Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell Physiol Biochem ; 52(5): 1003-1016, 2019.
Article in English | MEDLINE | ID: mdl-30977985

ABSTRACT

BACKGROUND/AIMS: The composition of the extracellular matrix (ECM) in the central nervous system (CNS) has several features that make it unique. For instance, it is remarkable for the presence of proteoglycans such as versican, brevican, and neurocan, some of which have been identified as substrates of different members of the ADAMTS family of secreted metalloproteases. Previous studies have associated ADAMTSs with the repair of the CNS, including recovery following degradation of glial scar tissue and the stimulation of axonal growth after brain injury. However, the involvement of ADAMTSs in diseases of the CNS is complex and not understood fully, and a current challenge is unraveling the precise roles of these metalloproteases in the brain. METHODS: ADAMTS12 and neurocan gene expression was examined by quantitative PCR. Western blot analysis was employed to detect ADAMTS12 and neurocan protein expression in cell lines, and immunostaining techniques were used to detect neurocan in mouse brain tissues. Neurocan cleavage using recombinant ADAMTS1, ADAMTS4, ADAMTS5, and ADAMTS12 metalloproteases was evaluated by western blotting. Cell adhesion and migration were assessed using uncoated culture dishes or dishes coated with Matrigel or ECM components. RESULTS: We identified neurocan as a novel component of brain ECM that can be cleaved by ADAMTS12. In addition, we showed that neurocan cleavage by ADAMTS12 altered the adhesive properties of the human neuroglioma H4 cell line. Moreover, immunohistochemical analysis of Adamts12-deficient mice revealed the significant accumulation of neurocan in the brain of neonatal mice. CONCLUSION: Overall, our results suggest that ADAMTS12 could be involved in the repair of the CNS through its ability to degrade neurocan. Moreover, it can be inferred that alterations in neurocan degradation processes could be associated with the pathogenesis of neurological disorders.


Subject(s)
ADAMTS Proteins/biosynthesis , ADAMTS Proteins/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Cranial Nerve Diseases/metabolism , Lectins, C-Type/metabolism , Nerve Tissue Proteins/metabolism , Proteoglycans/metabolism , Proteolysis , ADAMTS Proteins/genetics , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Chondroitin Sulfate Proteoglycans/genetics , Cranial Nerve Diseases/genetics , Cranial Nerve Diseases/pathology , Gene Expression Regulation , Humans , Lectins, C-Type/genetics , Mice , Nerve Tissue Proteins/genetics , Neurocan , Proteoglycans/genetics
3.
Mol Cell Biol ; 38(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29891514

ABSTRACT

Macrophages are prominent cells in acute and chronic inflammatory diseases. Recent studies highlight a role for macrophage proliferation post-monocyte recruitment under inflammatory conditions. Using an acute peritonitis model, we identify a significant defect in macrophage proliferation in mice lacking the leukocyte transmembrane protease ADAM17. The defect is associated with decreased levels of macrophage colony-stimulating factor 1 (CSF-1) in the peritoneum and is rescued by intraperitoneal injection of CSF-1. Cell surface CSF-1 (csCSF-1) is one of the substrates of ADAM17. We demonstrate that both infiltrated neutrophils and macrophages are major sources of csCSF-1. Furthermore, acute shedding of csCSF-1 following neutrophil extravasation is associated with elevated expression of iRhom2, a member of the rhomboid-like superfamily, which promotes ADAM17 maturation and trafficking to the neutrophil surface. Accordingly, deletion of hematopoietic iRhom2 is sufficient to prevent csCSF-1 release from neutrophils and macrophages and to prevent macrophage proliferation. In acute inflammation, csCSF-1 release and macrophage proliferation are self-limiting due to transient leukocyte recruitment and temporally restricted csCSF-1 expression. In chronic inflammation, such as atherosclerosis, the ADAM17-mediated lesional macrophage proliferative response is prolonged. Our results demonstrate a novel mechanism whereby ADAM17 promotes macrophage proliferation in states of acute and chronic inflammation.


