1
|
Cytoplasmic Tail of MT1-MMP: A Hub of MT1-MMP Regulation and Function. Int J Mol Sci 2023; 24:ijms24065068. [PMID: 36982142 PMCID: PMC10049710 DOI: 10.3390/ijms24065068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/03/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
MT1-MMP (MMP-14) is a multifunctional protease that regulates ECM degradation, activation of other proteases, and a variety of cellular processes, including migration and viability in physiological and pathological contexts. Both the localization and signal transduction capabilities of MT1-MMP are dependent on its cytoplasmic domain that constitutes the final 20 C-terminal amino acids, while the rest of the protease is extracellular. In this review, we summarize the ways in which the cytoplasmic tail is involved in regulating and enacting the functions of MT1-MMP. We also provide an overview of known interactors of the MT1-MMP cytoplasmic tail and the functional significance of these interactions, as well as further insight into the mechanisms of cellular adhesion and invasion that are regulated by the cytoplasmic tail.
Collapse
|
2
|
Humeres C, Shinde AV, Hanna A, Alex L, Hernández SC, Li R, Chen B, Conway SJ, Frangogiannis NG. Smad7 effects on TGF-β and ErbB2 restrain myofibroblast activation and protect from postinfarction heart failure. J Clin Invest 2022; 132:146926. [PMID: 34905511 PMCID: PMC8803336 DOI: 10.1172/jci146926] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/09/2021] [Indexed: 01/02/2023] Open
Abstract
Repair of the infarcted heart requires TGF-β/Smad3 signaling in cardiac myofibroblasts. However, TGF-β-driven myofibroblast activation needs to be tightly regulated in order to prevent excessive fibrosis and adverse remodeling that may precipitate heart failure. We hypothesized that induction of the inhibitory Smad, Smad7, may restrain infarct myofibroblast activation, and we examined the molecular mechanisms of Smad7 actions. In a mouse model of nonreperfused infarction, Smad3 activation triggered Smad7 synthesis in α-SMA+ infarct myofibroblasts, but not in α-SMA-PDGFRα+ fibroblasts. Myofibroblast-specific Smad7 loss increased heart failure-related mortality, worsened dysfunction, and accentuated fibrosis in the infarct border zone and in the papillary muscles. Smad7 attenuated myofibroblast activation and reduced synthesis of structural and matricellular extracellular matrix proteins. Smad7 effects on TGF-β cascades involved deactivation of Smad2/3 and non-Smad pathways, without any effects on TGF-β receptor activity. Unbiased transcriptomic and proteomic analysis identified receptor tyrosine kinase signaling as a major target of Smad7. Smad7 interacted with ErbB2 in a TGF-β-independent manner and restrained ErbB1/ErbB2 activation, suppressing fibroblast expression of fibrogenic proteases, integrins, and CD44. Smad7 induction in myofibroblasts serves as an endogenous TGF-β-induced negative feedback mechanism that inhibits postinfarction fibrosis by restraining Smad-dependent and Smad-independent TGF-β responses, and by suppressing TGF-β-independent fibrogenic actions of ErbB2.
Collapse
Affiliation(s)
- Claudio Humeres
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Arti V. Shinde
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Anis Hanna
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Linda Alex
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Silvia C. Hernández
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ruoshui Li
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Bijun Chen
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Simon J. Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nikolaos G. Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
3
|
Structure-based molecular insights into matrix metalloproteinase inhibitors in cancer treatments. Future Med Chem 2021; 14:35-51. [PMID: 34779649 DOI: 10.4155/fmc-2021-0246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Protease inhibitors are of considerable interest as anticancer agents. Matrix metalloproteinases (MMPs) were the earliest type of proteases considered as anticancer targets. The developments of MMP inhibitors (MMPIs) by pharmaceutical companies can be dated from the early 1980s. Thus far, none of the over 50 MMPIs entering clinical trials have been approved. This work summarizes the reported studies on the structure of MMPs and complexes with ligands and inhibitors, based on which, the authors analyzed the clinical failures of MMPIs in a structural biological manner. Furthermore, MMPs were systematically compared with urokinase, a protease-generating plasmin, which plays similar pathological roles in cancer development; the reasons for the clinical successes of urokinase inhibitors and the clinical failures of MMPIs are discussed.
Collapse
|
4
|
The Rebirth of Matrix Metalloproteinase Inhibitors: Moving Beyond the Dogma. Cells 2019; 8:cells8090984. [PMID: 31461880 PMCID: PMC6769477 DOI: 10.3390/cells8090984] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/22/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022] Open
Abstract
The pursuit of matrix metalloproteinase (MMP) inhibitors began in earnest over three decades ago. Initial clinical trials were disappointing, resulting in a negative view of MMPs as therapeutic targets. As a better understanding of MMP biology and inhibitor pharmacokinetic properties emerged, it became clear that initial MMP inhibitor clinical trials were held prematurely. Further complicating matters were problematic conclusions drawn from animal model studies. The most recent generation of MMP inhibitors have desirable selectivities and improved pharmacokinetics, resulting in improved toxicity profiles. Application of selective MMP inhibitors led to the conclusion that MMP-2, MMP-9, MMP-13, and MT1-MMP are not involved in musculoskeletal syndrome, a common side effect observed with broad spectrum MMP inhibitors. Specific activities within a single MMP can now be inhibited. Better definition of the roles of MMPs in immunological responses and inflammation will help inform clinic trials, and multiple studies indicate that modulating MMP activity can improve immunotherapy. There is a U.S. Food and Drug Administration (FDA)-approved MMP inhibitor for periodontal disease, and several MMP inhibitors are in clinic trials, targeting a variety of maladies including gastric cancer, diabetic foot ulcers, and multiple sclerosis. It is clearly time to move on from the dogma of viewing MMP inhibition as intractable.
Collapse
|
5
|
The Expanding Role of MT1-MMP in Cancer Progression. Pharmaceuticals (Basel) 2019; 12:ph12020077. [PMID: 31137480 PMCID: PMC6630478 DOI: 10.3390/ph12020077] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/16/2019] [Accepted: 05/18/2019] [Indexed: 12/21/2022] Open
Abstract
For over 20 years, membrane type 1 matrix metalloproteinase (MT1-MMP) has been recognized as a key component in cancer progression. Initially, the primary roles assigned to MT1-MMP were the activation of proMMP-2 and degradation of fibrillar collagen. Proteomics has revealed a great array of MT1-MMP substrates, and MT1-MMP selective inhibitors have allowed for a more complete mapping of MT1-MMP biological functions. MT1-MMP has extensive sheddase activities, is both a positive and negative regulator of angiogenesis, can act intracellularly and as a transcription factor, and modulates immune responses. We presently examine the multi-faceted role of MT1-MMP in cancer, with a consideration of how the diversity of MT1-MMP behaviors impacts the application of MT1-MMP inhibitors.
Collapse
|
6
|
de Vos IJHM, Tao EY, Ong SLM, Goggi JL, Scerri T, Wilson GR, Low CGM, Wong ASW, Grussu D, Stegmann APA, van Geel M, Janssen R, Amor DJ, Bahlo M, Dunn NR, Carney TJ, Lockhart PJ, Coull BJ, van Steensel MAM. Functional analysis of a hypomorphic allele shows that MMP14 catalytic activity is the prime determinant of the Winchester syndrome phenotype. Hum Mol Genet 2019; 27:2775-2788. [PMID: 29741626 PMCID: PMC6077784 DOI: 10.1093/hmg/ddy168] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/01/2018] [Indexed: 12/23/2022] Open
Abstract
Winchester syndrome (WS, MIM #277950) is an extremely rare autosomal recessive skeletal dysplasia characterized by progressive joint destruction and osteolysis. To date, only one missense mutation in MMP14, encoding the membrane-bound matrix metalloprotease 14, has been reported in WS patients. Here, we report a novel hypomorphic MMP14 p.Arg111His (R111H) allele, associated with a mitigated form of WS. Functional analysis demonstrated that this mutation, in contrast to previously reported human and murine MMP14 mutations, does not affect MMP14’s transport to the cell membrane. Instead, it partially impairs MMP14’s proteolytic activity. This residual activity likely accounts for the mitigated phenotype observed in our patients. Based on our observations as well as previously published data, we hypothesize that MMP14’s catalytic activity is the prime determinant of disease severity. Given the limitations of our in vitro assays in addressing the consequences of MMP14 dysfunction, we generated a novel mmp14a/b knockout zebrafish model. The fish accurately reflected key aspects of the WS phenotype including craniofacial malformations, kyphosis, short-stature and reduced bone density owing to defective collagen remodeling. Notably, the zebrafish model will be a valuable tool for developing novel therapeutic approaches to a devastating bone disorder.
Collapse
Affiliation(s)
- Ivo J H M de Vos
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.,Department of Dermatology, Maastricht University Medical Center+, Maastricht 6202 AZ, The Netherlands.,School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht 6200 MD, The Netherlands
| | - Evelyn Yaqiong Tao
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Sheena Li Ming Ong
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Julian L Goggi
- Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), Singapore 138667, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117593, Singapore
| | - Thomas Scerri
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville 3052, Australia
| | - Gabrielle R Wilson
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville 3052, Australia.,Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| | - Chernis Guai Mun Low
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Arnette Shi Wei Wong
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Dominic Grussu
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Alexander P A Stegmann
- School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht 6200 MD, The Netherlands.,Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht 6229 HX, The Netherlands
| | - Michel van Geel
- Department of Dermatology, Maastricht University Medical Center+, Maastricht 6202 AZ, The Netherlands.,School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht 6200 MD, The Netherlands.,Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht 6229 HX, The Netherlands
| | - Renske Janssen
- Department of Dermatology, Maastricht University Medical Center+, Maastricht 6202 AZ, The Netherlands.,School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht 6200 MD, The Netherlands
| | - David J Amor
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville 3052, Australia.,Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| | - Melanie Bahlo
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville 3052, Australia
| | - Norris R Dunn
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore 636921, Singapore
| | - Thomas J Carney
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore 636921, Singapore.,Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
| | - Paul J Lockhart
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville 3052, Australia.,Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| | - Barry J Coull
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Maurice A M van Steensel
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.,Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK.,Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore 636921, Singapore
| |
Collapse
|
7
|
Pahwa S, Bhowmick M, Amar S, Cao J, Strongin AY, Fridman R, Weiss SJ, Fields GB. Characterization and regulation of MT1-MMP cell surface-associated activity. Chem Biol Drug Des 2018; 93:1251-1264. [PMID: 30480376 DOI: 10.1111/cbdd.13450] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/18/2018] [Accepted: 11/11/2018] [Indexed: 12/19/2022]
Abstract
Quantitative assessment of MT1-MMP cell surface-associated proteolytic activity remains undefined. Presently, MT1-MMP was stably expressed and a cell-based FRET assay developed to quantify activity toward synthetic collagen-model triple-helices. To estimate the importance of cell surface localization and specific structural domains on MT1-MMP proteolysis, activity measurements were performed using a series of membrane-anchored MT1-MMP mutants and compared directly with those of soluble MT1-MMP. MT1-MMP activity (kcat /KM ) on the cell surface was 4.8-fold lower compared with soluble MT1-MMP, with the effect largely manifested in kcat . Deletion of the MT1-MMP cytoplasmic tail enhanced cell surface activity, with both kcat and KM values affected, while deletion of the hemopexin-like domain negatively impacted KM and increased kcat . Overall, cell surface localization of MT1-MMP restricts substrate binding and protein-coupled motions (based on changes in both kcat and KM ) for catalysis. Comparison of soluble and cell surface-bound MT2-MMP revealed 12.9-fold lower activity on the cell surface. The cell-based assay was utilized for small molecule and triple-helical transition state analog MMP inhibitors, which were found to function similarly in solution and at the cell surface. These studies provide the first quantitative assessments of MT1-MMP activity and inhibition in the native cellular environment of the enzyme.
