Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
2.
Clin Cancer Res ; 20(2): 344-57, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24150233

ABSTRACT

PURPOSE: This study investigated the functional and clinical significance of integrin αvß6 upregulation in myoepithelial cells of ductal carcinoma in situ (DCIS). EXPERIMENTAL DESIGN: Archival samples of DCIS and DCIS with associated invasion (n = 532) were analyzed for expression of αvß6 by immunohistochemistry and ability to predict recurrence and progression assessed in an independent, unique cohort of DCIS cases with long-term follow-up. Primary myoepithelial cells and myoepithelial cell lines, with and without αvß6 expression, were used to measure the effect of αvß6 on growth and invasion of tumor cell lines in vitro and in a xenograft mouse model. Involvement of TGFß signaling was established using mink lung epithelial cell (MLEC) assay and antibody inhibition, and expression and activation of matrix metalloproteinase (MMP)-9 established by Real Time-PCR and zymography. RESULTS: Expression of αvß6 is significantly associated with progression to invasive cancer (P < 0.006) and with recurrence over a median follow-up of 114 months in a series of matched DCIS cases treated with local excision. We show that expression of αvß6 drives myoepithelial cells to promote tumor cell invasion in vitro and enhances mammary tumor growth in vivo. The tumor-promoting effect of αvß6-positive myoepithelial cells is dependent on TGFß-driven upregulation of MMP9 and can be abrogated by inhibiting this pathway. CONCLUSION: These findings indicate that altered myoepithelial cells in DCIS predict disease progression and recurrence and show that upregulation of αvß6 on myoepithelial cells generates a tumor promoter function through TGFß upregulation of MMP-9. These data suggest that expression of αvß6 may be used to stratify patients with DCIS.


Subject(s)
Antigens, Neoplasm/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Integrins/genetics , Tumor Microenvironment/genetics , Animals , Antigens, Neoplasm/metabolism , Case-Control Studies , Cell Line , Cell Line, Tumor , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Follow-Up Studies , Humans , Immunohistochemistry , Integrins/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mink , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Prognosis , Transforming Growth Factor beta/metabolism , Tumor Burden/genetics
3.
J Biol Chem ; 288(23): 16282-16294, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23632023

ABSTRACT

Matrix metalloproteinase 8 (MMP-8) is a tumor-suppressive protease that cleaves numerous substrates, including matrix proteins and chemokines. In particular, MMP-8 proteolytically activates IL-8 and, thereby, regulates neutrophil chemotaxis in vivo. We explored the effects of expression of either a WT or catalytically inactive (E198A) mutant version of MMP-8 in human breast cancer cell lines. Analysis of serum-free conditioned media from three breast cancer cell lines (MCF-7, SK-BR-3, and MDA-MB-231) expressing WT MMP-8 revealed elevated levels of IL-6 and IL-8. This increase was mirrored at the mRNA level and was dependent on MMP-8 catalytic activity. However, sustained expression of WT MMP-8 by breast cancer cells was non-permissive for long-term growth, as shown by reduced colony formation compared with cells expressing either control vector or E198A mutant MMP-8. In long-term culture of transfected MDA-MB-231 cells, expression of WT but not E198A mutant MMP-8 was lost, with IL-6 and IL-8 levels returning to base line. Rare clonal isolates of MDA-MB-231 cells expressing WT MMP-8 were generated, and these showed constitutively high levels of IL-6 and IL-8, although production of the interleukins was no longer dependent upon MMP-8 activity. These studies support a causal connection between MMP-8 activity and the IL-6/IL-8 network, with an acute response to MMP-8 involving induction of the proinflammatory mediators, which may in part serve to compensate for the deleterious effects of MMP-8 on breast cancer cell growth. This axis may be relevant to the recognized ability of MMP-8 to orchestrate the innate immune system in inflammation in vivo.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Matrix Metalloproteinase 8/biosynthesis , Neoplasm Proteins/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Inflammation Mediators/metabolism , Interleukin-6/genetics , Interleukin-8/genetics , Matrix Metalloproteinase 8/genetics , Neoplasm Proteins/genetics
4.
J Cell Sci ; 123(Pt 21): 3808-16, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20940254

ABSTRACT

Blood vascular cells and lymphatic endothelial cells (BECs and LECs, respectively) form two separate vascular systems and are functionally distinct cell types or lineages with characteristic gene expression profiles. Interconversion between these cell types has not been reported. Here, we show that in conventional in vitro angiogenesis assays, human BECs of fetal or adult origin show altered gene expression that is indicative of transition to a lymphatic-like phenotype. This change occurs in BECs undergoing tubulogenesis in fibrin, collagen or Matrigel assays, but is independent of tube formation per se, because it is not inhibited by a metalloproteinase inhibitor that blocks tubulogenesis. It is also reversible, since cells removed from 3D tubules revert to a BEC expression profile upon monolayer culture. Induction of the lymphatic-like phenotype is partially inhibited by co-culture of HUVECs with perivascular cells. These data reveal an unexpected plasticity in endothelial phenotype, which is regulated by contact with the ECM environment and/or cues from supporting cells.


