Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Ecotoxicol Environ Saf ; 142: 544-554, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28482323

ABSTRACT

Bioassays of planarian neoplasia highlight the potential of these organisms as useful standards to assess whether environmental toxins such as cadmium promote tumorigenesis. These studies complement other investigations into the exceptional healing and regeneration of planarians - processes that are driven by a population of active stem cells, or neoblasts, which are likely transformed during planarian tumor growth. Our goal was to determine if planarian tumorigenesis assays are amenable to mechanistic studies of cadmium carcinogenesis. To that end we demonstrate, by examining both counts of cell populations by size, and instances of mitosis, that the activity of the stem cell population can be monitored. We also provide evidence that specific biomodulators can affect the potential of planarian neoplastic growth, in that an inhibitor of metalloproteinases effectively blocked the development of the lesions. From these results, we infer that neoblast activity does respond to cadmium-induced tumor growth, and that metalloproteinases are required for the progression of cancer in the planarian.


Subject(s)
Cadmium/toxicity , Carcinogens/toxicity , Cell Transformation, Neoplastic/chemically induced , Models, Biological , Planarians/drug effects , Animals , Benchmarking , Carcinogenicity Tests , Cell Transformation, Neoplastic/ultrastructure , Cocarcinogenesis , Mitosis/drug effects , Planarians/cytology , Regeneration/drug effects
2.
Nat Rev Cancer ; 17(1): 38-53, 2017 01.
Article in English | MEDLINE | ID: mdl-27932800

ABSTRACT

A compelling long-term goal of cancer biology is to understand the crucial players during tumorigenesis in order to develop new interventions. Here, we review how the four non-redundant tissue inhibitors of metalloproteinases (TIMPs) regulate the pericellular proteolysis of a vast range of matrix and cell surface proteins, generating simultaneous effects on tumour architecture and cell signalling. Experimental studies demonstrate the contribution of TIMPs to the majority of cancer hallmarks, and human cancers invariably show TIMP deregulation in the tumour or stroma. Of the four TIMPs, TIMP1 overexpression or TIMP3 silencing is consistently associated with cancer progression or poor patient prognosis. Future efforts will align mouse model systems with changes in TIMPs in patients, will delineate protease-independent TIMP function, will pinpoint therapeutic targets within the TIMP-metalloproteinase-substrate network and will use TIMPs in liquid biopsy samples as biomarkers for cancer prognosis.


Subject(s)
Neoplasms/metabolism , Neoplasms/physiopathology , Tissue Inhibitor of Metalloproteinases/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/physiology , Humans , Proteolysis , Signal Transduction , Tissue Inhibitor of Metalloproteinases/physiology , Tumor Microenvironment/physiology
3.
J Immunol ; 188(6): 2876-83, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22323541

ABSTRACT

Lymphocyte infiltration into epithelial tissues and proinflammatory cytokine release are key steps in autoimmune disease. Although cell-autonomous roles of lymphocytes are well studied in autoimmunity, much less is understood about the stromal factors that dictate immune cell function. Tissue inhibitor of metalloproteinases 3 (TIMP3) controls systemic cytokine bioavailability and signaling by inhibiting the ectodomain shedding of cytokines and their receptors. The role of TIMP3 in cytokine biology is emerging; however, its contribution to cellular immunology remains unknown. In this study, we show that TIMP3 produced by the hepatic stroma regulates the basal lymphocyte populations in the liver and prevents autoimmune hepatitis. TIMP3 deficiency in mice led to spontaneous accumulation and activation of hepatic CD4(+), CD8(+), and NKT cells. Treatment with Con A in a model of polyclonal T lymphocyte activation resulted in a greatly enhanced Th1 cytokine response and acute liver failure, which mechanistically depended on TNF signaling. Bone marrow chimeras demonstrated that TIMP3 derived from the stromal rather than hematopoietic compartment provided protection against autoimmunity. Finally, we identified hepatocytes as the major source of Timp3 in a resting liver, whereas significant Timp3 gene transcription was induced by hepatic stellate cells in the inflamed liver. These results uncover metalloproteinase inhibitors as critical stromal factors in regulating cellular immunity during autoimmune hepatitis.