Subject(s)
ADAM17 Protein/metabolism , Inflammation/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Neutrophils/metabolism , ADAM17 Protein/deficiency , ADAM17 Protein/genetics , Acute Disease , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cell Proliferation , Chronic Disease , Inflammation/pathology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Neutrophils/pathology , Peritonitis/metabolism , Peritonitis/pathology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Solubility
4.
Methods Mol Biol ; 1731: 57-64, 2018.
Article in English | MEDLINE | ID: mdl-29318543

ABSTRACT

Rhomboids are intramembrane serine proteases that cleave their substrates within or immediately adjacent to their transmembrane domains, a process known as regulated intramembrane proteolysis. In eukaryotes, two main types of rhomboid proteases can be distinguished based on their subcellular localization: mitochondrial rhomboids and secretase-type rhomboids that target the secretory pathway. The latter class can cleave and release the extracellular domain of all epidermal growth factor-like proteins in Drosophila and can liberate epidermal growth factor (EGF) in mammals, in a process known as ectodomain shedding. These released EGFs can then activate the EGF receptor (EGFR). EGFR signaling is crucial for mammalian development and is often deregulated in human cancer. Here we describe a cell-based protocol for detecting the ability of rhomboid proteases to release EGFR ligands into the medium. First, cells are transfected with the corresponding protease- and substrate-expressing vectors; second, cells condition the medium and accumulate shed protein. After this, protein lysates from cells and media are prepared and Western blotting is performed to detect the EGFR ligands that have been released into the medium.


Subject(s)
ErbB Receptors/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Epidermal Growth Factor/metabolism , Ligands , Mitochondria/metabolism , Protein Domains , Substrate Specificity
5.
Oncotarget ; 8(8): 13716-13729, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28099917

ABSTRACT

Fibulin-2 participates in the assembly of extracellular matrix components through interactions with multiple ligands and promotes contacts between cells and their surrounding environment. Consequently, identification of processes that could lead to an altered Fibulin-2 could have a major impact not only in the maintenance of tissue architecture and morphogenesis but also in pathological situations including cancer. Herein, we have investigated the ability of the secreted metalloproteases ADAMTS-4 and ADAMTS-5 to digest Fibulin-2. Using in vitro approaches and cultured breast cancer cell lines we demonstrate that Fibulin-2 is a better substrate for ADAMTS-5 than it is for ADAMTS-4. Moreover, Fibulin-2 degradation is associated to an enhancement of the invasive potential of T47D, MCF-7 and SK-BR-3 cells. We have also found that conditioned medium from MCF-7 cells that simultaneously overexpress Fibulin-2 and ADAMTS-5 significantly induced the migratory and invasive ability of normal breast fibroblasts using 3D collagen matrices. Immunohistochemical analysis highlights the close proximity or partial overlap of both Fibulin-2 and ADAMTS-5 in breast tumor samples. Additionally, proteolytic products derived from a potential degradation of Fibulin-2 by ADAMTS-5 were also identified in these samples. Finally, we also show that the cleavage of Fibulin-2 by ADAMTS-5 is counteracted by ADAMTS-12, a metalloprotease that interacts with Fibulin-2. Overall, our results provide direct evidence indicating that Fibulin-2 is a novel substrate of ADAMTS-5 and that this proteolysis could alter the cellular microenvironment affecting the balance between protumor and antitumor effects associated to both Fibulin-2 and the ADAMTSs metalloproteases.


Subject(s)
ADAMTS4 Protein/metabolism , ADAMTS5 Protein/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Breast Neoplasms/enzymology , Carcinogenesis , Cell Line, Tumor , Female , Fibroblasts/pathology , Humans , MCF-7 Cells , Spheroids, Cellular , Transfection , Tumor Microenvironment
6.
Nat Commun ; 8: 14174, 2017 01 27.
Article in English | MEDLINE | ID: mdl-28128203