Collapse
Affiliation(s)
- Sonia Pahwa
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Manishabrata Bhowmick
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Sabrina Amar
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida.,Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, Florida
| | - Jian Cao
- Departments of Medicine/Cancer Prevention and Pathology, Stony Brook University, Stony Brook, New York
| | - Alex Y Strongin
- Cancer Research Center, Sanford Burnham Prebys Medical Research Institute, La Jolla, California
| | - Rafael Fridman
- Department of Pathology and the Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Stephen J Weiss
- Division of Molecular Medicine & Genetics, Department of Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | - Gregg B Fields
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida.,Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, Florida.,The Scripps Research Institute/Scripps Florida, Jupiter, Florida
| |
Collapse
|
8
|
Thakur V, Zhang K, Savadelis A, Zmina P, Aguila B, Welford SM, Abdul-Karim F, Bonk KW, Keri RA, Bedogni B. The membrane tethered matrix metalloproteinase MT1-MMP triggers an outside-in DNA damage response that impacts chemo- and radiotherapy responses of breast cancer. Cancer Lett 2018; 443:115-124. [PMID: 30502358 DOI: 10.1016/j.canlet.2018.11.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 01/18/2023]
Abstract
Breast cancer is the second leading cause of death among women in the US. Targeted therapies exist, however resistance is common and patients resort to chemotherapy. Chemotherapy is also a main treatment for triple negative breast cancer (TNBC) patients; while radiation is delivered to patients with advanced disease to counteract metastasis. Yet, resistance to both chemo- and radiotherapy is still frequent, highlighting a need to provide novel sensitizers. We discovered that MT1-MMP modulates DNA damage responses (DDR) in breast cancer. MT1-MMP expression inversely correlates to chemotherapy response of breast cancer patients. Inhibition of MT1-MMP sensitizes TNBC cells to IR and doxorubicin in vitro, and in vivo in an orthotopic breast cancer model. Specifically, depletion of MT1-MMP causes stalling of replication forks and Double Strand Breaks (DBSs), leading to increased sensitivity to additional genotoxic stresses. These effects are mediated by integrinβ1, as a constitutive active integrinβ1 reverts replication defects and protects cells depleted of MT1-MMP from IR and chemotherapy. These data highlight a novel DNA damage response triggered by MT1-MMP-integrinβ1 and provide a new point of therapeutic targeting that may improve breast cancer patient outcomes.
Collapse
Affiliation(s)
- Varsha Thakur
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA; Department of Dermatology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Keman Zhang
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Alyssa Savadelis
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Patrick Zmina
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA; Department of Dermatology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Brittany Aguila
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Scott M Welford
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Fadi Abdul-Karim
- Department of Anatomic Pathology, Cleveland Clinic Foundation, Cleveland, OH, 44119, USA
| | - Kristen W Bonk
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ruth A Keri
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Barbara Bedogni
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA; Department of Dermatology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
9
|
Garmon T, Wittling M, Nie S. MMP14 Regulates Cranial Neural Crest Epithelial-to-Mesenchymal Transition and Migration. Dev Dyn 2018; 247:1083-1092. [PMID: 30079980 DOI: 10.1002/dvdy.24661] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 06/08/2018] [Accepted: 07/10/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Neural crest is a vertebrate specific cell population. Induced at lateral borders of the neural plate, neural crest cells (NCCs) subsequently undergo epithelial-to-mesenchymal transition (EMT) to detach from the neuroepithelium before migrating into various locations in the embryo. Despite the wealth of knowledge of transcription factors involved in this process, little is known about the effectors that directly regulate neural crest EMT and migration. RESULTS Here, we examined the activity of matrix metalloproteinase MMP14 in NCCs and found that MMP14 is expressed in both premigratory and migrating NCCs. Overexpression of MMP14 led to premature migration of NCCs, while down-regulation of MMP14 resulted in reduced neural crest migration. Transplantation experiment further showed that MMP14 is required in NCCs, whereas MMP2, which can be activated by MMP14, is required in the surrounding mesenchyme. in vitro explant culture showed that MMP14 is required for neural crest EMT but not for spreading. This is possibly mediated by the changes in cadherin levels, as decreasing MMP14 level led to increased cadherin expression and increasing MMP14 level led to reduced cadherin expression. CONCLUSIONS The results demonstrate that MMP14 is critical for neural crest EMT and migration, partially through regulating the levels of cadherins. Developmental Dynamics 247:1083-1092, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Taylor Garmon
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Megen Wittling
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia.,Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology, Atlanta, Georgia
| | - Shuyi Nie
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia.,Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology, Atlanta, Georgia.,Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
10
|
Sakr M, Li XY, Sabeh F, Feinberg TY, Tesmer JJG, Tang Y, Weiss SJ. Tracking the Cartoon mouse phenotype: Hemopexin domain-dependent regulation of MT1-MMP pericellular collagenolytic activity. J Biol Chem 2018; 293:8113-8127. [PMID: 29643184 DOI: 10.1074/jbc.ra117.001503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/23/2018] [Indexed: 11/06/2022] Open
Abstract
Following ENU mutagenesis, a phenodeviant line was generated, termed the "Cartoon mouse," that exhibits profound defects in growth and development. Cartoon mice harbor a single S466P point mutation in the MT1-MMP hemopexin domain, a 200-amino acid segment that is thought to play a critical role in regulating MT1-MMP collagenolytic activity. Herein, we demonstrate that the MT1-MMPS466P mutation replicates the phenotypic status of Mt1-mmp-null animals as well as the functional characteristics of MT1-MMP-/- cells. However, rather than a loss-of-function mutation acquired as a consequence of defects in MT1-MMP proteolytic activity, the S466P substitution generates a misfolded, temperature-sensitive mutant that is abnormally retained in the endoplasmic reticulum (ER). By contrast, the WT hemopexin domain does not play a required role in regulating MT1-MMP trafficking, as a hemopexin domain-deletion mutant is successfully mobilized to the cell surface and displays nearly normal collagenolytic activity. Alternatively, when MT1-MMPS466P-expressing cells are cultured at a permissive temperature of 25 °C that depresses misfolding, the mutant successfully traffics from the ER to the trans-Golgi network (ER → trans-Golgi network), where it undergoes processing to its mature form, mobilizes to the cell surface, and expresses type I collagenolytic activity. Together, these analyses define the Cartoon mouse as an unexpected gain-of-abnormal function mutation, wherein the temperature-sensitive mutant phenocopies MT1-MMP-/- mice as a consequence of eliciting a specific ER → trans-Golgi network trafficking defect.
Collapse
Affiliation(s)
- Moustafa Sakr
- Molecular Diagnostics and Therapeutics Department, Genetic Engineering and Biotechnology Research institute (GEBRI), University of Sadat City, Sadat City, Egypt 32897
| | - Xiao-Yan Li
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109; Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
| | - Farideh Sabeh
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109; Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
| | - Tamar Y Feinberg
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109; Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
| | - John J G Tesmer
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109; Departments of Medicinal Chemistry, Pharmacology, and Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Yi Tang
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109; Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
| | - Stephen J Weiss
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109; Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109; Departments of Medicinal Chemistry, Pharmacology, and Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109.
| |
Collapse
|
11
|
Stawowczyk M, Wellenstein MD, Lee SB, Yomtoubian S, Durrans A, Choi H, Narula N, Altorki NK, Gao D, Mittal V. Matrix Metalloproteinase 14 promotes lung cancer by cleavage of Heparin-Binding EGF-like Growth Factor. Neoplasia 2016; 19:55-64. [PMID: 28013056 PMCID: PMC5198728 DOI: 10.1016/j.neo.2016.11.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/31/2016] [Accepted: 11/07/2016] [Indexed: 11/24/2022]
Abstract
Molecularly targeted therapies benefit approximately 15–20% of non-small cell lung cancer (NSCLC) patients carrying specific drug-sensitive mutations. Thus, there is a clinically unmet need for the identification of novel targets for drug development. Here, we performed RNA-deep sequencing to identify altered gene expression between malignant and non-malignant lung tissue. Matrix Metalloproteinase 14 (MMP14), a membrane-bound proteinase, was significantly up-regulated in the tumor epithelial cells and intratumoral myeloid compartments in both mouse and human NSCLC. Overexpression of a soluble dominant negative MMP14 (DN-MMP14) or pharmacological inhibition of MMP14 blocked invasion of lung cancer cells through a collagen I matrix in vitro and reduced tumor incidence in an orthotopic K-RasG12D/+p53−/− mouse model of lung cancer. Additionally, MMP14 activity mediated proteolytic processing and activation of Heparin-Binding EGF-like Growth Factor (HB-EGF), stimulating the EGFR signaling pathway to increase proliferation and tumor growth. This study highlights the potential for development of therapeutic strategies that target MMP14 in NSCLC with particular focus on MMP14-HB-EGF axis.
Collapse
Affiliation(s)
- Marcin Stawowczyk
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA
| | - Max D Wellenstein
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA
| | - Sharrell B Lee
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA
| | - Shira Yomtoubian
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Department of pharmacology, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA
| | - Anna Durrans
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA
| | - Hyejin Choi
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA
| | - Navneet Narula
- Department of Pathology, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA
| | - Nasser K Altorki
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA
| | - Dingcheng Gao
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA.
| | - Vivek Mittal
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA; Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, 525 East 68th Street, NY, New York 10065, USA.