Subject(s)
Extracellular Matrix/immunology , Hepatitis, Autoimmune/immunology , Liver/immunology , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Tissue Inhibitor of Metalloproteinase-3/immunology , Animals , Cell Separation , Flow Cytometry , Hepatitis, Autoimmune/metabolism , Hepatitis, Autoimmune/pathology , Immunoblotting , Immunohistochemistry , Liver/cytology , Liver/metabolism , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/metabolism , Th1 Cells/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism
4.
J Clin Invest ; 120(8): 2731-44, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20628198

ABSTRACT

The cell death receptor Fas plays a role in the establishment of fulminant hepatitis, a major cause of drug-induced liver failure. Fas activation elicits extrinsic apoptotic and hepatoprotective signals; however, the mechanisms by which these signals are integrated during disease are unknown. Tissue inhibitor of metalloproteinases 3 (TIMP3) controls the critical sheddase a disintegrin and metalloproteinase 17 (ADAM17) and may dictate stress signaling. Using mice and cells lacking TIMP3, ADAM17, and ADAM17-regulated cell surface molecules, we have found that ADAM17-mediated ectodomain shedding of TNF receptors and EGF family ligands controls activation of multiple signaling cascades in Fas-induced hepatitis. We demonstrated that TNF signaling promoted hepatotoxicity, while excessive TNF receptor 1 (TNFR1) shedding in Timp3-/- mice was protective. Compound Timp3-/-Tnf-/- and Timp3-/-Tnfr1-/- knockout conferred complete resistance to Fas-induced toxicity. Loss of Timp3 enhanced metalloproteinase-dependent EGFR signaling due to increased release of the EGFR ligands TGF-alpha, amphiregulin, and HB-EGF, while depletion of shed amphiregulin resensitized Timp3-/- hepatocytes to apoptosis. Finally, adenoviral delivery of Adam17 prevented acetaminophen-induced liver failure in a clinically relevant model of Fas-dependent fulminant hepatitis. These findings demonstrate that TIMP3 and ADAM17 cooperatively dictate cytokine signaling during death receptor activation and indicate that regulated metalloproteinase activity integrates survival and death signals during acute hepatotoxic stress.


Subject(s)
ADAM Proteins/physiology , Apoptosis , Chemical and Drug Induced Liver Injury/pathology , ErbB Receptors/physiology , Hepatocytes/pathology , Liver Failure, Acute/pathology , Receptors, Tumor Necrosis Factor, Type I/physiology , Tissue Inhibitor of Metalloproteinase-3/physiology , ADAM17 Protein , Animals , Cells, Cultured , Cytoprotection , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction , fas Receptor/physiology
5.
J Biol Chem ; 284(43): 29893-904, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19625257

ABSTRACT

The pleiotropic cytokines, transforming growth factor beta1 (TGFbeta1), and tumor necrosis factor (TNF) play critical roles in tissue homeostasis in response to injury and are implicated in multiple human diseases and cancer. We reported that the loss of Timp3 (tissue inhibitor of metalloproteinase 3) leads to abnormal TNF signaling and cardiovascular function. Here we show that parallel deregulation of TGFbeta1 and TNF signaling in Timp3(-/-) mice amplifies their cross-talk at the onset of cardiac response to mechanical stress (pressure overload), resulting in fibrosis and early heart failure. Microarray analysis showed a distinct gene expression profile in Timp3(-/-) hearts, highlighting activation of TGFbeta1 signaling as a potential mechanism underlying fibrosis. Neonatal cardiomyocyte-cardiofibroblast co-cultures were established to measure fibrogenic response to agonists known to be induced following mechanical stress in vivo. A stronger response occurred in neonatal Timp3(-/-) co-cultures, as determined by increased Smad signaling and collagen expression, due to increased TNF processing and precocious proteolytic maturation of TGFbeta1 to its active form. The relationship between TGFbeta1 and TNF was dissected using genetic and pharmacological manipulations. Timp3(-/-)/Tnf(-/-) mice had lower TGFbeta1 than Timp3(-/-), and anti-TGFbeta1 antibody (1D11) negated the abnormal TNF response, indicating their reciprocal stimulatory effects, with each manipulation abolishing fibrosis and improving heart function. Thus, TIMP3 is a common innate regulator of TGFbeta1 and TNF in tissue response to injury. The matrix-bound TIMP3 balances the anti-inflammatory and proinflammatory processes toward constructive tissue remodeling.