ABSTRACT

Keratin 16 (K16) is a cytoskeletal scaffolding protein highly expressed at pressure-bearing sites of the mammalian footpad. It can be induced in hyperproliferative states such as wound healing, inflammation and cancer. Here we show that the inactive rhomboid protease RHBDF2 (iRHOM2) regulates thickening of the footpad epidermis through its interaction with K16. K16 expression is absent in the thinned footpads of irhom2-/- mice compared with irhom2+/+mice, due to reduced keratinocyte proliferation. Gain-of-function mutations in iRHOM2 underlie Tylosis with oesophageal cancer (TOC), characterized by palmoplantar thickening, upregulate K16 with robust downregulation of its type II keratin binding partner, K6. By orchestrating the remodelling and turnover of K16, and uncoupling it from K6, iRHOM2 regulates the epithelial response to physical stress. These findings contribute to our understanding of the molecular mechanisms underlying hyperproliferation of the palmoplantar epidermis in both physiological and disease states, and how this 'stress' keratin is regulated.


Subject(s)
Carrier Proteins/metabolism , Epidermis/physiology , Keratin-16/metabolism , Animals , Carrier Proteins/genetics , Cell Line , Cell Proliferation/physiology , Cytoskeleton/physiology , Down-Regulation , Epidermal Cells , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Fibroblasts , Gain of Function Mutation , Humans , Intracellular Signaling Peptides and Proteins , Keratin-6/metabolism , Keratinocytes/physiology , Keratoderma, Palmoplantar/genetics , Keratoderma, Palmoplantar/pathology , Male , Mice , Mice, Knockout , Pressure , RNA, Small Interfering/metabolism , Stress, Physiological/physiology , Tissue Culture Techniques , Up-Regulation , Wound Healing/physiology
7.
Oncotarget ; 5(5): 1253-64, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24457941

ABSTRACT

Balance between pro-tumor and anti-tumor effects may be affected by molecular interactions within tumor microenvironment. On this basis we searched for molecular partners of ADAMTS-12, a secreted metalloprotease that shows both oncogenic and tumor-suppressive effects. Using its spacer region as a bait in a yeast two-hybrid screen, we identified fibulin-2 as a potential ADAMTS-12-interacting protein. Fibulins are components of basement membranes and elastic matrix fibers in connective tissue. Besides this structural function, fibulins also play crucial roles in different biological events, including tumorigenesis. To examine the functional consequences of the ADAMTS-12/fibulin-2 interaction, we performed different in vitro assays using two breast cancer cell lines: the poorly invasive MCF-7 and the highly invasive MDA-MB-231. Overall our data indicate that this interaction promotes anti-tumor effects in breast cancer cells. To assess the in vivo relevance of this interaction, we induced tumors in nude mice using MCF-7 cells expressing both ADAMTS-12 and fibulin-2 that showed a remarkable growth deficiency. Additionally, we also found that ADAMTS-12 may elicit pro-tumor effects in the absence of fibulin-2. Immunohistochemical staining of breast cancer samples allowed the detection of both ADAMTS-12 and fibulin-2 in the connective tissue surrounding tumor area in less aggressive carcinomas. However, both proteins are hardly detected in more aggressive tumors. These data and survival analysis plots of breast cancer patients suggest that concomitant detection of ADAMTS-12 and fibulin-2 could be a good prognosis marker in breast cancer diagnosis.


Subject(s)
ADAM Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , ADAM Proteins/analysis , ADAM Proteins/genetics , ADAMTS Proteins , Animals , Breast Neoplasms/chemistry , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/genetics , Cell Movement , Cell Proliferation , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/analysis , Extracellular Matrix Proteins/genetics , Female , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Mice , Neoplasm Invasiveness , Prognosis , Spheroids, Cellular , Tumor Burden , Tumor Microenvironment
8.
Cancer Res ; 73(1): 395-405, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23108143