| |
Collapse
|
12
|
Thakur V, Bedogni B. The membrane tethered matrix metalloproteinase MT1-MMP at the forefront of melanoma cell invasion and metastasis. Pharmacol Res 2016; 111:17-22. [DOI: 10.1016/j.phrs.2016.05.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 01/18/2023]
|
13
|
Roth E, Cao J. miR-181 suppresses metastasis via MMP-14. Aging (Albany NY) 2015; 7:740-1. [PMID: 26527609 PMCID: PMC4637197 DOI: 10.18632/aging.100824] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 10/18/2015] [Indexed: 12/01/2022]
Affiliation(s)
- Eric Roth
- Department of Pharmacology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jian Cao
- Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
14
|
Terawaki S, Kitano K, Aoyama M, Mori T, Hakoshima T. MT1‐MMP recognition by ERM proteins and its implication in CD44 shedding. Genes Cells 2015; 20:847-59. [DOI: 10.1111/gtc.12276] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/05/2015] [Indexed: 01/21/2023]
Affiliation(s)
- Shin‐ichi Terawaki
- Structural Biology Laboratory Nara Institute of Science and Technology 8916‐5 Takayama Ikoma Nara 630‐0192 Japan
| | - Ken Kitano
- Structural Biology Laboratory Nara Institute of Science and Technology 8916‐5 Takayama Ikoma Nara 630‐0192 Japan
| | - Miki Aoyama
- Structural Biology Laboratory Nara Institute of Science and Technology 8916‐5 Takayama Ikoma Nara 630‐0192 Japan
| | - Tomoyuki Mori
- Structural Biology Laboratory Nara Institute of Science and Technology 8916‐5 Takayama Ikoma Nara 630‐0192 Japan
| | - Toshio Hakoshima
- Structural Biology Laboratory Nara Institute of Science and Technology 8916‐5 Takayama Ikoma Nara 630‐0192 Japan
| |
Collapse
|
15
|
Han KY, Dugas-Ford J, Lee H, Chang JH, Azar DT. MMP14 Cleavage of VEGFR1 in the Cornea Leads to a VEGF-Trap Antiangiogenic Effect. Invest Ophthalmol Vis Sci 2015; 56:5450-6. [PMID: 26284550 PMCID: PMC4544186 DOI: 10.1167/iovs.14-16248] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 05/28/2015] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To determine the possible antiangiogenic effect of metalloproteinase (MMP) 14 cleavage of vascular endothelial growth factor receptor 1 (VEGFR1) in the cornea. METHODS Recombinant mouse (rm) VEGFR1 was incubated with various concentrations of recombinant MMP14 to examine proteolysis in vitro. The reaction mixture was analyzed by SDS-PAGE and stained with Coomassie blue. The fragments resulting from rmVEGFR1 cleavage by MMP14 were subjected to Edman degradation, and the amino acid sequences were aligned with rmVEGFR1 sequences. Surface plasmon resonance was used to determine the equilibrium dissociation constant (KD) between MMP14 and rmVEGFR1. The KD value of rmVEGFR1 and the 59.8-kDa cleavage product binding to VEGF-A₁₆₅ was also determined. Cell proliferation assays were performed in the presence of VEGF-A₁₆₅ plus the 59.8-kDa VEGFR1 fragment or VEGF-A₁₆₅ alone. RESULTS Matrix metalloproteinase 14 binds and cleaves rmVEGFR1 to produce 59.8-kDa (N-terminal fragment, Ig domains 1-5), 35-kDa (C-terminal fragment containing IgG and His-tag), and 21-kDa (Ig domains 6-7) fragments. The 59.8-kDa fragment showed binding to VEGF-A₁₆₅ and inhibited VEGF-induced endothelial cell mitogenesis. CONCLUSIONS Our findings suggest that VEGFR1 cleavage by MMP14 in the cornea leads to a VEGF-trap effect, reducing the proangiogenic effect of VEGF-A₁₆₅, thereby reducing corneal angiogenesis.
Collapse
Affiliation(s)
- Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Jennifer Dugas-Ford
- Department of Ophthalmology and Visual Sciences Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Hyun Lee
- Center for Pharmaceutical Biotechnology and Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Dimitri T. Azar
- Department of Ophthalmology and Visual Sciences Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, United States
| |
Collapse
|
16
|
Li Y, Kuscu C, Banach A, Zhang Q, Pulkoski-Gross A, Kim D, Liu J, Roth E, Li E, Shroyer KR, Denoya PI, Zhu X, Chen L, Cao J. miR-181a-5p Inhibits Cancer Cell Migration and Angiogenesis via Downregulation of Matrix Metalloproteinase-14. Cancer Res 2015; 75:2674-85. [PMID: 25977338 DOI: 10.1158/0008-5472.can-14-2875] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 03/10/2015] [Indexed: 01/18/2023]
Abstract
Upregulation of matrix metalloproteinase MMP-14 (MT1-MMP) is associated with poor prognosis in cancer patients, but it is unclear how MMP-14 becomes elevated in tumors. Here, we show that miR-181a-5p is downregulated in aggressive human breast and colon cancers where its levels correlate inversely with MMP-14 expression. In clinical specimens, enhanced expression of MMP-14 was observed in cancer cells located at the invasive front of tumors where miR-181a-5p was downregulated relative to adjacent normal cells. Bioinformatics analyses defined a potential miR-181a-5p response element within the 3'-untranslated region of MMP-14 that was validated in reporter gene experiments. Ectopic miR-181a-5p reduced MMP-14 expression, whereas miR-181a-5p attenuation elevated MMP-14 expression. In support of a critical relationship between these two genes, miR-181a-5p-mediated reduction of MMP-14 levels was sufficient to decrease cancer cell migration, invasion, and activation of pro-MMP-2. Furthermore, this reduction in MMP-14 levels was sufficient to reduce in vivo invasion and angiogenesis in chick chorioallantoic membrane assays. Taken together, our results establish the regulation of MMP-14 in cancers by miR-181a-5p through a posttranscriptional mechanism, and they further suggest strategies to elevate miR-181a-5p to prevent cancer metastasis.
Collapse
Affiliation(s)
- Yiyi Li
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York. Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cem Kuscu
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York
| | - Anna Banach
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York
| | - Qian Zhang
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York
| | | | - Deborah Kim
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York
| | - Jingxuan Liu
- Department of Pathology, Stony Brook University, Stony Brook, New York
| | - Eric Roth
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York
| | - Ellen Li
- Department of Medicine/Division of Gastroenterology and Hepatology, Stony Brook University, Stony Brook, New York
| | - Kenneth R Shroyer
- Department of Pathology, Stony Brook University, Stony Brook, New York
| | - Paula I Denoya
- Department of Surgery, Stony Brook University, Stony Brook, New York
| | - Xiaoxia Zhu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Longhua Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jian Cao
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York.
| |
Collapse
|
17
|
Gao Y, Amar S, Pahwa S, Fields G, Kodadek T. Rapid lead discovery through iterative screening of one bead one compound libraries. ACS COMBINATORIAL SCIENCE 2015; 17:49-59. [PMID: 25434974 PMCID: PMC4294594 DOI: 10.1021/co500154e] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Primary hits that arise from screening one bead one compound (OBOC) libraries against a target of interest rarely have high potency. However, there has been little work focused on the development of an efficient workflow for primary hit improvement. In this study, we show that by characterizing the binding constants for all of the hits that arise from a screen, structure-activity relationship (SAR) data can be obtained to inform the design of "derivative libraries" of a primary hit that can then be screened under more demanding conditions to obtain improved compounds. Here, we demonstrate the rapid improvement of a primary hit against matrix metalloproteinase-14 using this approach.
Collapse
Affiliation(s)
- Yu Gao
- Departments
of Chemistry and Cancer Biology The Scripps Research Institutem 130
Scripps Way, Jupiter, Florida 33458, United States
| | - Sabrina Amar
- Torrey Pines Institute for Molecular Studies 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Sonia Pahwa
- Torrey Pines Institute for Molecular Studies 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Gregg Fields
- Torrey Pines Institute for Molecular Studies 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Thomas Kodadek
- Departments
of Chemistry and Cancer Biology The Scripps Research Institutem 130
Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
18
|
Ohkawara H, Ishibashi T, Sugimoto K, Ikeda K, Ogawa K, Takeishi Y. Membrane type 1-matrix metalloproteinase/Akt signaling axis modulates TNF-α-induced procoagulant activity and apoptosis in endothelial cells. PLoS One 2014; 9:e105697. [PMID: 25162582 PMCID: PMC4146507 DOI: 10.1371/journal.pone.0105697] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 07/28/2014] [Indexed: 11/18/2022] Open
Abstract
Membrane type 1–matrix metalloproteinase (MT1-MMP) functions as a signaling molecule in addition to a proteolytic enzyme. Our hypothesis was that MT1-MMP cooperates with protein kinase B (Akt) in tumor necrosis factor (TNF)-α-induced signaling pathways of vascular responses, including tissue factor (TF) procoagulant activity and endothelial apoptosis, in cultured human aortic endothelial cells (ECs). TNF-α (10 ng/mL) induced a decrease in Akt phosphorylation within 60 minutes in ECs. A chemical inhibitor of MMP, TIMP-2 and selective small interfering RNA (siRNA)-mediated suppression of MT1-MMP reversed TNF-α-triggered transient decrease of Akt phosphorylation within 60 minutes, suggesting that MT1-MMP may be a key regulator of Akt phosphorylation in TNF-α-stimulated ECs. In the downstream events, TNF-α increased TF antigen and activity, and suppressed the expression of thrombomodulin (TM) antigen. Inhibition of Akt markedly enhanced TNF-α-induced expression of TF antigen and activity, and further reduced the expression of TM antigen. Silencing of MT1-MMP by siRNA also reversed the changed expression of TF and TM induced by TNF-α. Moreover, TNF-α induced apoptosis of ECs through Akt- and forkhead box protein O1 (FoxO1)-dependent signaling pathway and nuclear factor-kB (NF-kB) activation. Knockdown of MT1-MMP by siRNA reversed apoptosis of ECs by inhibiting TNF-α-induced Akt-dependent regulation of FoxO1 in TNF-α-stimulated ECs. Immunoprecipitation demonstrated that TNF-α induced the changes in the associations between the cytoplasmic fraction of MT1-MMP and Akt in ECs. In conclusion, we show new evidence that MT1-MMP/Akt signaling axis is a key modifier for TNF-α-induced signaling pathways for modulation of procoagulant activity and apoptosis of ECs.
Collapse
Affiliation(s)
- Hiroshi Ohkawara
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
- * E-mail:
| | - Toshiyuki Ishibashi
- Department of Cardiovascular Medicine, Ohara General Hospital Medical Center, Fukushima, Japan
| | - Koichi Sugimoto
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Kazuhiko Ikeda
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Kazuei Ogawa
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Yasuchika Takeishi
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
19
|
Shaverdashvili K, Wong P, Ma J, Zhang K, Osman I, Bedogni B. MT1-MMP modulates melanoma cell dissemination and metastasis through activation of MMP2 and RAC1. Pigment Cell Melanoma Res 2014; 27:287-96. [PMID: 24387669 DOI: 10.1111/pcmr.12201] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 12/02/2013] [Indexed: 12/11/2022]
Abstract
Metastatic melanoma remains the deadliest of all skin cancers with a survival rate at five years of less than 15%. MT1-MMP is a membrane-associated matrix metalloproteinase that controls pericellular proteolysis and is an important, invasion-promoting, pro-tumorigenic MMP in cancer. We show that deregulation of MT1-MMP expression happens as early as the transition from nevus to primary melanoma and continues to increase during melanoma progression. Furthermore, MT1-MMP expression is associated with poor melanoma patient outcome, underscoring a pivotal role of MT1-MMP in melanoma pathogenesis. We demonstrate that MT1-MMP is directly required for melanoma cells to metastasize, as cells deprived of MT1-MMP fail to form distant metastasis in an orthotopic mouse melanoma model. We show that MT1-MMP affects cell invasion by activating its target MMP2. Importantly, we demonstrate, for the first time, that activation of MMP2 by MT1-MMP is required to sustain RAC1 activity and promote MT1-MMP-dependent cell motility. These data highlight a novel MT1-MMP/MMP2/RAC1 signaling axis in melanoma that may represent an intriguing molecular target for the treatment of invasive melanoma.
