extracellular matrix (ECM)/cell adhesion and 34% to inflammatory/immune response families. Based on gene data, we selected several antibodies to study fibrosis development during progression of hyperglycemia by immunohistochemistry. One-month-old GK and Wistar islets appeared to be similar. Two-month-old GK islets were strongly heterogenous in terms of ECM accumulation compared with Wistar islets. GK islet vascularization, labeled by von Willebrand factor, was altered after 1 month of mild hyperglycemia. Numerous macrophages (major histocompatibility complex class II(+) and CD68(+)) and granulocytes were found in/around GK islets. These data demonstrate that marked inflammatory reaction accompanies GK islet fibrosis and suggest that islet alterations in this nonobese model of type 2 diabetes develop in a way reminiscent of microangiopathy.
Collapse
Affiliation(s)
- Françoise Homo-Delarche
- Unité Mixte de Recherche 7059, National Center for Scientific Research, Diderot University, Paris, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Cavusoglu E, Ruwende C, Chopra V, Yanamadala S, Eng C, Clark LT, Pinsky DJ, Marmur JD. Tissue inhibitor of metalloproteinase-1 (TIMP-1) is an independent predictor of all-cause mortality, cardiac mortality, and myocardial infarction. Am Heart J 2006; 151:1101.e1-8. [PMID: 16644343 DOI: 10.1016/j.ahj.2006.02.029] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2005] [Accepted: 02/07/2006] [Indexed: 11/23/2022]
Abstract
BACKGROUND Matrix metalloproteinases and their inhibitors have been implicated in both vascular and ventricular remodeling, and in atherosclerotic plaque rupture. The prognostic value of plasma tissue inhibitor of metalloproteinase-1 (TIMP-1) levels in patients with established or suspected coronary artery disease is unknown. METHODS Tissue inhibitor of metalloproteinase-1 and matrix metalloproteinase-9 (MMP-9) levels, along with a number of other established biomarkers, were measured in 389 male patients undergoing coronary angiography at a Veterans Administration Medical Center. The patients were then followed prospectively for the occurrence of all-cause mortality, cardiac mortality, and myocardial infarction (MI). RESULTS Follow-up data at 24 months were available for 97% of the patients. For the entire cohort of patients, TIMP-1 was the only biomarker to independently predict all-cause mortality and MI. In addition, the ratio of TIMP-1 to matrix metalloproteinase-9 was independently predictive of cardiac mortality at 24 months. The 24-month survival rates for patients in the lower quartile (< 66.5 ng/mL), interquartile (66.5-100 ng/mL), and upper quartile (> 100 ng/mL) of plasma TIMP-1 values were 95.3%, 89.3%, and 72.2%, respectively (P < .001). Furthermore, when patients with chest pain were risk stratified into those with and without an acute coronary syndrome, TIMP-1 remained an independent predictor of all-cause mortality in both subgroups. CONCLUSIONS In a cohort of male patients undergoing coronary angiography, a single baseline determination of plasma TIMP-1 is independently predictive of the subsequent risk of death and MI.
Collapse
Affiliation(s)
- Erdal Cavusoglu
- Department of Medicine, State University of New York Health Science Center, Brooklyn, NY 11203-2098, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Crombez L, Marques B, Lenormand JL, Mouz N, Polack B, Trocme C, Toussaint B. High level production of secreted proteins: example of the human tissue inhibitor of metalloproteinases 1. Biochem Biophys Res Commun 2005; 337:908-15. [PMID: 16219294 DOI: 10.1016/j.bbrc.2005.09.136] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2005] [Accepted: 09/22/2005] [Indexed: 10/25/2022]
Abstract
The major difficulty for high-throughput screening of therapeutic protein candidates in experimental animal models of pathologies or for structural studies is their fast and efficient production. The tissue inhibitors of metalloproteinases (TIMPs) considered to play a role in many physiological and pathological processes, such as arthritis or cancer, by inhibiting matrix metalloproteinases or acting as signalling molecules, have always been produced with huge difficulties. We hereby propose a new method to overproduce human recombinant TIMP-1 by transient expression in HEK293E cells, followed by a one-step chromatography purification, yielding in only 2 weeks, dozens of milligrams of pure, stable, glycosylated and active protein for in vitro and in vivo studies. This easy to set up, rapid, and efficient method could be applied for any naturally secreted mammalian protein.
Collapse
Affiliation(s)
- L Crombez
- Groupe de Recherche et d'Etude du Processus Inflammatoire, MENRT EA 2938, DBPC/Enzymologie, Centre Hospitalier Universitaire de Grenoble, BP 217, 38043 Grenoble Cedex 9, France.
| | | | | | | | | | | | | |
Collapse
|
44
|
Puhakka HL, Turunen P, Rutanen J, Hiltunen MO, Turunen MP, Yla-Herttuala S. Tissue Inhibitor of Metalloproteinase 1 Adenoviral Gene Therapy Alone Is Equally Effective in Reducing Restenosis as Combination Gene Therapy in a Rabbit Restenosis Model. J Vasc Res 2005; 42:361-7. [PMID: 16037681 DOI: 10.1159/000087120] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2005] [Accepted: 05/14/2005] [Indexed: 11/19/2022] Open
Abstract
Neointimal formation is a common feature after angioplasty, bypass grafting and stenting. Angioplasty damages endothelium, causing pathological changes in arteries which lead to smooth muscle cell proliferation, synthesis of extracellular matrix components and eventually restenosis formation. Adenoviruses offer an efficient transgene expression in the vascular system. In this study, we compared the effects of different gene combinations. We wanted to find out whether adenoviral catheter-mediated delivery of an additive combination of the vascular endothelial growth factor (VEGF)-A with VEGF-C is more effective than the combination of tissue inhibitor of metalloproteinase 1 (TIMP-1) alone or with VEGF-C in a rabbit balloon denudation model. Additionally, we wanted to clarify whether the combination therapy prolongs the treatment effect. It was found that TIMP-1 alone prevents restenosis and that the combination of VEGF-A and VEGF-C has a similar effect at the 2-week time point. However, the combination of VEGF-A and VEGF-C lost the treatment effect at the 4-week time point due to the catch-up growth of neointima. On the other hand, TIMP-1 and the combination of TIMP-1 with VEGF-C still had an extended treatment effect at the 4-week time point. When considering the gene combination used in this study, it is concluded that gene therapy with adenoviral TIMP-1 alone is sufficient in reducing restenosis and that combination gene therapy does not bring any significant advantages.
Collapse
Affiliation(s)
- Hanna L Puhakka
- A.I. Virtanen Institute, University of Kuopio, Kuopio, Finland
| | | | | | | | | | | |
Collapse
|