Subject(s)
Cell Transdifferentiation , Endothelium, Vascular/metabolism , Lymphatic Vessels/metabolism , Lymphoid Progenitor Cells/metabolism , Microtubules/metabolism , Adult , Cell Differentiation , Cell Lineage , Cells, Cultured , Coculture Techniques , Collagen/metabolism , Drug Combinations , Endothelium, Vascular/pathology , Extracellular Matrix , Fibrin/metabolism , Humans , Laminin/metabolism , Lymphatic Vessels/pathology , Lymphoid Progenitor Cells/pathology , Neovascularization, Physiologic , Phenotype , Proteoglycans/metabolism , Tissue Engineering
5.
Cancer Res ; 70(17): 6902-12, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20663911

ABSTRACT

Vascular disrupting agents (VDA) offer a strategy to starve solid tumors of nutrients and oxygen concomitant with tumor shrinkage. Several VDAs have progressed into early clinical trials, but their therapeutic value seems to be compromised by systemic toxicity. In this report, we describe the design and characterization of a novel VDA, ICT2588, that is nontoxic until activated specifically in the tumor by membrane-type 1 matrix metalloproteinase (MT1-MMP). HT1080 cancer cells expressing MT1-MMP were selectively chemosensitive to ICT2588, whereas MCF7 cells that did not express MT1-MMP were nonresponsive. Preferential hydrolysis of ICT2588 to its active metabolite (ICT2552) was observed in tumor homogenates of HT1080 relative to MCF7 homogenates, mouse plasma, and liver homogenate. ICT2588 activation was inhibited by the MMP inhibitor ilomastat. In HT1080 tumor-bearing mice, ICT2588 administration resulted in the formation of the active metabolite, diminution of tumor vasculature, and hemorrhagic necrosis of the tumor. The antitumor activity of ICT2588 was superior to its active metabolite, exhibiting reduced toxicity, improved therapeutic index, enhanced pharmacodynamic effect, and greater efficacy. Coadministration of ICT2588 with doxorubicin resulted in a significant antitumor response (22.6 d growth delay), which was superior to the administration of ICT2588 or doxorubicin as a single agent, including complete tumor regressions. Our findings support the clinical development of ICT2588, which achieves selective VDA targeting based on MT-MMP activation in the tumor microenvironment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Colchicine/analogs & derivatives , Fibrosarcoma/drug therapy , Matrix Metalloproteinases, Membrane-Associated/metabolism , Matrix Metalloproteinases/metabolism , Oligopeptides/pharmacology , Thiourea/analogs & derivatives , Angiogenesis Inhibitors/pharmacokinetics , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/enzymology , Colchicine/pharmacokinetics , Colchicine/pharmacology , Doxorubicin/pharmacology , Female , Fibrosarcoma/blood supply , Fibrosarcoma/enzymology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Oligopeptides/pharmacokinetics , Thiourea/pharmacokinetics , Thiourea/pharmacology , Tissue Distribution , Xenograft Model Antitumor Assays
6.
J Immunol ; 184(11): 6492-503, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20483790

ABSTRACT

Inflammatory tissue destruction is central to pathology in CNS tuberculosis (TB). We hypothesized that microglial-derived matrix metalloproteinases (MMPs) have a key role in driving such damage. Analysis of all of the MMPs demonstrated that conditioned medium from Mycobacterium tuberculosis-infected human monocytes (CoMTb) stimulated greater MMP-1, -3, and -9 gene expression in human microglial cells than direct infection. In patients with CNS TB, MMP-1/-3 immunoreactivity was demonstrated in the center of brain granulomas. Concurrently, CoMTb decreased expression of the inhibitors, tissue inhibitor of metalloproteinase-2, -3, and -4. MMP-1/-3 secretion was significantly inhibited by dexamethasone, which reduces mortality in CNS TB. Surface-enhanced laser desorption ionization time-of-flight analysis of CoMTb showed that TNF-alpha and IL-1beta are necessary but not sufficient for upregulating MMP-1 secretion and act synergistically to drive MMP-3 secretion. Chemical inhibition and promoter-reporter analyses showed that NF-kappaB and AP-1 c-Jun/FosB heterodimers regulate CoMTb-induced MMP-1/-3 secretion. Furthermore, NF-kappaB p65 and AP-1 c-Jun subunits were upregulated in biopsy granulomas from patients with cerebral TB. In summary, functionally unopposed, network-dependent microglial MMP-1/-3 gene expression and secretion regulated by NF-kappaB and AP-1 subunits were demonstrated in vitro and, for the first time, in CNS TB patients. Dexamethasone suppression of MMP-1/-3 gene expression provides a novel mechanism explaining the benefit of steroid therapy in these patients.