Collapse
Affiliation(s)
- Khvaramze Shaverdashvili
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | | | | | | |
Collapse
|
20
|
Evensen NA, Li J, Yang J, Yu X, Sampson NS, Zucker S, Cao J. Development of a high-throughput three-dimensional invasion assay for anti-cancer drug discovery. PLoS One 2013; 8:e82811. [PMID: 24349367 PMCID: PMC3859626 DOI: 10.1371/journal.pone.0082811] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 11/06/2013] [Indexed: 01/15/2023] Open
Abstract
The lack of three-dimensional (3-D) high-throughput (HT) screening assays designed to identify anti-cancer invasion drugs is a major hurdle in reducing cancer-related mortality, with the key challenge being assay standardization. Presented is the development of a novel 3-D invasion assay with HT potential that involves surrounding cell-collagen spheres within collagen to create a 3-D environment through which cells can invade. Standardization was achieved by designing a tooled 96-well plate to create a precisely designated location for the cell-collagen spheres and by using dialdehyde dextran to inhibit collagen contraction, maintaining uniform size and shape. This permits automated readout for determination of the effect of inhibitory compounds on cancer cell invasion. Sensitivity was demonstrated by the ability to distinguish varying levels of invasiveness of cancer cell lines, and robustness was determined by calculating the Z-factor. A Z-factor of 0.65 was obtained by comparing the effects of DMSO and anti-β1-integrin antibody, an inhibitory reagent, on the invasion of Du145 cancer cells, suggesting this novel assay is suitable for large scale drug discovery. As proof of principle, the NCI Diversity Compound Library was screened against human invasive cancer cells. Nine compounds exhibiting high potency and low toxicity were identified, including DX-52-1, a compound previously reported to inhibit cell migration, a critical determinant of cancer invasion. The results indicate that this innovative HT platform is a simple, precise, and easy to replicate 3-D invasion assay for anti-cancer drug discovery.
Collapse
Affiliation(s)
- Nikki A. Evensen
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York, United States of America
| | - Jian Li
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York, United States of America
| | - Jie Yang
- Department of Preventive Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Xiaojun Yu
- Department of Tissue Engineering, Stevens Institute of Technology, Hoboken, New Jersey, United States of America
| | - Nicole S. Sampson
- Department of Chemistry, Stony Brook University, Stony Brook, New York, United States of America, and
| | - Stanley Zucker
- Department of Research, Veterans Affair Medical Center, Northport, New York, United States of America
| | - Jian Cao
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail:
| |
Collapse
|
21
|
Evensen NA, Kuscu C, Nguyen HL, Zarrabi K, Dufour A, Kadam P, Hu YJ, Pulkoski-Gross A, Bahou WF, Zucker S, Cao J. Unraveling the role of KIAA1199, a novel endoplasmic reticulum protein, in cancer cell migration. J Natl Cancer Inst 2013; 105:1402-16. [PMID: 23990668 DOI: 10.1093/jnci/djt224] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cell migration is a critical determinant of cancer metastasis, and a better understanding of the genes involved will lead to the identification of novel targets aimed at preventing cancer dissemination. KIAA1199 has been shown to be upregulated in human cancers, yet its role in cancer progression was hitherto unknown. METHODS Clinical relevance was assessed by examining KIAA1199 expression in human cancer specimens. In vitro and in vivo studies were employed to determine the function of KIAA1199 in cancer progression. Cellular localization of KIAA1199 was microscopically determined. SNAP-tag pull-down assays were used to identify binding partner(s) of KIAA1199. Calcium levels were evaluated using spectrofluorometric and fluorescence resonance energy transfer analyses. Signaling pathways were dissected by Western blotting. Student t test was used to assess differences. All statistical tests were two-sided. RESULTS KIAA1199 was upregulated in invasive breast cancer specimens and inversely associated with patient survival rate. Silencing of KIAA1199 in MDA-MB-435 cancer cells resulted in a mesenchymal-to-epithelial transition that reduced cell migratory ability in vitro (75% reduction; P < .001) and decreased metastasis in vivo (80% reduction; P < .001). Gain-of-function assays further demonstrated the role of KIAA1199 in cell migration. KIAA1199-enhanced cell migration required endoplasmic reticulum (ER) localization, where it forms a stable complex with the chaperone binding immunoglobulin protein (BiP). A novel ER-retention motif within KIAA1199 that is required for its ER localization, BiP interaction, and enhanced cell migration was identified. Mechanistically, KIAA1199 was found to mediate ER calcium leakage, and the resultant increase in cytosolic calcium ultimately led to protein kinase C alpha activation and cell migration. CONCLUSIONS KIAA1199 serves as a novel cell migration-promoting gene and plays a critical role in maintaining cancer mesenchymal status.
Collapse
Affiliation(s)
- Nikki A Evensen
- Affiliations of authors: Department of Medicine/Cancer Prevention (NAE, CK, H-LN, KZ, AD, AP-G, JC), Department of Pathology (YH, JC), Department of Medicine/Hematology & Oncology (WFB, SZ), Stony Brook University, Stony Brook, NY; Department of Research, Veterans Affair Medical Center, Northport, NY (H-LN, PK, SZ); Centre for Blood Research and Departments of Biochemistry and Molecular Biology and Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC, Canada (AD)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Murray MY, Birkland TP, Howe JD, Rowan AD, Fidock M, Parks WC, Gavrilovic J. Macrophage migration and invasion is regulated by MMP10 expression. PLoS One 2013; 8:e63555. [PMID: 23691065 PMCID: PMC3653827 DOI: 10.1371/journal.pone.0063555] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 04/03/2013] [Indexed: 12/31/2022] Open
Abstract
This study was designed to identify metalloproteinase determinants of macrophage migration and led to the specific hypothesis that matrix metalloproteinase 10 (MMP10/stromelysin-2) facilitates macrophage migration. We first profiled expression of all MMPs in LPS-stimulated primary murine bone marrow-derived macrophages and Raw264.7 cells and found that MMP10 was stimulated early (3 h) and down-regulated later (24 h). Based on this pattern of expression, we speculated that MMP10 plays a role in macrophage responses, such as migration. Indeed, using time lapse microscopy, we found that RNAi silencing of MMP10 in primary macrophages resulted in markedly reduced migration, which was reversed with exogenous active MMP10 protein. Mmp10 (-/-) bone marrow-derived macrophages displayed significantly reduced migration over a two-dimensional fibronectin matrix. Invasion of primary wild-type macrophages into Matrigel supplemented with fibronectin was also markedly impaired in Mmp10 (-/-) cells. MMP10 expression in macrophages thus emerges as an important moderator of cell migration and invasion. These findings support the hypothesis that MMP10 promotes macrophage movement and may have implications in understanding the control of macrophages in several pathologies, including the abnormal wound healing response associated with pro-inflammatory conditions.
Collapse
Affiliation(s)
- Megan Y. Murray
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
| | - Timothy P. Birkland
- Center for Lung Biology, University of Washington, Seattle, Washington, United States of America
| | - Jonathan D. Howe
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
| | - Andrew D. Rowan
- Musculoskeletal Research Group, Institute of Cellular Medicine, The Medical School, Newcastle University, Newcastle, United Kingdom
| | - Mark Fidock
- Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| | - William C. Parks
- Center for Lung Biology, University of Washington, Seattle, Washington, United States of America
| | - Jelena Gavrilovic
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
- * E-mail:
| |
Collapse
|
23
|
Barcelona PF, Jaldín-Fincati JR, Sánchez MC, Chiabrando GA. Activated α2-macroglobulin induces Müller glial cell migration by regulating MT1-MMP activity through LRP1. FASEB J 2013; 27:3181-97. [PMID: 23640058 DOI: 10.1096/fj.12-221598] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In retinal proliferative diseases, Müller glial cells (MGCs) acquire migratory abilities. However, the mechanisms that regulate this migration remain poorly understood. In addition, proliferative disorders associated with enhanced activities of matrix metalloprotease 2 (MMP-2) and MMP-9 also present increased levels of the protease inhibitor α2-macroglobulin (α2M) and its receptor, the low-density lipoprotein receptor-related protein 1 (LRP1). In the present work, we investigated whether the protease activated form of α2M, α2M*, and LRP1 are involved with the MGC migratory process. By performing wound-scratch migration and zymography assays, we demonstrated that α2M* induced cell migration and proMMP-2 activation in the human Müller glial cell line, MIO-M1. This induction was blocked when LRP1 and MT1-MMP were knocked down with siRNA techniques. Using fluorescence microscopy and biochemical procedures, we found that α2M* induced an increase in LRP1 and MT1-MMP accumulation in early endosomes, followed by endocytic recycling and intracellular distribution of MT1-MMP toward cellular protrusions. Moreover, Rab11-dominant negative mutant abrogated MT1-MMP recycling pathway, cell migration, and proMMP-2 activation induced by α2M*. In conclusion, α2M*, through its receptor LRP1, induces cellular migration of Müller glial cells by a mechanism that involves MT1-MMP intracellular traffic to the plasma membrane by a Rab11-dependent recycling pathway.
Collapse
Affiliation(s)
- Pablo F Barcelona
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Cientificas y Tecnicas (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, (5000) Córdoba, Argentina
| | | | | | | |
Collapse
|
24
|
Dissecting Major Signaling Pathways throughout the Development of Prostate Cancer. Prostate Cancer 2013; 2013:920612. [PMID: 23738079 PMCID: PMC3657461 DOI: 10.1155/2013/920612] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 03/25/2013] [Accepted: 03/28/2013] [Indexed: 01/28/2023] Open
Abstract
Prostate cancer (PCa) is one of the most common malignancies found in males. The development of PCa involves several mutations in prostate epithelial cells, usually linked to developmental changes, such as enhanced resistance to apoptotic death, constitutive proliferation, and, in some cases, to differentiation into an androgen deprivation-resistant phenotype, leading to the appearance of castration-resistant PCa (CRPCa), which leads to a poor prognosis in patients. In this review, we summarize recent findings concerning the main deregulations into signaling pathways that will lead to the development of PCa and/or CRPCa. Key mutations in some pathway molecules are often linked to a higher prevalence of PCa, by directly affecting the respective cascade and, in some cases, by deregulating a cross-talk node or junction along the pathways. We also discuss the possible environmental and nonenvironmental inducers for these mutations, as well as the potential therapeutic strategies targeting these signaling pathways. A better understanding of how some risk factors induce deregulation of these signaling pathways, as well as how these deregulated pathways affect the development of PCa and CRPCa, will further help in the development of new treatments and prevention strategies for this disease.
Collapse
|
25
|
Abstract
The matrix metalloproteinases (MMPs) exhibit a broad array of activities, some catalytic and some non-catalytic in nature. An overall lack of selectivity has rendered small molecule, active site targeted MMP inhibitors problematic in execution. Inhibitors that favor few or individual members of the MMP family often take advantage of interactions outside the enzyme active site. We presently focus on peptide-based MMP inhibitors and probes that do not incorporate conventional Zn2+ binding groups. In some cases, these inhibitors and probes function by binding only secondary binding sites (exosites), while others bind both exosites and the active site. A myriad of MMP mediated-activities beyond selective catalysis can be inhibited by peptides, particularly cell adhesion, proliferation, motility, and invasion. Selective MMP binding peptides comprise highly customizable, unique imaging agents. Areas of needed improvement for MMP targeting peptides include binding affinity and stability.