Subject(s)
Endomyocardial Fibrosis/metabolism , Signal Transduction , Tissue Inhibitor of Metalloproteinase-3 , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Coculture Techniques , Collagen/biosynthesis , Collagen/genetics , Endomyocardial Fibrosis/genetics , Gene Expression Profiling , Gene Expression Regulation/genetics , Humans , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/genetics , Tumor Necrosis Factor-alpha/genetics
6.
Hepatology ; 47(1): 177-85, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18008367

ABSTRACT

UNLABELLED: Matrix metalloproteinases (MMPs) have been implicated in the hepatic injury induced after cold ischemia-warm reperfusion (CI-WR), by altering the extracellular matrix (ECM), but their precise role remains unknown. The hepatic MMP expression was evaluated after 2 conditions of CI (4 degrees C for 24 and 42 hours: viable and nonviable livers) followed by different periods of WR, using isolated perfused rat livers. CI-WR induced moderate changes in hepatic MMP transcript levels not influenced by CI duration, whereas gelatinase activities accumulated in liver effluents. Therefore, the protective effect of a new phosphinic MMP inhibitor, RXP409, was tested after prolonged CI. RXP409 (10 microM) was added to the University of Wisconsin solution, and livers were preserved for 42 hours (4 degrees C), then reperfused for 1 hour in Krebs solution (37 degrees C), containing 20% erythrocytes. Liver viability parameters were recorded, and the extent of cell necrosis was evaluated on liver biopsies, using trypan blue nuclear uptake. Treatment with RXP409 significantly improved liver function (transaminase release and bile secretion) and liver injury. In particular, the MMP inhibitor significantly modified the extent of cell death from large clusters of necrotic hepatocytes as found in control livers (2%-60% of liver biopsies; mean, 26% +/- 9%) to isolated necrotic hepatocytes as found in treated livers (0.2%-12%; mean, 3% +/- 2%) (P < 0.05). CONCLUSION: These data demonstrate that MMPs, by altering the ECM, play a major role in liver CI-WR injury leading to extensive hepatocyte necrosis and that their inhibition might prove to be a new strategy in improving preservation solutions.


Subject(s)
Cold Ischemia/adverse effects , Liver Diseases/enzymology , Matrix Metalloproteinases/metabolism , Phosphinic Acids/therapeutic use , Reperfusion Injury/enzymology , Tissue Inhibitor of Metalloproteinases/metabolism , Tryptophan/analogs & derivatives , Animals , Apoptosis/drug effects , Endothelial Cells/drug effects , Enzyme Inhibitors/therapeutic use , Fibronectins/metabolism , Gene Expression , Hepatocytes/drug effects , Liver Diseases/metabolism , Male , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/genetics , Necrosis/prevention & control , Phosphinic Acids/pharmacology , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Time Factors , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Survival/drug effects , Tryptophan/pharmacology , Tryptophan/therapeutic use
7.
Am J Respir Crit Care Med ; 176(11): 1098-107, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17673693