Subject(s)
Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/metabolism , Microglia/metabolism , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Tuberculosis, Central Nervous System/metabolism , Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Expression , Gene Expression Profiling , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/immunology , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/immunology , Microglia/immunology , Microscopy, Confocal , Monocytes/immunology , Monocytes/metabolism , Mycobacterium tuberculosis/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , Tuberculosis, Central Nervous System/genetics , Tuberculosis, Central Nervous System/immunology , Up-Regulation
7.
Clin Cancer Res ; 16(7): 2022-35, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20305301

ABSTRACT

PURPOSE: Head and neck squamous cell carcinomas (HNSCC) are characterized by high morbidity and mortality, largely due to the high invasive and metastatic potential of these tumors, high recurrence rates, and low treatment responses. Proteinases have been implicated in several aspects of tumor growth and metastasis in a broad range of tumors including HNSCC. EXPERIMENTAL DESIGN: Comprehensive expression profiling of proteinases [matrix metalloproteinases (MMPs), A disintegrin and metalloproteinase (ADAMs), and ADAMs with thrombospondin motif (ADAMTSs)] and their inhibitors [tissue inhibitor of metalloproteinases (TIMPs)] was done using quantitative real-time reverse transcription-PCR analysis of a large cohort of tissue samples representing the tumor (n = 83), the invasive margin (n = 41), and the adjacent tissue (n = 41) from 83 HNSCC patients, along with normal tissue controls (n = 13), as well as cell lines established from tumors of 34 HNSCC patients. RESULTS: The results show specifically elevated gene expression of several proteinases, including MMP1, MMP3, MMP10, and MMP13 within tumor tissue and peritumoral adjacent tissue. In addition, the results identify several novel HNSCC-associated proteinases, including ADAM8, ADAM9, ADAM17, ADAM28, ADAMTS1, ADAMTS8, and ADAMTS15. There were also significant differences in proteinase expression based on clinical parameters, i.e., tumor location, grade, and local invasion. MMP13 expression was significantly higher in large (>4 cm) locally invasive tumors (P < 0.05). MMP9 expression was significantly decreased in tumors with regional metastasis, whereas increased expression of ADAM8 was noted in the metastatic tumors (P < 0.001 for both). CONCLUSIONS: These findings suggest the HNSCC degradome as a valuable source of diagnostic, predictive, and prognostic molecular markers for these malignant tumors.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Gene Expression Profiling , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Protein Processing, Post-Translational , ADAM Proteins/genetics , ADAM Proteins/metabolism , Adult , Aged , Aged, 80 and over , Animals , Body Fluids/metabolism , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Cricetinae , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Humans , Male , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Metabolome/genetics , Middle Aged , Protein Processing, Post-Translational/genetics , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism
8.
Methods Mol Biol ; 622: 159-73, 2010.
Article in English | MEDLINE | ID: mdl-20135281

ABSTRACT

Quantitative reverse transcriptase polymerase chain reaction enables the accurate quantification of gene expression in cultured cells or small tissue samples. In this chapter, we describe the use of Taqman((R)) technology to measure expression of matrix metalloproteinases and related genes.


Subject(s)
Gene Expression Profiling/methods , Matrix Metalloproteinases/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Tissue Inhibitor of Metalloproteinases/metabolism , Animals , Gene Expression Regulation, Enzymologic , Humans , Matrix Metalloproteinases/metabolism , RNA/isolation & purification , RNA Probes , Reverse Transcription/genetics , Tissue Inhibitor of Metalloproteinases/genetics
9.
Am J Pathol ; 175(3): 1226-34, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19661439