Collapse
|
26
|
Li J, Zucker S, Pulkoski-Gross A, Kuscu C, Karaayvaz M, Ju J, Yao H, Song E, Cao J. Conversion of stationary to invasive tumor initiating cells (TICs): role of hypoxia in membrane type 1-matrix metalloproteinase (MT1-MMP) trafficking. PLoS One 2012; 7:e38403. [PMID: 22679501 PMCID: PMC3367975 DOI: 10.1371/journal.pone.0038403] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 05/04/2012] [Indexed: 01/11/2023] Open
Abstract
Emerging evidence has implicated the role of tumor initiating cells (TICs) in the process of cancer metastasis. The mechanism underlying the conversion of TICs from stationary to invasive remains to be characterized. In this report, we employed less invasive breast cancer TICs, SK-3rd, that displays CD44high/CD24low with high mammosphere-forming and tumorigenic capacities, to investigate the mechanism by which stationary TICs are converted to invasive TICs. Invasive ability of SK-3rd TICs was markedly enhanced when the cells were cultured under hypoxic conditions. Given the role of membrane type 1-matrix metalloproteinase (MT1-MMP) in cancer invasion/metastasis, we explored a possible involvement of MT1-MMP in hypoxia-induced TIC invasion. Silencing of MT1-MMP by a shRNA approach resulted in diminution of hypoxia-induced cell invasion in vitro and metastasis in vivo. Under hypoxic conditions, MT1-MMP redistributed from cytoplasmic storage pools to the cell surface of TICs, which coincides with the increased cell invasion. In addition, CD44, a cancer stem-like cell marker, inversely correlated with increased cell surface MT1-MMP. Interestingly, cell surface MT1-MMP gradually disappeared when the hypoxia-treated cells were switched to normoxia, suggesting the plasticity of TICs in response to oxygen content. Furthermore, we dissected the pathways leading to upregulated MT1-MMP in cytoplasmic storage pools under normoxic conditions, by demonstrating a cascade involving Twist1-miR10b-HoxD10 leading to enhanced MT1-MMP expression in SK-3rd TICs. These observations suggest that MT1-MMP is a key molecule capable of executing conversion of stationary TICs to invasive TICs under hypoxic conditions and thereby controlling metastasis.
Collapse
Affiliation(s)
- Jian Li
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York, United States of America
| | - Stanley Zucker
- Department of Research, Veterans Affair Medical Center, Northport, New York, United States of America
- * E-mail: (JC); (SZ)
| | - Ashleigh Pulkoski-Gross
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York, United States of America
| | - Cem Kuscu
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York, United States of America
| | - Mihriban Karaayvaz
- Department of Pathology, Stony Brook University, Stony Brook, New York, United States of America
| | - Jingfang Ju
- Department of Pathology, Stony Brook University, Stony Brook, New York, United States of America
| | - Herui Yao
- Department of Breast Surgery, the Memorial Hospital of Sun-Yat-Sen University, Guangzhou, China
| | - Erwei Song
- Department of Breast Surgery, the Memorial Hospital of Sun-Yat-Sen University, Guangzhou, China
| | - Jian Cao
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York, United States of America
- Department of Pathology, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail: (JC); (SZ)
| |
Collapse
|
27
|
Shimizu-Hirota R, Xiong W, Baxter BT, Kunkel SL, Maillard I, Chen XW, Sabeh F, Liu R, Li XY, Weiss SJ. MT1-MMP regulates the PI3Kδ·Mi-2/NuRD-dependent control of macrophage immune function. Genes Dev 2012; 26:395-413. [PMID: 22345520 DOI: 10.1101/gad.178749.111] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Macrophages play critical roles in events ranging from host defense to obesity and cancer, where they infiltrate affected tissues and orchestrate immune responses in tandem with the remodeling of the extracellular matrix (ECM). Despite the dual roles played by macrophages in inflammation, the functions of macrophage-derived proteinases are typically relegated to tissue-invasive or -degradative events. Here we report that the membrane-tethered matrix metalloenzyme MT1-MMP not only serves as an ECM-directed proteinase, but unexpectedly controls inflammatory gene responses wherein MT1-MMP(-/-) macrophages mount exaggerated chemokine and cytokine responses to immune stimuli both in vitro and in vivo. MT1-MMP modulates inflammatory responses in a protease-independent fashion in tandem with its trafficking to the nuclear compartment, where it triggers the expression and activation of a phosphoinositide 3-kinase δ (PI3Kδ)/Akt/GSK3β signaling cascade. In turn, MT1-MMP-dependent PI3Kδ activation regulates the immunoregulatory Mi-2/NuRD nucleosome remodeling complex that is responsible for controlling macrophage immune response. These findings identify a novel role for nuclear MT1-MMP as a previously unsuspected transactivator of signaling networks central to macrophage immune responses.
Collapse
Affiliation(s)
- Ryoko Shimizu-Hirota
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, 48109, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Remacle AG, Golubkov VS, Shiryaev SA, Dahl R, Stebbins JL, Chernov AV, Cheltsov AV, Pellecchia M, Strongin AY. Novel MT1-MMP small-molecule inhibitors based on insights into hemopexin domain function in tumor growth. Cancer Res 2012; 72:2339-49. [PMID: 22406620 DOI: 10.1158/0008-5472.can-11-4149] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Membrane type-1 matrix metalloproteinase (MT1-MMP) is a promising drug target in malignancy. The structure of MT1-MMP includes the hemopexin domain (PEX) that is distinct from and additional to the catalytic domain. Current MMP inhibitors target the conserved active site in the catalytic domain and, as a result, repress the proteolytic activity of multiple MMPs instead of MT1-MMP alone. In our search for noncatalytic inhibitors of MT1-MMP, we compared the protumorigenic activity of wild-type MT1-MMP with an MT1-MMP mutant lacking PEX (ΔPEX). In contrast to MT1-MMP, ΔPEX did not support tumor growth in vivo, and its expression resulted in small fibrotic tumors that contained increased levels of collagen. Because these findings suggested an important role for PEX in tumor growth, we carried out an inhibitor screen to identify small molecules targeting the PEX domain of MT1-MMP. Using the Developmental Therapeutics Program (National Cancer Institute/NIH), virtual ligand screening compound library as a source and the X-ray crystal structure of PEX as a target, we identified and validated a novel PEX inhibitor. Low dosage, intratumoral injections of PEX inhibitor repressed tumor growth and caused a fibrotic, ΔPEX-like tumor phenotype in vivo. Together, our findings provide a preclinical proof of principle rationale for the development of novel and selective MT1-MMP inhibitors that specifically target the PEX domain.
Collapse
Affiliation(s)
- Albert G Remacle
- Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Basu B, Correa de Sampaio P, Mohammed H, Fogarasi M, Corrie P, Watkins NA, Smethurst PA, English WR, Ouwehand WH, Murphy G. Inhibition of MT1-MMP activity using functional antibody fragments selected against its hemopexin domain. Int J Biochem Cell Biol 2011; 44:393-403. [PMID: 22138224 DOI: 10.1016/j.biocel.2011.11.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 11/04/2011] [Accepted: 11/17/2011] [Indexed: 01/07/2023]
Abstract
The membrane associated MMP, MT1-MMP, is a critical pericellular protease involved in tumour cell invasion and angiogenesis and is highly up-regulated in numerous human cancers. It therefore represents an exciting new therapeutic cancer-specific target. We have generated recombinant human scFv antibodies against the non-catalytic, hemopexin domain of MT1-MMP that modulate its interactions with collagen. One of these is an effective inhibitor of the invasive capacity of cancer cells and of angiogenesis in model systems. This demonstrates that targeting sites outside the catalytic domain presents a potential novel approach to proteinase inhibition that could have applications in cancer therapeutics.
Collapse
Affiliation(s)
- B Basu
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Mahimkar R, Alfonso-Jaume MA, Cape LM, Dahiya R, Lovett DH. Graded activation of the MEK1/MT1-MMP axis determines renal epithelial cell tumor phenotype. Carcinogenesis 2011; 32:1806-14. [PMID: 21965271 DOI: 10.1093/carcin/bgr216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Activation of Raf/Ras/mitogen-activated protein kinase (MEK)/mitogen-activated protein kinase signaling and elevated expression of membrane type-1 matrix metalloproteinase (MT1-MMP) are associated with von Hippel-Lindau gene alterations in renal cell carcinoma. We postulated that the degree of MEK activation was related to graded expression of MT1-MMP and the resultant phenotype of renal epithelial tumors. Madin Darby canine kidney epithelial cells transfected with a MEK1 expression plasmid yielded populations with morphologic phenotypes ranging from epithelial, mixed epithelial/mesenchymal to mesenchymal. Clones were analyzed for MEK1 activity, MT1-MMP expression and extent of epithelial-mesenchymal transition. Phenotypes of the MDCK-MEK1 clones were evaluated in vivo with nu/nu mice. Tissue microarray of renal cell cancers was quantitatively assessed for expression of phosphorylated MEK1 and MT1-MMP proteins and correlations drawn to Fuhrman nuclear grade. Graded increases in the MEK signaling module were associated with graded induction of epithelial-mesenchymal transition of the MDCK cells and induction of MT1-MMP transcription and synthesis. Inhibition of MEK1 and MT1-MMP activity reversed the epithelial-mesenchymal transition. Tumors generated by epithelial, mixed epithelial/mesenchymal and mesenchymal MDCK clones demonstrated a gradient of phenotypes extending from well-differentiated, fully encapsulated non-invasive tumors to tumors with an anaplastic morphology, high Fuhrman nuclear score, neoangiogenesis and invasion. Tumor microarray demonstrated a statistically significant association between the extent of phosphorylated MEK1, MT1-MMP expression and nuclear grade. We conclude that graded increases in the MEK1 signaling module are correlated with M1-MMP expression, renal epithelial cell tumor phenotype, invasive activity and nuclear grade. Phosphorylated MEK1 and MT1-MMP may represent novel, and mechanistic, biomarkers for the assessment of renal cell carcinoma.
Collapse
Affiliation(s)
- Rajeev Mahimkar
- Department of Medicine, San Francisco Department of Veterans Affairs Medical Center, University of California, San Francisco, CA 94121, USA
| | | | | | | | | |
Collapse
|
31
|
Nguyen HL, Zucker S, Zarrabi K, Kadam P, Schmidt C, Cao J. Oxidative stress and prostate cancer progression are elicited by membrane-type 1 matrix metalloproteinase. Mol Cancer Res 2011; 9:1305-18. [PMID: 21849471 DOI: 10.1158/1541-7786.mcr-11-0033] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Oxidative stress caused by high levels of reactive oxygen species (ROS) has been correlated with prostate cancer aggressiveness. Expression of membrane-type 1 matrix metalloproteinase (MT1-MMP), which has been implicated in cancer invasion and metastasis, is associated with advanced prostate cancer. We show here that MT1-MMP plays a key role in eliciting oxidative stress in prostate cancer cells. Stable MT1-MMP expression in less invasive LNCaP prostate cancer cells with low endogenous MT1-MMP increased activity of ROS, whereas MT1-MMP knockdown in DU145 cells with high endogenous MT1-MMP decreased activity of ROS. Expression of MT1-MMP increased oxidative DNA damage in LNCaP and in DU145 cells, indicating that MT1-MMP-mediated induction of ROS caused oxidative stress. MT1-MMP expression promoted a more aggressive phenotype in LNCaP cells that was dependent on elaboration of ROS. Blocking ROS activity using the ROS scavenger N-acetylcysteine abrogated MT1-MMP-mediated increase in cell migration and invasion. MT1-MMP-expressing LNCaP cells displayed an enhanced ability to grow in soft agar that required increased ROS. Using cells expressing MT1-MMP mutant cDNAs, we showed that ROS activation entails cell surface MT1-MMP proteolytic activity. Induction of ROS in prostate cancer cells expressing MT1-MMP required adhesion to extracellular matrix proteins and was impeded by anti-β1 integrin antibodies. These results highlight a novel mechanism of malignant progression in prostate cancer cells that involves β1 integrin-mediated adhesion, in concert with MT1-MMP proteolytic activity, to elicit oxidative stress and induction of a more invasive phenotype.