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF/CSF2) is a cytokine produced in the hematologic compartment that may enhance antitumor immune responses, mainly by activation of dendritic cells. Here, we show that more than one-third of human colorectal tumors exhibit aberrant DNA demethylation of the GM-CSF promoter and overexpress the cytokine. Mouse engraftment experiments with autologous and homologous colon tumors engineered to repress the ectopic secretion of GM-CSF revealed the tumor-secreted GM-CSF to have an immune-associated antitumor effect. Unexpectedly, an immune-independent antitumor effect was observed that depended on the ectopic expression of GM-CSF receptor subunits by tumors. Cancer cells expressing GM-CSF and its receptor did not develop into tumors when autografted into immunocompetent mice. Similarly, 100% of the patients with human colon tumors that overexpressed GM-CSF and its receptor subunits survived at least 5 years after diagnosis. These data suggest that expression of GM-CSF and its receptor subunits by colon tumors may be a useful marker for prognosis as well as for patient stratification in cancer immunotherapy.


Subject(s)
Colorectal Neoplasms/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Animals , Biomarkers, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , DNA Methylation , Enzyme-Linked Immunosorbent Assay , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Prognosis , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Tissue Array Analysis , Transfection
9.
Exp Hematol ; 41(2): 172-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23085464

ABSTRACT

Matrix metalloprotease-9 (MMP9) plays a critical role in acute myeloid leukemia (AML) by increasing the invasive properties of malignant myeloblasts. The role of this enzyme in high-risk myelodysplastic diseases (MDS) and the effect of azacitidine on its expression in MDS and AML have not been studied in detail. In this work, we have analyzed the effect of different concentrations of azacitidine in two well-established, MDS-derived, acute myeloid leukemic cell lines: MOLM-13 and SKM-1. We have demonstrated that 1 µmol/L azacitidine decreases MMP9 DNA methylation levels and that this is correlated with a significant increase in messenger RNA expression in both cell lines. Surprisingly, changes in protein levels were minor. This paradoxic effect is explained by the drug-dependent induction of apoptosis that reduces the amount of active secreting cells. A balance between induced expression and apoptosis was established at an azacitidine concentration of 0.2 µmol/L in MOLM-13 cells. This dose significantly increased the invasive capacity of viable cells, as measured in the Matrigel assay. To evaluate the clinical relevance of this observation, we have examined the effect of azacitidine on MMP9 expression in bone marrow from five patients with MDS, with the finding that this drug significantly increased MMP9 protein levels in all analyzed patients after six cycles of treatment. Based on these results, we conclude that azacitidine increases MMP9 expression and may enhance invasiveness in vitro. Because all five patients relapsed, these findings might explain, at least partially, the clinical failure of the drug and the progression to a more aggressive disease.


Subject(s)
Anemia, Refractory, with Excess of Blasts/drug therapy , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites/therapeutic use , Azacitidine/pharmacology , Leukemia, Monocytic, Acute/pathology , Leukemia, Myelomonocytic, Acute/pathology , Matrix Metalloproteinase 9/biosynthesis , Neoplasm Proteins/biosynthesis , Aged , Aged, 80 and over , Anemia, Refractory, with Excess of Blasts/enzymology , Anemia, Refractory, with Excess of Blasts/pathology , Antimetabolites/adverse effects , Apoptosis/drug effects , Azacitidine/adverse effects , Azacitidine/therapeutic use , Catalysis/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , DNA Methylation/drug effects , Disease Progression , Drug Resistance, Neoplasm , Enzyme Induction/drug effects , Female , Humans , Male , Matrix Metalloproteinase 9/genetics , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasms, Second Primary/drug therapy , Neoplasms, Second Primary/enzymology , Neoplasms, Second Primary/pathology , Promoter Regions, Genetic/drug effects , Risk
10.
J Biol Chem ; 287(47): 39554-63, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23019333

ABSTRACT

ADAMTSs (a disintegrin and metalloprotease with thrombospondin domains) are a family of enzymes with both proteolytic and protein interaction functions, which have been implicated in distinct pathologies. In this work, we have investigated the putative role of ADAMTS-12 in inflammation by using a mouse model deficient in this metalloprotease. Control and mutant mice were subjected to different experimental conditions to induce colitis, endotoxic sepsis, and pancreatitis. We have observed that Adamts12-deficient mice exhibit more severe inflammation and a delayed recovery from these challenges compared with their wild-type littermates. These changes are accompanied by an increase in inflammatory markers including several cytokines, as assessed by microarray expression analysis and proteomic-based approaches. Interestingly, the clinical symptoms observed in Adamts12-deficient mice are also concomitant with an elevation in the number of neutrophils in affected tissues. Finally, isolation and in vitro culture of human neutrophils demonstrate that the presence of ADAMTS-12 induces neutrophil apoptosis. On the basis of these results, we propose that ADAMTS-12 is implicated in the inflammatory response by modulating normal neutrophil apoptosis.