ABSTRACT

Brk, a tyrosine kinase expressed in a majority of breast tumors, but not normal mammary tissue, promotes breast carcinoma cell proliferation. Normal epithelial cells are dependent on cell-cell or cell-matrix interactions for survival and undergo apoptosis after disruption of these interactions. Tumor cells are less sensitive to the induction of apoptosis and are predicted to have the potential to disseminate. We investigated whether Brk has further roles in breast tumor progression by relating its expression to tumor grade and demonstrating its role in the regulation of carcinoma cell survival under non-adherent conditions. Brk expression was determined by reverse transcription PCR on RNA extracted from surgical samples of human breast cancers. Breast carcinoma cell survival in suspension culture was examined when Brk protein levels were suppressed by RNA interference. Additionally, the effect of experimentally overexpressing Brk in otherwise Brk-negative breast carcinoma cells was assessed. Brk mRNA expression was notably higher in grade 3 breast tumors, as compared with lower tumor grades. In suspension culture, Brk suppression increased the rate of cell death, as compared with controls, and this cell death program exhibited characteristics of autophagy but not of apoptosis. Conversely, experimental expression of Brk in Brk-negative cells increased cell survival whereas kinase-inactive Brk did not. Therefore, Brk enhances breast carcinoma cell survival in suspension, suggesting a role for Brk in supporting breast cancer cell dissemination.


Subject(s)
Autophagy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Neoplasm Proteins/biosynthesis , Protein-Tyrosine Kinases/biosynthesis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Cell Adhesion , Cell Proliferation , Cell Survival , Cells, Cultured , Disease Progression , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Proteins/antagonists & inhibitors , Polymerase Chain Reaction , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA Interference , RNA, Neoplasm/analysis
10.
Arthritis Res Ther ; 11(3): R96, 2009.
Article in English | MEDLINE | ID: mdl-19549314

ABSTRACT

INTRODUCTION: The molecular mechanisms underlying cartilage destruction in osteoarthritis are poorly understood. Proteolysis is a key feature in the turnover and degradation of cartilage extracellular matrix where the focus of research has been on the metzincin family of metalloproteinases. However, there is strong evidence to indicate important roles for other catalytic classes of proteases, with both extracellular and intracellular activities. The aim of this study was to profile the expression of the majority of protease genes in all catalytic classes in normal human cartilage and that from patients with osteoarthritis (OA) using a quantitative method. METHODS: Human cartilage was obtained from femoral heads at joint replacement for either osteoarthritis or following fracture to the neck of femur (NOF). Total RNA was purified, and expression of genes assayed using Taqman low-density array quantitative RT-PCR. RESULTS: A total of 538 protease genes were profiled, of which 431 were expressed in cartilage. A total of 179 genes were differentially expressed in OA versus NOF cartilage: eight aspartic proteases, 44 cysteine proteases, 76 metalloproteases, 46 serine proteases and five threonine proteases. Wilcoxon ranking as well as the LogitBoost-NR machine learning approach were used to assign significance to each gene, with the most highly ranked genes broadly similar using each method. CONCLUSIONS: This study is the most complete quantitative analysis of protease gene expression in cartilage to date. The data help give direction to future research on the specific function(s) of individual proteases or protease families in cartilage and may help to refine anti-proteolytic strategies in OA.


Subject(s)
Cartilage, Articular/pathology , Gene Expression Profiling/methods , ADAM Proteins/biosynthesis , ADAM Proteins/genetics , Adult , Aged , Aged, 80 and over , Cartilage, Articular/enzymology , Cartilage, Articular/physiology , Enzyme Precursors/biosynthesis , Enzyme Precursors/genetics , Female , Femur Head/enzymology , Femur Head/metabolism , Femur Head/pathology , Humans , Male , Matrix Metalloproteinases/biosynthesis , Matrix Metalloproteinases/genetics , Middle Aged , Osteoarthritis/enzymology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Reverse Transcriptase Polymerase Chain Reaction
11.
Breast Cancer Res ; 11(2): R24, 2009.
Article in English | MEDLINE | ID: mdl-19405959