Collapse
Affiliation(s)
- Hoang-Lan Nguyen
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | | | | | | | | | | |
Collapse
|
32
|
Zarrabi K, Dufour A, Li J, Kuscu C, Pulkoski-Gross A, Zhi J, Hu Y, Sampson NS, Zucker S, Cao J. Inhibition of matrix metalloproteinase 14 (MMP-14)-mediated cancer cell migration. J Biol Chem 2011; 286:33167-77. [PMID: 21795678 DOI: 10.1074/jbc.m111.256644] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Matrix metalloproteinases (MMPs) have been shown to be key players in both extracellular matrix remodeling and cell migration during cancer metastasis. MMP-14, a membrane-anchored MMP, in particular, is closely associated with these processes. The hemopexin (PEX) domain of MMP-14 has been proposed as the modulating region involved in the molecular cross-talk that initiates cell migration through homodimerization of MMP-14 as well as heterodimerization with the cell surface adhesion molecule CD44. In this study, minimal regions required for function within the PEX domain were investigated through a series of substitution mutations. Blades I and IV were found to be involved in cell migration. We found that blade IV is necessary for MMP-14 homodimerization and that blade I is required for CD44 MMP-14 heterodimerization. Cross-talk between MMP-14 and CD44 results in phosphorylation of EGF receptor and downstream activation of the MAPK and PI3K signaling pathways involved in cell migration. Based on these mutagenesis analyses, peptides mimicking the essential outermost strand motifs within the PEX domain of MMP-14 were designed. These synthetic peptides inhibit MMP-14-enhanced cell migration in a dose-dependent manner but have no effect on the function of other MMPs. Furthermore, these peptides interfere with cancer metastasis without affecting primary tumor growth. Thus, targeting the MMP-14 hemopexin domain represents a novel approach to inhibit MMP-14-mediated cancer dissemination.
Collapse
Affiliation(s)
- Kevin Zarrabi
- Department of Medicine/Cancer Prevention, Stony Brook University, Stony Brook, New York 11794, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Dufour A, Sampson NS, Li J, Kuscu C, Rizzo RC, Deleon JL, Zhi J, Jaber N, Liu E, Zucker S, Cao J. Small-molecule anticancer compounds selectively target the hemopexin domain of matrix metalloproteinase-9. Cancer Res 2011; 71:4977-88. [PMID: 21646471 DOI: 10.1158/0008-5472.can-10-4552] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Lack of target specificity by existing matrix metalloproteinase (MMP) inhibitors has hindered antimetastatic cancer drug discovery. Inhibitors that bind to noncatalytic sites of MMPs and disrupt protease signaling function have the potential to be more specific and selective. In this work, compounds that target the hemopexin (PEX) domain of MMP-9 were identified using an in silico docking approach and evaluated using biochemical and biological approaches. Two of the selected compounds interfere with MMP-9-mediated cancer cell migration and proliferation in cells expressing exogenous or endogenous MMP-9. Furthermore, these inhibitors do not modulate MMP-9 catalytic activity. The lead compound, N-[4-(difluoromethoxy)phenyl]-2-[(4-oxo-6-propyl-1H-pyrimidin-2-yl)sulfanyl]-acetamide, specifically binds to the PEX domain of MMP-9, but not other MMPs. This interaction between the compound and the PEX domain results in the abrogation of MMP-9 homodimerization and leads to blockage of a downstream signaling pathway required for MMP-9-mediated cell migration. In a tumor xenograft model, this pyrimidinone retarded MDA-MB-435 tumor growth and inhibited lung metastasis. Thus, we have shown for the first time that a novel small-molecule interacts specifically with the PEX domain of MMP-9 and inhibits tumor growth and metastasis by reducing cell migration and proliferation.
Collapse
Affiliation(s)
- Antoine Dufour
- Departments of Medicine, Chemistry, Molecular and Cellular Biology, and Applied Mathematics, and Bioinformatics Facility, Stony Brook University, Stony Brook, NY 11794, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Mechanisms of liver fibrosis are complex and varied. Among them, metabolic factors are particularly important in the development of fibrosis associated with nonalcoholic steatohepatitis (NASH). These factors are some of the "multiple parallel hits" responsible for liver damage during NASH. Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome. Major profibrogenic protagonists, such as hepatic stellate cells and Kupffer cells, are activated by insulin resistance, apoptosis and local inflammation. Relations between steatosis, insulin resistance and fibrosis are complex. Initially, simple steatosis may be a way to store deleterious free fatty acid in neutral triglycerides. If the lipid storage threshold is exceeded, steatosis may become associated with lipotoxicity. Similarly, interindividual variations of adipose tissue expandability might explain various phenotypes, ranging from "metabolically obese patients with normal weight" to "metabolically normal morbidly obese patients". The metabolic abnormalities in subcutaneous and visceral adipose tissue are insulin resistance and low-grade inflammation, which are associated with increased release of free fatty acid flux and changes in adipocytokines production such as leptin, adiponectin and interleukin 6. The nuclear transcription factor peroxisome proliferator-activated receptor gamma (PPARγ) and the endocannabinoid system might have important roles in liver fibrogenesis and are potential therapeutic targets. Finally, with the development of new molecular tools, gut microbiota has been recently identified for its pleiotropic functions, including metabolism regulation. Better knowledge of these mechanisms should lead to new strategies for the treatment of metabolic factors that play a key role in liver injuries.
Collapse
Affiliation(s)
- Rodolphe Anty
- Institut National de la Santé et de la Recherche Médicale, U895, Team 8, Hepatic Complications in Obesity, Nice F-06204, Cedex 3, France.
| | | |
Collapse
|
35
|
Sela-Passwell N, Trahtenherts A, Krüger A, Sagi I. New opportunities in drug design of metalloproteinase inhibitors: combination between structure-function experimental approaches and systems biology. Expert Opin Drug Discov 2011; 6:527-42. [PMID: 22646077 DOI: 10.1517/17460441.2011.560936] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION MMPs (matrix metalloproteinases) and ADAMs (a disintegrin and metalloproteinases) are endopeptidases central to the degradation and remodeling of the extracellular matrix. These proteases also exhibit regulatory activity in cell signaling pathways and thus tissue homeostasis under normal conditions and in many diseases. Consequently, individual members of the MMP and ADAM protein families were identified as important therapeutic targets. However, designing effective inhibitors in vivo for this class of enzymes appears to be extremely challenging. This is attributed to the broad structural similarity of their active sites and to the dynamic functional interconnectivity of MMPs with other proteases, their inhibitors, and substrates (the so-called degradome) in healthy and disease tissues. AREAS COVERED The article covers the progress in designing metalloproteinase inhibitors, based on recent advancements in our understanding of enzyme structures and their function as master regulators. It also discusses the potential of utilizing structure-based drug design strategies in conjunction with systems biology experimental approaches for designing potent and therapeutically effective metalloproteinase inhibitors. EXPERT OPINION We highlight the use of protein-based drug design strategies, for example, antibodies and protein scaffolds, targeting extracatalytic domains, which are central to proteolytic and non-proteolytic enzyme functions. Such rationally designed function-blocking inhibitors may create new opportunities in disease management and in emerging therapies that require control of dysregulated MMP activity without causing severe side effects. Importantly, the lessons learned from studying these protein-based inhibitors can be implemented to design new and effective small or medium sized synthetic antagonists.
Collapse
Affiliation(s)
- Netta Sela-Passwell
- The Weizmann Institute of Science, Department of Biological Regulation , Rehovot 76100 , Israel
| | | | | | | |
Collapse
|
36
|
Bialek J, Kunanuvat U, Hombach-Klonisch S, Spens A, Stetefeld J, Sunley K, Lippert D, Wilkins JA, Hoang-Vu C, Klonisch T. Relaxin Enhances the Collagenolytic Activity and In Vitro Invasiveness by Upregulating Matrix Metalloproteinases in Human Thyroid Carcinoma Cells. Mol Cancer Res 2011; 9:673-87. [DOI: 10.1158/1541-7786.mcr-10-0411] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
37
|
Tochowicz A, Goettig P, Evans R, Visse R, Shitomi Y, Palmisano R, Ito N, Richter K, Maskos K, Franke D, Svergun D, Nagase H, Bode W, Itoh Y. The dimer interface of the membrane type 1 matrix metalloproteinase hemopexin domain: crystal structure and biological functions. J Biol Chem 2011; 286:7587-600. [PMID: 21193411 PMCID: PMC3045013 DOI: 10.1074/jbc.m110.178434] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 12/03/2010] [Indexed: 11/06/2022] Open
Abstract
Homodimerization is an essential step for membrane type 1 matrix metalloproteinase (MT1-MMP) to activate proMMP-2 and to degrade collagen on the cell surface. To uncover the molecular basis of the hemopexin (Hpx) domain-driven dimerization of MT1-MMP, a crystal structure of the Hpx domain was solved at 1.7 Å resolution. Two interactions were identified as potential biological dimer interfaces in the crystal structure, and mutagenesis studies revealed that the biological dimer possesses a symmetrical interaction where blades II and III of molecule A interact with blades III and II of molecule B. The mutations of amino acids involved in the interaction weakened the dimer interaction of Hpx domains in solution, and incorporation of these mutations into the full-length enzyme significantly inhibited dimer-dependent functions on the cell surface, including proMMP-2 activation, collagen degradation, and invasion into the three-dimensional collagen matrix, whereas dimer-independent functions, including gelatin film degradation and two-dimensional cell migration, were not affected. These results shed light on the structural basis of MT1-MMP dimerization that is crucial to promote cellular invasion.
Collapse
Affiliation(s)
- Anna Tochowicz
- From the Arbeitsgruppe Proteinaseforschung, Max-Planck-Institut fuer Biochemie, Am Klopferspitz 18, D-82152 Martinsried, Germany
| | - Peter Goettig
- From the Arbeitsgruppe Proteinaseforschung, Max-Planck-Institut fuer Biochemie, Am Klopferspitz 18, D-82152 Martinsried, Germany
| | - Richard Evans
- the Department of Matrix Biology, Kennedy Institute of Rheumatology Division, Faculty of Medicine, Imperial College London, 65 Aspenlea Rd., London W6 8LH, United Kingdom
| | - Robert Visse
- the Department of Matrix Biology, Kennedy Institute of Rheumatology Division, Faculty of Medicine, Imperial College London, 65 Aspenlea Rd., London W6 8LH, United Kingdom
| | - Yasuyuki Shitomi
- the Department of Matrix Biology, Kennedy Institute of Rheumatology Division, Faculty of Medicine, Imperial College London, 65 Aspenlea Rd., London W6 8LH, United Kingdom
| | - Ralf Palmisano
- the Department of Matrix Biology, Kennedy Institute of Rheumatology Division, Faculty of Medicine, Imperial College London, 65 Aspenlea Rd., London W6 8LH, United Kingdom
| | - Noriko Ito
- the Department of Matrix Biology, Kennedy Institute of Rheumatology Division, Faculty of Medicine, Imperial College London, 65 Aspenlea Rd., London W6 8LH, United Kingdom
| | - Klaus Richter
- the Institut für Organische Chemie und Biochemie, Technische Universität München, Lichtenbergstrasse 4, 85747 München, Germany, and
| | - Klaus Maskos
- From the Arbeitsgruppe Proteinaseforschung, Max-Planck-Institut fuer Biochemie, Am Klopferspitz 18, D-82152 Martinsried, Germany
| | - Daniel Franke
- the European Molecular Biology Laboratory, Hamburg Outstation, Deutsches Elektronen Synchrotron, Notkestrasse 85, D-22603 Hamburg, Germany
| | - Dmitri Svergun
- the European Molecular Biology Laboratory, Hamburg Outstation, Deutsches Elektronen Synchrotron, Notkestrasse 85, D-22603 Hamburg, Germany
| | - Hideaki Nagase
- the Department of Matrix Biology, Kennedy Institute of Rheumatology Division, Faculty of Medicine, Imperial College London, 65 Aspenlea Rd., London W6 8LH, United Kingdom
| | - Wolfram Bode
- From the Arbeitsgruppe Proteinaseforschung, Max-Planck-Institut fuer Biochemie, Am Klopferspitz 18, D-82152 Martinsried, Germany
| | - Yoshifumi Itoh
- the Department of Matrix Biology, Kennedy Institute of Rheumatology Division, Faculty of Medicine, Imperial College London, 65 Aspenlea Rd., London W6 8LH, United Kingdom
| |
Collapse
|
38
|
Furin Functions as a Nonproteolytic Chaperone for Matrix Metalloproteinase-28: MMP-28 Propeptide Sequence Requirement. Biochem Res Int 2010; 2011:630319. [PMID: 21152186 PMCID: PMC2989691 DOI: 10.1155/2011/630319] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2010] [Revised: 09/16/2010] [Accepted: 09/27/2010] [Indexed: 01/16/2023] Open
Abstract
Although MMP-28 is involved in numerous important physiologic and pathologic conditions, the mechanisms of action of this secreted proteinase is not well understood. We now have demonstrated that furin serves as an intermolecular chaperone for MMP-28 secretion by interacting with the propeptide domain of MMP-28. Employing COS-1 cells transfected with MMP-28 cDNA, protein levels of MMP-28 were quite low in conditioned media as compared to cell lysates. Coexpression of MMP-28 with furin cDNA resulted in markedly enhanced MMP-28 secretion. Contrary to expectation, cleavage of MMP-28 at the furin consensus sequence did not occur and proteolytic inactive furin was equally effective in enhancing MMP-28 secretion. Furin and MMP-28 coimmunoprecipitated and were partially coimmunolocalized in the cytoplasm of transfected cells. Cotransfection with furin cDNA also enhanced MMP-28 induced cell migration. In conclusion, our data provide a novel mechanism for MMP-28 function in cells in which furin serves as an intermolecular chaperone.
Collapse
|
39
|
Dufour A, Zucker S, Sampson NS, Kuscu C, Cao J. Role of matrix metalloproteinase-9 dimers in cell migration: design of inhibitory peptides. J Biol Chem 2010; 285:35944-56. [PMID: 20837483 DOI: 10.1074/jbc.m109.091769] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Non-proteolytic activities of matrix metalloproteinases (MMPs) have recently been shown to impact cell migration, but the precise mechanism remains to be understood. We previously demonstrated that the hemopexin (PEX) domain of MMP-9 is a prerequisite for enhanced cell migration. Using a biochemical approach, we now report that dimerization of MMP-9 through the PEX domain appears necessary for MMP-9-enhanced cell migration. Following a series of substitution mutations within the MMP-9 PEX domain, blade IV was shown to be critical for homodimerization, whereas blade I was required for heterodimerization with CD44. Blade I and IV mutants showed diminished enhancement of cell migration compared with wild type MMP-9-transfected cells. Peptides mimicking motifs in the outermost strands of the first and fourth blades of the MMP-9 PEX domain were designed. These peptides efficiently blocked MMP-9 dimer formation and inhibited motility of COS-1 cells overexpressing MMP-9, HT-1080, and MDA-MB-435 cells. Using a shRNA approach, CD44 was found to be a critical molecule in MMP-9-mediated cell migration. Furthermore, an axis involving a MMP-9-CD44-EGFR signaling pathway in cell migration was identified using antibody array and specific receptor tyrosine kinase inhibitors. In conclusion, we dissected the mechanism of pro-MMP-9-enhanced cell migration and developed structure-based inhibitory peptides targeting MMP-9-mediated cell migration.
Collapse
Affiliation(s)
- Antoine Dufour
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794, USA
| | | | | | | | | |
Collapse
|
40
|
Wickramasinghe RD, Ko Ferrigno P, Roghi C. Peptide aptamers as new tools to modulate clathrin-mediated internalisation--inhibition of MT1-MMP internalisation. BMC Cell Biol 2010; 11:58. [PMID: 20653933 PMCID: PMC2919464 DOI: 10.1186/1471-2121-11-58] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Accepted: 07/23/2010] [Indexed: 12/23/2022] Open
Abstract
Background Peptide aptamers are combinatorial protein reagents that bind to targets with a high specificity and a strong affinity thus providing a molecular tool kit for modulating the function of their targets in vivo. Results Here we report the isolation of a peptide aptamer named swiggle that interacts with the very short (21 amino acid long) intracellular domain of membrane type 1-metalloproteinase (MT1-MMP), a key cell surface protease involved in numerous and crucial physiological and pathological cellular events. Expression of swiggle in mammalian cells was found to increase the cell surface expression of MT1-MMP by impairing its internalisation. Swiggle interacts with the LLY573 internalisation motif of MT1-MMP intracellular domain, thus disrupting the interaction with the μ2 subunit of the AP-2 internalisation complex required for endocytosis of the protease. Interestingly, swiggle-mediated inhibition of MT1-MMP clathrin-mediated internalisation was also found to promote MT1-MMP-mediated cell migration. Conclusions Taken together, our results provide further evidence that peptide aptamers can be used to dissect molecular events mediated by individual protein domains, in contrast to the pleiotropic effects of RNA interference techniques.
Collapse
Affiliation(s)
- Rochana D Wickramasinghe
- Department of Oncology, Cambridge Research Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | | | | |
Collapse
|
41
|
Zhang L, Yang M, Yang D, Cavey G, Davidson P, Gibson G. Molecular interactions of MMP-13 C-terminal domain with chondrocyte proteins. Connect Tissue Res 2010; 51:230-9. [PMID: 20073988 DOI: 10.3109/03008200903288902] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Matrix metalloproteinases (MMP)-13 activity is necessary for normal skeletal development and plays a central role in cartilage degeneration associated with osteoarthritis (OA). The studies we described here examine the interactions of the hemopexin domain of MMP-13 with proteins secreted by human chondrocytes in culture. The hemopexin domain of the MMPs and many other proteins in which this structure is found mediates protein function by forming the primary site of interaction with other proteins. We have modified a tandem affinity expression tag (hTAP) to enable efficient expression of the tagged bait protein. In this case the MMP-13 C-terminal domain (CTD) comprises hinge and hemopexin domain, and we immobilized the fusion construct on a column of agarose bound immunoglobin G. The MMP-13 CTD affinity column so generated enabled the efficient and gentle isolation of interacting proteins from the culture medium of human articular chondrocytes. TIMP1 and alpha2-macroglobulin previously shown to interact with MMP-13 as well as several proteins, fibronectin, type VI collagen and xylosyltransferase 1 and several proteoglycans, decorin, syndecan 4 and serglycin not previously recognized as interacting with MMP-13 were identified by mass spectrometry. The interaction between isolated proteins and MMP-13 CTD was verified by yeast two hybrid analysis. We also demonstrated serglycin expression by chondrocytes for the first time and its co localization with MMP-13 in a cytoplasmic granular morphology. The consequence of these interactions remains to be demonstrated, however; binding to MMP-13 suggests a role in the regulation of cartilage degradation.
Collapse
Affiliation(s)
- Liang Zhang
- Bone and Joint Center, Henry Ford Hospital, Detroit, Michigan, USA
| | | | | | | | | | | |
Collapse
|
42
|
Poincloux R, Lizárraga F, Chavrier P. Matrix invasion by tumour cells: a focus on MT1-MMP trafficking to invadopodia. J Cell Sci 2009; 122:3015-24. [PMID: 19692588 DOI: 10.1242/jcs.034561] [Citation(s) in RCA: 361] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
When migrating away from a primary tumour, cancer cells interact with and remodel the extracellular matrix (ECM). Matrix metalloproteinases (MMPs), and in particular the transmembrane MT1-MMP (also known as MMP-14), are key enzymes in tumour-cell invasion. Results from recent in vitro studies highlight that MT1-MMP is implicated both in the breaching of basement membranes by tumour cells and in cell invasion through interstitial type-I collagen tissues. Remarkably, MT1-MMP accumulates at invadopodia, which are specialized ECM-degrading membrane protrusions of invasive cells. Here we review current knowledge about MT1-MMP trafficking and its importance for the regulation of protease activity at invadopodia. In invasive cells, endocytosis of MT1-MMP by clathrin- and caveolae-dependent pathways can be counteracted by several mechanisms, which leads to protease stabilization at the cell surface and increased pericellular degradation of the matrix. Furthermore, the recent identification of cellular components that control delivery of MT1-MMP to invadopodia brings new insight into mechanisms of cancer-cell invasion and reveals potential pharmacological targets.
Collapse
Affiliation(s)
- Renaud Poincloux
- CNRS, UMR144, Membrane and Cytoskeleton Dynamics, and Institut Curie, Paris, France
| | | | | |
Collapse
|
43
|
Induction of a MT1-MMP and MT2-MMP-dependent basement membrane transmigration program in cancer cells by Snail1. Proc Natl Acad Sci U S A 2009; 106:20318-23. [PMID: 19915148 DOI: 10.1073/pnas.0910962106] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The ability of carcinoma cells arising at primary sites to cross their underlying basement membrane (BM), a specialized form of extracellular matrix that subtends all epithelial cells, and to access the host vasculature are central features of the malignant phenotype. The initiation of the invasive phenotype has been linked to the aberrant expression of zinc-finger transcriptional repressors, like Snail1, which act by triggering an epithelial-mesenchymal cell-like transformation (EMT-like) via the regulation of largely undefined, downstream effectors. Herein, we find that Snail1 induces cancer cells to (i) degrade and perforate BM barriers, (ii) initiate angiogenesis, and (iii) and intravasate vascular networks in vivo via a matrix metalloproteinase (MMP)-dependent process. Unexpectedly, the complete Snail1 invasion program can be recapitulated by expressing directly either of the membrane-anchored metalloproteinases, MT1-MMP or MT2-MMP. The pro-invasive, angiogenic, and metastatic activities of MT1-MMP and MT2-MMP are unique relative to all other metalloproteinase family members and cannot be mimicked in vivo by the secreted MMPs, MMP-1, MMP-2, MMP-3, MMP-7, MMP-9, or MMP-13. Further, siRNA-specific silencing of MT1-MMP and MT2-MMP ablates completely the ability of Snail1 to drive cancer cell BM invasion, induce angiogenesis, or trigger intravasation. Taken together, these data demonstrate that MT1-MMP and MT2-MMP cooperatively function as direct-acting, pro-invasive factors that confer Snail1-triggered cells with the key activities most frequently linked to morbidity and mortality in cancer.
Collapse
|
44
|
Abstract
Human mesenchymal stem cells (hMSCs) localized to bone marrow, nonhematopoietic organs, as well as perivascular niches are postulated to traffic through type I collagen-rich stromal tissues to first infiltrate sites of tissue damage, inflammation, or neoplasia and then differentiate. Nevertheless, the molecular mechanisms supporting the ability of hMSCs to remodel 3-dimensional (3D) collagenous barriers during trafficking or differentiation remain undefined. Herein, we demonstrate that hMSCs degrade and penetrate type I collagen networks in tandem with the expression of a 5-member set of collagenolytic matrix metalloproteinases (MMPs). Specific silencing of each of these proteases reveals that only a single membrane-tethered metalloenzyme, termed MT1-MMP, plays a required role in hMSC-mediated collagenolysis, 3D invasion, and intravasation. Further, once confined within type I collagen-rich tissue, MT1-MMP also controls hMSC differentiation in a 3D-specific fashion. Together, these data demonstrate that hMSC invasion and differentiation programs fall under the control of the pericellular collagenase, MT1-MMP.
Collapse
|
45
|
Mori H, Gjorevski N, Inman JL, Bissell MJ, Nelson CM. Self-organization of engineered epithelial tubules by differential cellular motility. Proc Natl Acad Sci U S A 2009; 106:14890-5. [PMID: 19706461 PMCID: PMC2736456 DOI: 10.1073/pnas.0901269106] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Indexed: 02/06/2023] Open
Abstract
Patterning of developing tissues arises from a number of mechanisms, including cell shape change, cell proliferation, and cell sorting from differential cohesion or tension. Here, we reveal that differences in cell motility can also lead to cell sorting within tissues. Using mosaic engineered mammary epithelial tubules, we found that cells sorted depending on their expression level of the membrane-anchored collagenase matrix metalloproteinase (MMP)-14. These rearrangements were independent of the catalytic activity of MMP14 but absolutely required the hemopexin domain. We describe a signaling cascade downstream of MMP14 through Rho kinase that allows cells to sort within the model tissues. Cell speed and persistence time were enhanced by MMP14 expression, but only the latter motility parameter was required for sorting. These results indicate that differential directional persistence can give rise to patterns within model developing tissues.
Collapse
Affiliation(s)
- Hidetoshi Mori
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and
| | - Nikolce Gjorevski
- Departments of Chemical Engineering and Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Jamie L. Inman
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and
| | - Mina J. Bissell
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and
| | - Celeste M. Nelson
- Departments of Chemical Engineering and Molecular Biology, Princeton University, Princeton, NJ 08544
| |
Collapse
|
46
|
Deshmukh HS, McLachlan A, Atkinson JJ, Hardie WD, Korfhagen TR, Dietsch M, Liu Y, Di PYP, Wesselkamper SC, Borchers MT, Leikauf GD. Matrix metalloproteinase-14 mediates a phenotypic shift in the airways to increase mucin production. Am J Respir Crit Care Med 2009; 180:834-45. [PMID: 19661247 DOI: 10.1164/rccm.200903-0328oc] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Induced mainly by cigarette smoking, chronic obstructive pulmonary disease (COPD) is a global public health problem characterized by progressive difficulty in breathing and increased mucin production. Previously, we reported that acrolein levels found in COPD sputum could activate matrix metalloproteinase-9 (MMP9). OBJECTIVES To determine whether acrolein increases expression and activity of MMP14, a critical membrane-bound endopeptidase that can initial a MMP-activation cascade. METHODS MMP14 activity and adduct formation were measured following direct acrolein treatment. MMP14 expression and activity was measured in human airway epithelial cells. MMP14 immunohistochemistry was performed with COPD tissue, and in acrolein- or tobacco-exposed mice. MEASUREMENTS AND MAIN RESULTS In a cell-free system, acrolein, in concentrations equal to those found in COPD sputum, directly adducted cysteine 319 in the MMP14 hemopexin-like domain and activated MMP14. In cells, acrolein increased MMP14 activity, which was inhibited by a proprotein convertase inhibitor, hexa-d-arginine. In the airway epithelium of COPD subjects, immunoreactive MMP14 protein increased. In mouse lung, acrolein or tobacco smoke increased lung MMP14 activity and protein. In cells, acrolein-induced MMP14 transcripts were inhibited by an epidermal growth factor receptor (EGFR) neutralizing antibody, EGFR kinase inhibitor, metalloproteinase inhibitor, or mitogen-activated protein kinase (MAPK) 3/2 or MAPK8 inhibitors, but not a MAPK14 inhibitor. Decreasing the MMP14 protein and activity in vitro by small interfering (si)RNA to MMP14 diminished the acrolein-induced MUC5AC transcripts. In acrolein-exposed mice or transgenic mice with lung-specific transforming growth factor-alpha (an EGFR ligand) expression, lung MMP14 and MUC5AC levels increased and these effects were inhibited by a EGFR inhibitor, erlotinib. CONCLUSIONS Taken together, these findings implicate acrolein-induced MMP14 expression and activity in mucin production in COPD.
Collapse
Affiliation(s)
- Hitesh S Deshmukh
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sabeh F, Li XY, Saunders TL, Rowe RG, Weiss SJ. Secreted versus membrane-anchored collagenases: relative roles in fibroblast-dependent collagenolysis and invasion. J Biol Chem 2009; 284:23001-11. [PMID: 19542530 DOI: 10.1074/jbc.m109.002808] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Fibroblasts degrade type I collagen, the major extracellular protein found in mammals, during events ranging from bulk tissue resorption to invasion through the three-dimensional extracellular matrix. Current evidence suggests that type I collagenolysis is mediated by secreted as well as membrane-anchored members of the matrix metalloproteinase (MMP) gene family. However, the roles played by these multiple and possibly redundant, degradative systems during fibroblast-mediated matrix remodeling is undefined. Herein, we use fibroblasts isolated from Mmp13(-/-), Mmp8(-/-), Mmp2(-/-), Mmp9(-/-), Mmp14(-/-) and Mmp16(-/-) mice to define the functional roles for secreted and membrane-anchored collagenases during collagen-resorptive versus collagen-invasive events. In the presence of a functional plasminogen activator-plasminogen axis, secreted collagenases arm cells with a redundant collagenolytic potential that allows fibroblasts harboring single deficiencies for either MMP-13, MMP-8, MMP-2, or MMP-9 to continue to degrade collagen comparably to wild-type fibroblasts. Likewise, Mmp14(-/-) or Mmp16(-/-) fibroblasts retain near-normal collagenolytic activity in the presence of plasminogen via the mobilization of secreted collagenases, but only Mmp14 (MT1-MMP) plays a required role in the collagenolytic processes that support fibroblast invasive activity. Furthermore, by artificially tethering a secreted collagenase to the surface of Mmp14(-/-) fibroblasts, we demonstrate that localized pericellular collagenolytic activity differentiates the collagen-invasive phenotype from bulk collagen degradation. Hence, whereas secreted collagenases arm fibroblasts with potent matrix-resorptive activity, only MT1-MMP confers the focal collagenolytic activity necessary for supporting the tissue-invasive phenotype.
Collapse
Affiliation(s)
- Farideh Sabeh
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | |
Collapse
|
48
|
Aplin AC, Zhu WH, Fogel E, Nicosia RF. Vascular regression and survival are differentially regulated by MT1-MMP and TIMPs in the aortic ring model of angiogenesis. Am J Physiol Cell Physiol 2009; 297:C471-80. [PMID: 19494241 DOI: 10.1152/ajpcell.00019.2009] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This study was designed to investigate the role of matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs (TIMPs) in the reabsorption of neovessels in collagen gel cultures of rat and mouse aortic rings. Aortic angiogenesis was associated with collagen lysis and production of the matrix-degrading enzymes MMP-2, MMP-9, and membrane-type MMP (MT1-MMP, or MMP-14). Vascular growth and regression were not affected by disruption of MMP-2 or MMP-9. In addition, no effect on vascular regression was observed by blocking plasmin, a protease implicated in the activation of MMPs, with epsilon-aminocaproic acid or by adding plasminogen, which caused a modest increase in vascular proliferation. Conversely, angiogenesis was blocked and vessels stabilized by inhibiting MT1-MMP with neutralizing antibodies, TIMP-2, TIMP-3, or TIMP-4. TIMP-1, which blocks MMP-2 and MMP-9 but is a poor inhibitor of MT1-MMP, had no antiangiogenic effect. However, TIMP-1 prolonged the survival of neovessels following angiogenesis. Vascular regression was accelerated in aortic cultures from TIMP-1- and TIMP-2-deficient mice. The vascular survival effect of anti-MT1-MMP antibodies and TIMPs with MT1-MMP inhibitory activity was associated with complete inhibition of collagen lysis. In contrast, TIMP-1 had no anticollagenolytic effect. These results indicate that MT1-MMP plays a critical role not only in angiogenesis but also in vascular regression and demonstrate that TIMPs with anti-MT1-MMP activity have opposite effects on angiogenic outcomes depending on the stage of the angiogenic process. This study also suggests the existence of a TIMP-1-mediated alternate pathway of vascular survival that is unrelated to MT1-MMP inhibitory activity.
Collapse
Affiliation(s)
- A C Aplin
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|
49
|
Sela-Passwell N, Rosenblum G, Shoham T, Sagi I. Structural and functional bases for allosteric control of MMP activities: can it pave the path for selective inhibition? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1803:29-38. [PMID: 19406173 DOI: 10.1016/j.bbamcr.2009.04.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 04/20/2009] [Accepted: 04/21/2009] [Indexed: 01/01/2023]
Abstract
The zinc-dependent matrix metalloproteinases (MMPs) belong to a large family of structurally homologous enzymes. These enzymes are involved in a wide variety of biological processes ranging from physiological cell proliferation and differentiation to pathological states associated with tumor metastasis, inflammation, tissue degeneration, and cell death. Controlling the enzymatic activity of specific individual MMPs by antagonist molecules is highly desirable, first, for studying their individual roles, and second as potential therapeutic agents. However, blocking the enzymatic activity with synthetic small inhibitors appears to be an extremely difficult task. Thus, this is an unmet need presumably due to the high structural homology between MMP catalytic domains. Recent reports have recognized a potential role for exosite or allosteric protein regions, distinct from the extended catalytic pocket, in mediating MMP activation and substrate hydrolysis. This raises the possibility that MMP enzymatic and non-enzymatic activities may be modified via antagonist molecules targeted to such allosteric sites or to alternative enzyme domains. In this review, we discuss the structural and functional bases for potential allosteric control of MMPs and highlight potential alternative enzyme domains as targets for designing highly selective MMP inhibitors.
Collapse
|
50
|
Abstract
Membrane-type 1 matrix metalloproteinase (MT1-MMP) is a zinc-dependent proteinase found in cholesterol-rich lipid rafts on the plasma membrane. MT1-MMP hydrolyzes extracellular matrix (ECM) proteins, activates pro-matrix metalloproteinase-2 (proMMP-2) and plays an important role in ECM remodeling, cancer cell migration and metastasis. The role of caveolin-1, an integral protein of caveolae, in the activation of MT1-MMP remains largely unknown. Here, we show that the expression of caveolin-1 attenuates the activation of proMMP-2, reduces proteolytic cleavage of ECM and inhibits cell migration. We utilized the cytoplasmic tail domain deletion (DeltaCT) or the E240A mutant of MT1-MMP. Co-expression of caveolin-1 with the wild-type or the DeltaCT MT1-MMP decreased the proMMP-2 activation and inhibited collagen degradation and cell migration. Caveolin-1 had no effect on the catalytically inert E240A MT1-MMP. Our findings suggest that caveolin-1 is essential in the down-regulation of MT1-MMP activity by promoting internalization from the cell surface.
Collapse
Affiliation(s)
- Hye-Nan Kim
- Department of Biotechnology, Hannam University, Daejeon 305-811, Korea
| | | |
Collapse
|