Subject(s)
ADAM Proteins/metabolism , Colitis/enzymology , Endotoxemia/enzymology , Neutrophils/enzymology , Pancreatitis/enzymology , ADAM Proteins/genetics , ADAM Proteins/immunology , ADAMTS Proteins , Animals , Apoptosis/genetics , Apoptosis/immunology , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Endotoxemia/genetics , Endotoxemia/immunology , Endotoxemia/metabolism , Endotoxemia/pathology , Gene Expression Profiling , Gene Expression Regulation/immunology , Humans , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Neutrophils/immunology , Neutrophils/pathology , Oligonucleotide Array Sequence Analysis , Pancreatitis/genetics , Pancreatitis/immunology , Pancreatitis/pathology
11.
Cancer Lett ; 325(2): 132-8, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-22781395

ABSTRACT

The human fibulin family consists of seven complex extracellular glycoproteins originally characterized as components of elastic fibers in connective tissue. However, beyond its structural role, fibulins are involved in complex biological processes such as cell adhesion, migration or proliferation. Indeed, they have proved to be essential elements in normal physiology, as shown by mouse models lacking these proteins, that evidence several developmental abnormalities and pathological features. Their relevance is also apparent in tumorigenesis, an aspect that has started to be intensely studied. Distinct fibulins are expressed in both tumor and stromal cells and are subjected to multiple expression regulations with either anti or pro-tumor effects. The mechanistic insights that underlie these observations are now commencing to emerge, portraying these proteins as very versatile and active constituents of connective tissue. The aim of this review is to highlight the most relevant connections between fibulins and cancer.


Subject(s)
Calcium-Binding Proteins/physiology , Cell Transformation, Neoplastic , ADAM Proteins/physiology , ADAMTS1 Protein , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Elastic Tissue/metabolism , Elastic Tissue/ultrastructure , Elastin/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Gene Silencing , Humans , Mice , Mice, Knockout , Microfibrils/metabolism , Microfibrils/ultrastructure , Neoplasm Proteins/physiology , Neoplasms/metabolism , Neoplasms/ultrastructure , Oligopeptides , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology
12.
Hum Mutat ; 32(6): E2148-75, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21618342

ABSTRACT

We performed a mutational analysis of the 19 disintegrin-metalloproteinases (ADAMs) genes in human cutaneous metastatic melanoma and identified eight to be somatically mutated in 79 samples, affecting 34% of the melanoma tumors analyzed. Functional analysis of the two frequently mutated ADAM genes, ADAM29 and ADAM7 demonstrated that the mutations affect adhesion of melanoma cells to specific extracellular matrix proteins and in some cases increase their migration ability. This suggests that mutated ADAM genes could play a role in melanoma progression.


Subject(s)
ADAM Proteins/genetics , Melanoma/genetics , Membrane Glycoproteins/genetics , Skin Diseases/genetics , Adolescent , Adult , Cell Adhesion/genetics , Cell Movement/genetics , DNA Mutational Analysis , Extracellular Matrix Proteins/genetics , Female , Humans , Male , Melanoma/secondary , Middle Aged , Mutation/genetics , Neoplasm Metastasis , Skin Diseases/pathology
13.
J Mol Med (Berl) ; 89(6): 611-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21365186

ABSTRACT

A disintegrin and metalloprotease with thrombospondin domains (ADAMTS)-4 (aggrecanase-1) and ADAMTS-5 (aggrecanase-2) are metalloproteases involved in articular cartilage degradation and represent potential therapeutic targets in arthritis treatment. We explore herein the ability of different natural compounds to specifically block the destructive action of these enzymes. Following a preliminary screening using carboxymethylated transferrin as substrate, we focused our interest on luteolin due to its inhibitory effect on ADAMTS-4 and ADAMTS-5 activities using aggrecan and fluorogenic peptides as substrates. However, matrix metalloproteinases (MMPs) activities on these substrates result less affected by this flavonoid. Moreover, incubation of mouse chondrogenic ATDC5 cells in the presence of luteolin clearly decreases the release of aggrecan fragments mediated by aggrecanases under the same conditions in which aggrecanolysis mediated by MMPs is detected. Additionally, glycosaminoglycan levels in culture medium of murine cartilage explants stimulated with interleukin-1-alpha plus retinoic acid are reduced by the presence of the flavonoid. This inhibition takes place through blockade of ADAMTS-mediated aggrecanolysis, while MMPs activity is not or poorly affected. These results suggest that luteolin could be employed as a prototypic modifying disease-agent to create new chondroprotective compounds aimed to specifically block the unwanted aggrecanase activities in arthritic diseases.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Chondrocytes/drug effects , Luteolin/pharmacology , Procollagen N-Endopeptidase/antagonists & inhibitors , ADAMTS4 Protein , ADAMTS5 Protein , Aggrecans/metabolism , Animals , Arthritis/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Endopeptidases/metabolism , Extracellular Matrix Proteins/metabolism , Glycosaminoglycans/analysis , Interleukin-1alpha/pharmacology , Luteolin/metabolism , Matrix Metalloproteinases/metabolism , Metalloproteases/chemistry , Metalloproteases/metabolism , Mice , Tretinoin/pharmacology
14.
Mol Cancer ; 9: 170, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20591152

ABSTRACT

BACKGROUND: Wnt factors control cell differentiation through semi-independent molecular cascades known as the beta-catenin-dependent (canonical) and -independent (non-canonical) Wnt signalling pathways. Genetic and epigenetic alteration of components of the canonical Wnt signalling pathway is one of the primary mechanisms underlying colon cancer. Despite increasing evidence of the role of the non-canonical pathways in tumourigenesis, however, the underlying molecular mechanisms are poorly understood. RESULTS: Here we report that the receptor tyrosine kinase-like orphan receptor 2 (ROR2), a transmembrane receptor for Wnt factors that activates non-canonical pathways, is frequently repressed by aberrant promoter hypermethylation in human colon cancer cell lines and primary tumours. By restoring ROR2 activity in colon cancer cells harbouring ROR2 promoter hypermethylation, we show that the role of ROR2 in colon cancer cells is mediated, at least in part, by canonical Wnt and that its epigenetic-dependent loss can be pro-tumourigenic. CONCLUSIONS: Our data show the importance of epigenetic alterations of ROR2 in colon cancer, highlighting the close interconnection between canonical and non-canonical Wnt signalling pathways in this type of tumour.


Subject(s)
Colonic Neoplasms/genetics , Epigenesis, Genetic , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Wnt Proteins/physiology , Colonic Neoplasms/pathology , DNA Methylation , Humans , Promoter Regions, Genetic
15.
J Cell Sci ; 122(Pt 16): 2906-13, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19638407

ABSTRACT

Proteases have long been associated with tumor progression, given their ability to degrade extracellular matrix components and facilitate invasion and metastasis. However, recent findings indicate that different proteases can also act as tumor-suppressor enzymes. We have recently reported that lung carcinoma cells expressing the ADAMTS-12 metalloprotease show a remarkable impairment of growth in immunodeficient mice as compared with parental cells. Here, we show that ADAMTS12 promoter is hypermethylated in cancer cell lines and tumor tissues. Interestingly, ADAMTS12 expression in the stromal cells surrounding epithelial malignant cells is higher than in the paired normal tissues. Moreover, the expression of this metalloprotease in colon fibroblasts co-cultured with colon cancer cell lines is higher than in those cultured alone. Furthermore, the expression of ADAMTS-12 by these fibroblasts is linked with an anti-proliferative effect on tumor cells. Based on these findings, we hypothesize that ADAMTS-12 is a novel anti-tumor protease that can reduce the proliferative properties of tumor cells. This function is lost by epigenetic silencing in tumor cells, but concurrently induced in stromal cells, probably as part of a response of the normal tissue aimed at controlling the progression of cancer.


Subject(s)
ADAM Proteins/genetics , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Gene Silencing , Stromal Cells/enzymology , Transcriptional Activation/genetics , ADAM Proteins/biosynthesis , ADAMTS Proteins , Cell Line, Tumor , Colonic Neoplasms/pathology , DNA Methylation , Disease Progression , Enzyme Induction , Gene Expression Regulation, Neoplastic , Humans , Promoter Regions, Genetic/genetics , Stromal Cells/pathology
16.
Cancer Res ; 69(11): 4926-34, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19458070

ABSTRACT

Matrix metalloproteinases have been traditionally linked to cancer dissemination through their ability to degrade most extracellular matrix components, thus facilitating invasion and metastasis of tumor cells. However, recent functional studies have revealed that some metalloproteases, including several members of the ADAMTS family, also exhibit tumor suppressor properties. In particular, ADAMTS1, ADAMTS9, and ADAMTS18 have been found to be epigenetically silenced in malignant tumors of different sources, suggesting that they may function as tumor suppressor genes. Herein, we show that ADAMTS15 is genetically inactivated in colon cancer. We have performed a mutational analysis of the ADAMTS15 gene in human colorectal carcinomas, with the finding of four mutations in 50 primary tumors and 6 colorectal cancer cell lines. Moreover, functional in vitro and in vivo studies using HCT-116 and SW-620 colorectal cancer cells and severe combined immunodeficient mice have revealed that ADAMTS15 restrains tumor growth and invasion. Furthermore, the presence of ADAMTS15 in human colorectal cancer samples showed a negative correlation with the histopathologic differentiation grade of the corresponding tumors. Collectively, these results provide evidence that extracellular proteases, including ADAMTS15, may be targets of inactivating mutations in human cancer and further validate the concept that secreted metalloproteases may show tumor suppressor properties.


Subject(s)
ADAM Proteins/genetics , Carcinoma/genetics , Colorectal Neoplasms/genetics , Gene Silencing/physiology , ADAM Proteins/metabolism , ADAMTS Proteins , Amino Acid Sequence , Animals , Base Sequence , Caco-2 Cells , Carcinoma/metabolism , Carcinoma/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Mutational Analysis , HCT116 Cells , Humans , Mice , Mice, SCID , Models, Biological , Molecular Sequence Data , Neoplasm Invasiveness , Sequence Homology , Tumor Cells, Cultured
17.
J Cell Sci ; 120(Pt 20): 3544-52, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17895370

ABSTRACT

Members of the ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family of proteolytic enzymes are implicated in a variety of physiological processes, such as collagen maturation, organogenesis, angiogenesis, reproduction and inflammation. Moreover, deficiency or overexpression of certain ADAMTS proteins is directly involved in serious human diseases, including cancer. However, the functional roles of other family members, such as ADAMTS12, remain unknown. Here, by using different in vitro and in vivo approaches, we have evaluated the possible role of ADAMTS12 in the development and progression of cancer. First, we show that expression of ADAMTS12 in Madin-Darby canine kidney (MDCK) cells prevents the tumorigenic effects of hepatocyte growth factor (HGF) by blocking the activation of the Ras-MAPK signalling pathway and that this regulation involves the thrombospondin domains of the metalloproteinase. We also show that addition of recombinant human ADAMTS12 to bovine aortic endothelial cells (BAE-1 cells) abolishes their ability to form tubules upon stimulation with vascular endothelial growth factor (VEGF). Additionally, tumours induced in immunodeficient SCID mice injected with A549 cells overexpressing ADAMTS12 show a remarkable growth deficiency in comparison with tumours formed in animals injected with parental A549 cells. Overall, our data suggest that ADAMTS12 confers tumour-protective functions upon cells that produce this proteolytic enzyme.


Subject(s)
ADAM Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neoplasms, Experimental/pathology , ras Proteins/metabolism , ADAM Proteins/chemistry , ADAMTS Proteins , Animals , Cattle , Cell Line , Cell Line, Tumor , Dogs , Hepatocyte Growth Factor/metabolism , Humans , MAP Kinase Signaling System , Mice , Mice, SCID , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism
18.
Front Biosci ; 12: 4661-9, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17485402

ABSTRACT

The large and growing number of protease genes identified in the human genome, more than 560, reflects the complexity and relevance of these enzymes in multiple biological processes. As part of our studies on the human degradome--which is defined as the complete set of human protease genes--we have recently identified and cloned three complex polyserine proteases called polyserases. Polyserase-1 is a member of the type-II transmembrane serine protease (TTSP) family of proteolytic enzymes that undergoes a series of post-translational processing events to generate three distinct and independent serine protease domains called serase-1, -2, and -3. Polyserase-2 is a secreted enzyme that also possesses three serine protease domains, but they remain as an integral part of the initial protein product. Finally, polyserase-3 is also a secreted enzyme that contains two serine protease domains embedded in the same polypeptide chain. Despite all three human polyserases share this complex molecular design characterized by the presence of several catalytic domains in their structure, they also exhibit distinctive features including unique expression patterns and different enzymatic properties. At present, the putative functional advantages derived from the complex structural organization of polyserases remain unknown, but the widespread occurrence of these enzymes in mammalian degradomes provides additional evidence about the complexity of proteolytic systems in these organisms.


Subject(s)
Serine Endopeptidases/metabolism , Animals , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Serine Endopeptidases/chemistry
19.
BMC Biochem ; 7: 9, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16566820

ABSTRACT

BACKGROUND: We have previously described the identification and characterization of polyserase-1 and polyserase-2, two human serine proteases containing three different catalytic domains within the same polypeptide chain. Polyserase-1 shows a complex organization and it is synthesized as a membrane-bound protein which can generate three independent serine protease domains as a consequence of post-translational processing events. The two first domains are enzymatically active. By contrast, polyserase-2 is an extracellular glycosylated protein whose three protease domains remain embedded in the same chain, and only the first domain possesses catalytic activity. RESULTS: Following our interest in the study of the human degradome, we have cloned a human liver cDNA encoding polyserase-3, a new protease with tandem serine protease domains in the same polypeptide chain. Comparative analysis of polyserase-3 with the two human polyserases described to date, revealed that this novel polyprotein is more closely related to polyserase-2 than to polyserase-1. Thus, polyserase-3 is a secreted protein such as polyserase-2, but lacks additional domains like the type II transmembrane motif and the low-density lipoprotein receptor module present in the membrane-anchored polyserase-1. Moreover, analysis of post-translational mechanisms operating in polyserase-3 maturation showed that its two protease domains remain as integral parts of the same polypeptide chain. This situation is similar to that observed in polyserase-2, but distinct from polyserase-1 whose protease domains are proteolytically released from the original chain to generate independent units. Immunolocalization studies indicated that polyserase-3 is secreted as a non-glycosylated protein, thus being also distinct from polyserase-2, which is a heavily glycosylated protein. Enzymatic assays indicated that recombinant polyserase-3 degrades the alpha-chain of fibrinogen as well as pro-urokinase-type plasminogen activator (pro-uPA). Northern blot analysis showed that polyserase-3 exhibits a unique expression pattern among human polyserases, being predominantly detected in testis, liver, heart and ovary, as well as in several tumor cell lines. CONCLUSION: These findings contribute to define the growing group of human polyserine proteases composed at present by three different proteins. All of them share a complex structural design with several catalytic units in a single polypeptide but also show specific features in terms of enzymatic properties, expression patterns and post-translational maturation mechanisms.


Subject(s)
Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Amino Acid Sequence , Cloning, Molecular , Dimerization , Humans , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Serine Endopeptidases/genetics , Structural Homology, Protein , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...