ABSTRACT

INTRODUCTION: The stromal microenvironment has a profound influence on tumour cell behaviour. In tumours, the extracellular matrix (ECM) composition differs from normal tissue and allows novel interactions to influence tumour cell function. The ECM protein tenascin-C (TNC) is frequently up-regulated in breast cancer and we have previously identified two novel isoforms - one containing exon 16 (TNC-16) and one containing exons 14 plus 16 (TNC-14/16). METHODS: The present study has analysed the functional significance of this altered TNC isoform profile in breast cancer. TNC-16 and TNC-14/16 splice variants were generated using PCR-ligation and over-expressed in breast cancer cells (MCF-7, T47D, MDA-MD-231, MDA-MB-468, GI101) and human fibroblasts. The effects of these variants on tumour cell invasion and proliferation were measured and compared with the effects of the large (TNC-L) and fully spliced small (TNC-S) isoforms. RESULTS: TNC-16 and TNC-14/16 significantly enhanced tumour cell proliferation (P < 0.05) and invasion, both directly (P < 0.01) and as a response to transfected fibroblast expression (P < 0.05) with this effect being dependent on tumour cell interaction with TNC, because TNC-blocking antibodies abrogated these responses. An analysis of 19 matrix metalloproteinases (MMPs) and tissue inhibitor of matrix metalloproteinases 1 to 4 (TIMP 1 to 4) revealed that TNC up-regulated expression of MMP-13 and TIMP-3 two to four fold relative to vector, and invasion was reduced in the presence of MMP inhibitor GM6001. However, this effect was not isoform-specific but was elicited equally by all TNC isoforms. CONCLUSIONS: These results demonstrate a dual requirement for TNC and MMP in enhancing breast cancer cell invasion, and identify a significant role for the tumour-associated TNC-16 and TNC-14/16 in promoting tumour invasion, although these isoform-specific effects appear to be mediated through MMP-independent mechanisms.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Matrix Metalloproteinases/metabolism , Tenascin/physiology , Alternative Splicing , Blotting, Western , Breast Neoplasms/genetics , Cell Adhesion , Cell Movement , Cell Proliferation , Female , Humans , Matrix Metalloproteinases/genetics , Neoplasm Invasiveness , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism , Up-Regulation , Tissue Inhibitor of Metalloproteinase-4
12.
Arterioscler Thromb Vasc Biol ; 29(9): 1251-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19325140

ABSTRACT

OBJECTIVE: Impairment of transforming growth factor (TGF)-beta1 signaling accelerates atherosclerosis in experimental mice. However, it is uncertain whether increased TGF-beta1 expression would retard atherosclerosis. The role of TGF-beta1 in aneurysm formation is also controversial. We tested whether overexpression of active TGF-beta1 in hyperlipidemic mice affects atherogenesis and aortic dilation. METHODS AND RESULTS: We generated apolipoprotein E-null mice with transgenes that allow regulated overexpression of active TGF-beta1 in their hearts. Compared to littermate controls, these mice had elevated cardiac and plasma TGF-beta1, less aortic root atherosclerosis (P< or =0.002), fewer lesions in the thoracic and abdominal aortae (P< or =0.01), less aortic root dilation (P<0.001), and fewer pseudoaneurysms (P=0.02). Mechanistic studies revealed no effect of TGF-beta1 overexpression on plasma lipids or cytokines, or on peripheral lymphoid organ cells. However, aortae of TGF-beta1-overexpressing mice had fewer T-lymphocytes, more collagen, less lipid, lower expression of inflammatory cytokines and matrix metalloproteinase-13, and higher expression of tissue inhibitor of metalloproteinase-2. CONCLUSIONS: When overexpressed in the heart and plasma, TGF-beta1 is an antiatherogenic, vasculoprotective cytokine that limits atherosclerosis and prevents aortic dilation. These actions are associated with significant changes in cellularity, collagen and lipid accumulation, and gene expression in the artery wall.


Subject(s)
Aneurysm, False/prevention & control , Aortic Aneurysm/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Hyperlipidemias/metabolism , Myocardium/metabolism , Transforming Growth Factor beta1/metabolism , Aneurysm, False/genetics , Aneurysm, False/metabolism , Aneurysm, False/pathology , Animals , Aortic Aneurysm/genetics , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Collagen/metabolism , Dilatation, Pathologic , Disease Models, Animal , Female , Gene Expression Regulation , Hyperlipidemias/complications , Hyperlipidemias/genetics , Hyperlipidemias/pathology , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction , T-Lymphocytes/immunology , Time Factors , Transforming Growth Factor beta1/blood , Transforming Growth Factor beta1/genetics
13.
Mol Carcinog ; 48(7): 618-25, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19058297

ABSTRACT

Matrix metalloproteinases (MMPs) have been linked to the metastatic potential of tumor cells due to their ability to degrade the extracellular matrix. MMP-3 (stromelysin-1) is upregulated in a wide variety of human tumors. We used the MMTV-PyMT breast cancer model to determine if MMP-3 is involved in tumorigenesis and metastatic growth. In this model the stromal expression of MMP-3 mRNA resembles the predominant MMP-3 expression pattern observed in human ductal breast carcinomas. We studied a cohort of 63 PyMT transgenic mice, either deficient for MMP-3 or wild-type controls. The degree of metastasis did not differ significantly between the two groups of mice, although the median lung metastasis volume was more than threefold increased in MMTV-PyMT mice deficient in MMP-3. Likewise, primary tumor growth rate and lymph node metastasis were not significantly affected by MMP-3-deficiency. By comparing mRNA levels in MMP-3-deficient PyMT tumors with PyMT wild-type tumors we excluded compensatory transcriptional changes of other MMPs or their specific inhibitors. Thus, we conclude that genetic ablation of MMP-3 does not significantly affect tumor growth and metastasis in the MMTV-PyMT model.


Subject(s)
Matrix Metalloproteinase 3/metabolism , Neoplasm Metastasis , Neoplasms, Experimental/pathology , Animals , Base Sequence , DNA Primers , Female , Hydrolysis , Male , Matrix Metalloproteinase 3/genetics , Mice , Mice, Transgenic , Neoplasms, Experimental/enzymology , Polymerase Chain Reaction , RNA, Messenger/genetics
14.
J Immunol ; 182(1): 49-53, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109134

ABSTRACT

MHC class I-related chain (MIC) A/B are transmembrane proteins expressed in pathological conditions that are ligands for the activating receptor NKG2D found on cytotoxic lymphocytes. Soluble NKG2D ligands are detected in sera of patients suffering from multiple types of cancer where they are associated with reduced levels of receptor expression and compromised function of NK and CTLs. In this study, we report the identification of a metalloproteinase involved in the cleavage process of MIC; inhibition and knockdown of ADAM17/TACE blocks the shedding of these proteins. Strikingly, the recruitment of both enzyme and substrate to detergent-resistant membrane microdomains is crucial for efficient proteolysis. These findings provide a novel insight into the molecular mechanisms of MIC shedding.


Subject(s)
ADAM Proteins/physiology , Histocompatibility Antigens Class I/metabolism , Membrane Microdomains/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM17 Protein , Cell Line , Cell Line, Tumor , Dipeptides/pharmacology , Humans , Hydrolysis/drug effects , Membrane Microdomains/enzymology , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Solubility , Thiophenes/pharmacology
15.
Oncol Rep ; 20(6): 1561-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19020742

ABSTRACT

Prostate cancer cells exist under hypoxic conditions. Hypoxia has a detrimental effect on the efficacy of treatment and final outcome in patients with prostate cancer. There have been a large number of endogenous markers of hypoxia described previously across a range of cancer types, both in vitro and in vivo. The aim of this study was to evaluate the expression of a range of hypoxia-associated genes within benign prostatic hypertrophy (BPH) and prostate cancer tissue. Messenger RNA was extracted from primary prostate tissue obtained from 67 men with benign prostatic hypertrophy or prostate cancer (Gleason score 5 to 10). Real-time polymerase chain reaction was performed to quantify the expression levels of 12 hypoxia-associated genes in these tissues. Expression of lysyl oxidase (LOX) and glucose transporter-1 (GLUT-1) genes were significantly higher in prostate cancer compared with BPH tissue (P<0.05) and correlated with Gleason score (LOX: R=0.297, P=0.015; GLUT-1: R=0.274, P=0.026). HIF-2alpha had a negative correlation with Gleason score (R= -0.309, P=0.012). The remaining hypoxia-associated genes did not show any specific pattern of expression in prostate tissue. Numerous molecules have been proposed as endogenous markers of hypoxia. The findings of this study illustrate that not all hypoxia-associated molecules are relevant to prostate cancer in vivo. However, LOX and GLUT-1 are candidate markers of hypoxia in prostate cancer and may prove useful in identifying patients with hypoxic prostate cancer. Not all hypoxia-associated molecules are relevant in prostate cancer in vivo.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glucose Transporter Type 1/metabolism , Hypoxia , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein-Lysine 6-Oxidase/metabolism , DNA Primers/chemistry , Dimerization , Disease Progression , Humans , Male , Models, Biological , Oxygen/metabolism , Prostatic Neoplasms/enzymology , RNA, Neoplasm/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
Mol Aspects Med ; 29(5): 258-89, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18762209