ABSTRACT

RATIONALE: Different sensitivities to profibrotic compounds such as bleomycin are observed among mouse strains. OBJECTIVES: To identify genetic factors contributing to the outcome of lung injury. METHODS: Physiological comparison of C57BL/6 (sensitive) and BALB/c (resistant) mice challenged by intratracheal bleomycin instillation revealed several early differences: global gene expression profiles were thus established from lungs derived from the two strains, in the absence of any bleomycin administration. MEASUREMENTS AND MAIN RESULTS: Expression of 25 genes differed between the two strains. Among them, two molecules, not previously associated with pulmonary fibrosis, were identified. The first corresponded to dipeptidyl-peptidase I (DPPI), a cysteine peptidase (also known as cathepsin C) essential for the activation of serine proteinases produced by immune/inflammatory cells. The second corresponded to tissue inhibitor of matrix metalloproteinase-3, which also inhibits members of the ADAM (a disintegrin and metalloproteinase) family, such as the tumor necrosis factor-converting enzyme. In functional studies performed in the bleomycin-induced lung fibrosis model, the level of expression of these two genes was closely correlated with specific early events associated with lung fibrosis, namely activation of polymorphonuclear neutrophil-derived serine proteases and tumor necrosis factor-alpha-dependent inflammatory syndrome. Surprisingly, genetic deletion of DPPI in the context of a C57BL/6 genetic background did not protect against bleomycin-mediated fibrosis, suggesting additional function(s) for this key enzyme. CONCLUSIONS: This study highlights the importance of the early inflammatory events that follow bleomycin instillation in the development of lung fibrosis, and describes for the first time the roles that DPPI and tissue inhibitor of matrix metalloproteinase-3 may play in this process.


Subject(s)
Bleomycin , Cathepsin C/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Apoptosis , Bleomycin/administration & dosage , Bronchoalveolar Lavage Fluid/chemistry , Cathepsin C/deficiency , Eosinophilia/chemically induced , Eosinophilia/etiology , Gene Expression Profiling , Genetic Predisposition to Disease , Instillation, Drug , Interleukin-5/metabolism , Lung/metabolism , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred BALB C/genetics , Mice, Inbred BALB C/metabolism , Mice, Inbred C57BL/genetics , Mice, Inbred C57BL/metabolism , Mice, Knockout , Neutrophils/enzymology , Oligonucleotide Array Sequence Analysis , Pneumonia/physiopathology , Serine Endopeptidases/metabolism , Species Specificity , Trachea , Tumor Necrosis Factor-alpha/metabolism
8.
Nucleic Acids Res ; 34(12): e87, 2006 Jul 19.
Article in English | MEDLINE | ID: mdl-16855282

ABSTRACT

Two collections of oligonucleotides have been designed for preparing pangenomic human and mouse microarrays. A total of 148,993 and 121,703 oligonucleotides were designed against human and mouse transcripts. Quality scores were created in order to select 25,342 human and 24,109 mouse oligonucleotides. They correspond to: (i) a BLAST-specificity score; (ii) the number of expressed sequence tags matching each probe; (iii) the distance to the 3' end of the target mRNA. Scores were also used to compare in silico the two microarrays with commercial microarrays. The sets described here, called RNG/MRC collections, appear at least as specific and sensitive as those from the commercial platforms. The RNG/MRC collections have now been used by an Anglo-French consortium to distribute more than 3500 microarrays to the academic community. Ad hoc identification of tissue-specific transcripts and a approximately 80% correlation with hybridizations performed on Affymetrix GeneChiptrade mark suggest that the RNG/MRC microarrays perform well. This work provides a comprehensive open resource for investigators working on human and mouse transcriptomes, as well as a generic method to generate new microarray collections in other organisms. All information related to these probes, as well as additional information about commercial microarrays have been stored in a freely-accessible database called MEDIANTE.