ABSTRACT

The ADAMs (a disintegrin and metalloproteinase) are a fascinating family of transmembrane and secreted proteins with important roles in regulating cell phenotype via their effects on cell adhesion, migration, proteolysis and signalling. Though all ADAMs contain metalloproteinase domains, in humans only 13 of the 21 genes in the family encode functional proteases, indicating that at least for the other eight members, protein-protein interactions are critical aspects of their biological functions. The functional ADAM metalloproteinases are involved in "ectodomain shedding" of diverse growth factors, cytokines, receptors and adhesion molecules. The archetypal activity is shown by ADAM-17 (tumour necrosis factor-alpha convertase, TACE), which is the principal protease involved in the activation of pro-TNF-alpha, but whose sheddase functions cover a broad range of cell surface molecules. In particular, ADAM-17 is required for generation of the active forms of Epidermal Growth Factor Receptor (EGFR) ligands, and its function is essential for the development of epithelial tissues. Several other ADAMs have important sheddase functions in particular tissue contexts. Another major family member, ADAM-10, is a principal player in signalling via the Notch and Eph/ephrin pathways. For a growing number of substrates, foremost among them being Notch, cleavage by ADAM sheddases is essential for their subsequent "regulated intramembrane proteolysis" (RIP), which generates cleaved intracellular domains that translocate to the nucleus and regulate gene transcription. Several ADAMs play roles in spermatogenesis and sperm function, potentially by effecting maturation of sperm and their adhesion and migration in the uterus. Other non-catalytic ADAMs function in the CNS via effects on guidance mechanisms. The ADAM family are thus fundamental to many control processes in development and homeostasis, and unsurprisingly they are also linked to pathological states when their functions are dysregulated, including cancer, cardiovascular disease, asthma, Alzheimer's disease. This review will provide an overview of current knowledge of the human ADAMs, discussing their structure, function, regulation and disease involvement.


Subject(s)
ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM Proteins/chemistry , ADAM Proteins/classification , Animals , Disease , Enzyme Activation , Enzyme Inhibitors/metabolism , Evolution, Molecular , Humans , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Conformation , Protein Structure, Tertiary
17.
PLoS One ; 3(8): e2959, 2008 Aug 13.
Article in English | MEDLINE | ID: mdl-18698413

ABSTRACT

Matrix metalloproteinase (MMP) 13 (collagenase 3) is an extracellular matrix remodeling enzyme that is induced in myofibroblasts during the earliest invasive stages of human breast carcinoma, suggesting that it is involved in tumor progression. During progression of mammary carcinomas in the polyoma virus middle T oncogene mouse model (MMTV-PyMT), Mmp13 mRNA was strongly upregulated concurrently with the transition to invasive and metastatic carcinomas. As in human tumors, Mmp13 mRNA was found in myofibroblasts of invasive grade II and III carcinomas, but not in benign grade I and II mammary intraepithelial neoplasias. To determine if MMP13 plays a role in tumor progression, we crossed MMTV-PyMT mice with Mmp13 deficient mice. The absence of MMP13 did not influence tumor growth, vascularization, progression to more advanced tumor stages, or metastasis to the lungs, and the absence of MMP13 was not compensated for by expression of other MMPs or tissue inhibitor of metalloproteinases. However, an increased fraction of thin collagen fibrils was identified in MMTV-PyMT;Mmp13(-/-) compared to MMTV-PyMT;Mmp13(+/+) tumors, showing that collagen metabolism was altered in the absence of MMP13. We conclude that the expression pattern of Mmp13 mRNA in myofibroblasts of invasive carcinomas in the MMTV-PyMT breast cancer model recapitulates the expression pattern observed in human breast cancer. Our results suggest that MMP13 is a marker of carcinoma-associated myofibroblasts of invasive carcinoma, even though it does not make a major contribution to tumor progression in the MMTV-PyMT breast cancer model.


Subject(s)
Antigens, Polyomavirus Transforming/pharmacology , Fibroblasts/enzymology , Fibroblasts/virology , Mammary Neoplasms, Animal/enzymology , Mammary Neoplasms, Animal/pathology , Matrix Metalloproteinase 13/biosynthesis , Animals , DNA Primers , Disease Progression , Female , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/blood supply , Matrix Metalloproteinase 13/deficiency , Matrix Metalloproteinase 13/genetics , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasm Staging , Neovascularization, Pathologic
18.
Cancer Res ; 68(8): 2755-63, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18413742

ABSTRACT

Collagenase-2 (matrix metalloproteinase-8, MMP-8) is an MMP mainly produced by neutrophils and associated with many inflammatory conditions. We have previously described that MMP-8 plays a protective role in cancer through its ability to regulate the inflammatory response induced by carcinogens. Moreover, it has been reported that experimental manipulation of the expression levels of this enzyme alters the metastatic behavior of human breast cancer cells. In this work, we have used mutant mice deficient in MMP-8 and syngenic melanoma and lung carcinoma tumor cells lines overexpressing this enzyme to further explore the putative antimetastatic potential of MMP-8. We report herein that MMP-8 prevents metastasis formation through the modulation of tumor cell adhesion and invasion. Thus, tumor cells overexpressing MMP-8 have an increased adhesion to extracellular matrix proteins, whereas their invasive ability through Matrigel is substantially reduced when compared with control cells. Analysis of MMP-8 in breast cancer patients revealed that the expression of this metalloproteinase by breast tumors correlates with a lower incidence of lymph node metastasis and confers good prognosis to these patients. On this basis, we propose that MMP-8 is a tumor protective factor, which also has the ability to reduce the metastatic potential of malignant cells in both mice and human.


Subject(s)
Cell Adhesion/physiology , Lung Neoplasms/pathology , Matrix Metalloproteinase 8/deficiency , Melanoma/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Animals , Cell Division , Cell Line, Tumor , Cell Movement , Crosses, Genetic , Humans , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 8/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
J Biol Chem ; 283(14): 9465-74, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18245084

ABSTRACT

Matrix metalloproteinases (MMPs) have been implicated in a variety of human diseases, including neuroimmunological disorders such as multiple sclerosis. However, the recent finding that some MMPs play paradoxical protective roles in these diseases has made necessary the detailed study of the specific function of each family member in their pathogenesis. To determine the relevance of collagenase-2 (MMP-8) in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, we have performed two different analyses involving genetic and biochemical approaches. First, we have analyzed the development of EAE in mutant mouse deficient in MMP-8, with the finding that the absence of this proteolytic enzyme is associated with a marked reduction in the clinical symptoms of EAE. We have also found that MMP-8(-/-) mice exhibit a marked reduction in central nervous system-infiltrating cells and demyelinating lesions. As a second approach, we have carried out a pharmacological inhibition of MMP-8 with a selective inhibitor against this protease (IC(50) = 0.4 nM). These studies have revealed that the administration of the MMP-8 selective inhibitor to mice with EAE also reduces the severity of the disease. Based on these findings, we conclude that MMP-8 plays an important role in EAE development and propose that this enzyme may be a novel therapeutic target in human neuro-inflammatory diseases such as multiple sclerosis.


Subject(s)
Central Nervous System/enzymology , Encephalomyelitis, Autoimmune, Experimental/enzymology , Matrix Metalloproteinase 8/metabolism , Multiple Sclerosis/enzymology , Animals , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase Inhibitors , Mice , Mice, Knockout , Multiple Sclerosis/drug therapy , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use
20.
Mol Cancer Res ; 6(3): 383-94, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18296648

ABSTRACT

Adamalysins [a disintegrin and metalloproteinase (ADAM)] are a family of cell surface transmembrane proteins that have broad biological functions encompassing proteolysis, adhesion, and cell signal regulation. We previously showed that the cytoplasmic domain of ADAM-15 interacts with Src family protein tyrosine kinases and the adaptor protein growth factor receptor binding protein 2 (Grb2). In the present study, we have cloned and characterized four alternatively spliced forms of ADAM-15, which differ only in their cytoplasmic domains. We show that the four ADAM-15 variants were differentially expressed in human mammary carcinoma tissues compared with normal breast. The expression of the individual isoforms did not correlate with age, menopausal status, tumor size or grade, nodal status, Nottingham Prognostic Index, or steroid hormone receptor status. However, higher levels of two isoforms (ADAM-15A and ADAM-5B) were associated with poorer relapse-free survival in node-negative patients, whereas elevated ADAM-15C correlated with better relapse-free survival in node-positive, but not in node-negative, patients. The expression of ADAM-15A and ADAM-15B variants in MDA-MB-435 cells had differential effects on cell morphology, with adhesion, migration, and invasion enhanced by expression of ADAM-15A, whereas ADAM-15B led to reduced adhesion. Using glutathione S-transferase pull-down assays, we showed that the cytoplasmic domains of ADAM-15A, ADAM-15B, and ADAM-15C show equivalent abilities to interact with extracellular signal-regulated kinase and the adaptor molecules Grb2 and Tks5/Fish, but associate in an isoform-specific fashion with Nck and the Src and Brk tyrosine kinases. These data indicate that selective expression of ADAM-15 variants in breast cancers could play an important role in determining tumor aggressiveness by interplay with intracellular signaling pathways.


Subject(s)
ADAM Proteins/genetics , Breast Neoplasms/genetics , Genetic Variation , Membrane Proteins/genetics , ADAM Proteins/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Cytoplasm/physiology , Female , Humans , Lymphatic Metastasis , Membrane Proteins/metabolism , Middle Aged , Postmenopause , Premenopause , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...