Subject(s)
Databases, Nucleic Acid , Gene Expression Profiling , Mice/genetics , Oligonucleotide Array Sequence Analysis , Oligonucleotide Probes/chemistry , Animals , Expressed Sequence Tags , Humans , Internet , Mice/metabolism , Transcription, Genetic
9.
Transpl Int ; 18(4): 444-52, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15773965

ABSTRACT

Prolonged ischemia used in liver surgery and/or transplantation causes cellular damage resulting in apoptosis and necrosis. Ischemia-reperfusion (I/R) led Kupffer cells to pro-inflammatory cytokines secretion [tumor necrosis factor (TNF)-alpha, interleukin-1] which involve chemokines secretion by hepatocytes. These chemokines have neutrophil chemotactic properties and neutrophils are involved in the development of I/R-induced necrosis. The aim of this study was to specify the consequence of partial oxygen pressure variation on hepatocyte chemokines synthesis and to verify if intermittent hypoxia and/or preconditioning could decrease it. It was performed on primary cultured mice hepatocytes and Kupffer cells, subjected to continuous, intermittent hypoxia or preconditioning phases, mimicking surgical processes. The chemokine secretion was evaluated by RNase protection assay and enzyme-linked immunosorbent assay method. Only monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-2 (MIP-2) mRNA formation were observed, especially after 1-h hypoxia followed by 10-h (for MCP-1) or 24-h reoxygenation (for MIP-2). In conclusion, TNF-alpha and coculture with Kupffer cells increased hepatocyte chemokines mRNA transcription, whereas surgical split up protocols (intermittent hypoxia and preconditioning) had no significant effect.


Subject(s)
Cell Hypoxia/physiology , Chemokines/genetics , Hepatocytes/physiology , Ischemic Preconditioning , Oxygen/pharmacology , Transcription, Genetic/drug effects , Animals , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL8 , Hepatocytes/drug effects , Male , Mice , Mice, Inbred Strains , Monocyte Chemoattractant Proteins/genetics , Monocyte Chemoattractant Proteins/metabolism
10.
J Biol Chem ; 280(2): 1272-83, 2005 Jan 14.
Article in English | MEDLINE | ID: mdl-15509588

ABSTRACT

Stromelysin-3 (ST3, MMP-11) has been shown to be strongly overexpressed in stromal fibroblasts of most invasive human carcinomas. However, the molecular mechanisms leading to ST3 expression in nonmalignant fibroblasts remain unknown. The aim of the present study was to analyze the signaling pathways activated in normal pulmonary fibroblasts after their interaction with non-small cell lung cancer (NSCLC) cells and leading to ST3 expression. The use of selective signaling pathway inhibitors showed that conventional and novel protein kinase Cs (PKC) were required for ST3 induction, whereas Src kinases exerted a negative control. We observed by both conventional and real time confocal microscopy that green fluorescent protein-tagged PKCalpha and PKCepsilon, but not PKCdelta, transfected in fibroblasts, accumulate selectively at the cell-cell contacts between fibroblasts and tumor cells. In agreement, RNAi-mediated depletion of PKCalpha and PKCepsilon, but not PKCdelta significantly decreased co-culture-dependent ST3 production. Finally, a tetracycline-inducible expression model allowed us to confirm the central role of these PKC isoforms and the negative regulatory function of c-Src in the control of ST3 expression. Altogether, our data emphasize signaling changes occurring in the tumor microenvironment that may define new stromal targets for therapeutic intervention.


Subject(s)
Metalloendopeptidases/biosynthesis , Neoplasms/metabolism , Neoplasms/pathology , Protein Kinase C/metabolism , Cell Adhesion , Cell Line , Cell Line, Tumor , Cell Membrane/enzymology , Coculture Techniques , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/metabolism , Green Fluorescent Proteins , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Matrix Metalloproteinase 11 , Metalloendopeptidases/metabolism , Protein Kinase C/biosynthesis , Protein Kinase C/genetics , Protein Transport , Signal Transduction/drug effects , Tetracycline/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL