Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Res ; 78(5): 1293-1307, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29282220

ABSTRACT

Inflammasomes are key regulators of innate immunity in chronic inflammatory disorders and autoimmune diseases, but their role in inflammation-associated tumorigenesis remains ill-defined. Here we reveal a protumorigenic role in gastric cancer for the key inflammasome adaptor apoptosis-related speck-like protein containing a CARD (ASC) and its effector cytokine IL18. Genetic ablation of ASC in the gp130F/F spontaneous mouse model of intestinal-type gastric cancer suppressed tumorigenesis by augmenting caspase-8-like apoptosis in the gastric epithelium, independently from effects on myeloid cells and mucosal inflammation. This phenotype was characterized by reduced activation of caspase-1 and NF-κB activation and reduced expression of mature IL18, but not IL1ß, in gastric tumors. Genetic ablation of IL18 in the same model also suppressed gastric tumorigenesis, whereas blockade of IL1ß and IL1α activity upon genetic ablation of the IL1 receptor had no effect. The specific protumorigenic role for IL18 was associated with high IL18 gene expression in the gastric tumor epithelium compared with IL1ß, which was preferentially expressed in immune cells. Supporting an epithelial-specific role for IL18, we found it to be highly secreted from human gastric cancer cell lines. Moreover, IL18 blockade either by a neutralizing anti-IL18 antibody or by CRISPR/Cas9-driven deletion of ASC augmented apoptosis in human gastric cancer cells. In clinical specimens of human gastric cancer tumors, we observed a significant positive correlation between elevated mature IL18 protein and ASC mRNA levels. Collectively, our findings reveal the ASC/IL18 signaling axis as a candidate therapeutic target in gastric cancer.Significance: Inflammasome activation that elevates IL18 helps drive gastric cancer by protecting cancer cells against apoptosis, with potential implications for new therapeutic strategies in this setting. Cancer Res; 78(5); 1293-307. ©2017 AACR.


Subject(s)
Apoptosis , CARD Signaling Adaptor Proteins/metabolism , CARD Signaling Adaptor Proteins/physiology , Cell Transformation, Neoplastic/pathology , Inflammation/pathology , Interleukin-18/metabolism , Stomach Neoplasms/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CARD Signaling Adaptor Proteins/genetics , Cell Proliferation , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cytokine Receptor gp130/physiology , Follow-Up Studies , Humans , Immunity, Innate/immunology , Inflammasomes , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-18/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Prognosis , Signal Transduction , Stomach Neoplasms/immunology , Stomach Neoplasms/metabolism , Tumor Cells, Cultured
2.
Atherosclerosis ; 238(2): 321-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25545330

ABSTRACT

OBJECTIVE: Interleukin (IL)-6 is a key modulator of the acute phase response (APR), and while both are implicated in atherosclerosis, the pathological role of specific IL-6 signalling cascades is ill-defined. Since IL-6 employs the cytokine receptor gp130 to primarily activate the STAT3 pathway, here we evaluate whether gp130-dependent STAT3 activation modulates atherosclerosis. METHODS: High-fat diet-induced atherosclerosis was established in ApoE(-/-) mice crossed with gp130(F/F) knock-in mice displaying elevated gp130-dependent STAT3 activation and production of the APR protein, serum amyloid A (SAA). Also generated were gp130(F/F):Stat3(-/+):ApoE(-/-) mice displaying genetically-normalised STAT3 activation and SAA levels, and bone marrow chimeras involving ApoE(-/-) and gp130(F/F):ApoE(-/-) mice. At 10 weeks post high-fat diet, aortic atherosclerotic lesions, including the presence of CD68(+) macrophages, and plasma lipid and SAA profiles, were assessed. RESULTS: Aortic plaque development and plasma triglyceride levels in gp130(F/F):ApoE(-/-) mice were significantly reduced (3-fold, P < 0.001) compared to ApoE(-/-) littermates. By contrast, in gp130(F/F):ApoE(-/-) mice, atherosclerotic plaques contained augmented CD68(+) macrophage infiltrates, and plasma SAA levels were elevated, compared to ApoE(-/-) mice. Atherosclerotic lesion development and plasma triglyceride levels in gp130(F/F):ApoE(-/-) and gp130(F/F):Stat3(-/+):ApoE(-/-) mice were comparable, despite a significant (P < 0.05) reduction in macrophage numbers in lesions, and also plasma SAA levels, in gp130(F/F):Stat3(-/+):ApoE(-/-) mice. Aortic plaque development and plasma triglyceride levels were comparable in ApoE(-/-) mice reconstituted with gp130(F/F):ApoE(-/-) (ApoE(F/F:ApoE)) or ApoE(-/-) (ApoE(ApoE)) bone marrow cells. CONCLUSIONS: Deregulation of gp130/STAT3 signalling augments the APR and macrophage infiltration during atherosclerosis without impacting on the development of aortic plaques.


Subject(s)
Aorta/metabolism , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Cytokine Receptor gp130/metabolism , Signal Transduction , Animals , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Chemotaxis , Cytokine Receptor gp130/genetics , Diet, High-Fat , Disease Models, Animal , Female , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mutation , Plaque, Atherosclerotic , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Serum Amyloid A Protein/metabolism , Triglycerides/blood
3.
J Immunol ; 192(9): 4425-35, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24696235

ABSTRACT

Circulating levels of a soluble type I IFNR are elevated in diseases, such as chronic inflammation, infections, and cancer, but whether it functions as an antagonist, agonist, or transporter is unknown. In this study, we elucidate the in vivo importance of the soluble type I IFNAR, soluble (s)IFNAR2a, which is generated by alternative splicing of the Ifnar2 gene. A transgenic mouse model was established to mimic the 10-15-fold elevated expression of sIFNAR2a observed in some human diseases. We generated transgenic mouse lines, designated SolOX, in which the transgene mRNA and protein-expression patterns mirrored the expression patterns of the endogenous gene. SolOX were demonstrated to be more susceptible to LPS-mediated septic shock, a disease model in which type I IFN plays a crucial role. This effect was independent of "classical" proinflammatory cytokines, such as TNF-α and IL-6, whose levels were unchanged. Because the increased levels of sIFNAR2a did not affect the kinetics of the increased interferonemia, this soluble receptor does not potentiate its ligand signaling by improving IFN pharmacokinetics. Mechanistically, increased levels of sIFNAR2a are likely to facilitate IFN signaling, as demonstrated in spleen cells overexpressing sIFNAR2a, which displayed quicker, higher, and more sustained activation of STAT1 and STAT3. Thus, the soluble IFNR is an important agonist of endogenous IFN actions in pathophysiological processes and also is likely to modulate the therapeutic efficacy of clinically administered IFNs.


Subject(s)
Interferon Type I/immunology , Receptor, Interferon alpha-beta/immunology , Shock, Septic/immunology , Signal Transduction/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Immunophenotyping , Inflammation/immunology , Inflammation/metabolism , Interferon Type I/metabolism , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Receptor, Interferon alpha-beta/metabolism , Shock, Septic/metabolism , Toll-Like Receptor 4/metabolism
4.
Cancer Cell ; 22(4): 466-78, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23079657

ABSTRACT

Gastric cancer (GC) is associated with chronic inflammation; however, the molecular mechanisms promoting tumorigenesis remain ill defined. Using a GC mouse model driven by hyperactivation of the signal transducer and activator of transcription (STAT)3 oncogene, we show that STAT3 directly upregulates the epithelial expression of the inflammatory mediator Toll-like receptor (TLR)2 in gastric tumors. Genetic and therapeutic targeting of TLR2 inhibited gastric tumorigenesis, but not inflammation, characterized by reduced proliferation and increased apoptosis of the gastric epithelium. Increased STAT3 pathway activation and TLR2 expression were also associated with poor GC patient survival. Collectively, our data reveal an unexpected role for TLR2 in the oncogenic function of STAT3 that may represent a therapeutic target in GC.


Subject(s)
Cell Transformation, Neoplastic , STAT3 Transcription Factor/physiology , Stomach Neoplasms/etiology , Toll-Like Receptor 2/physiology , Animals , Cell Proliferation , Cell Survival , Cytokine Receptor gp130/physiology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Humans , Inflammation/complications , Male , Mice , Mice, Inbred C57BL , Up-Regulation
5.
J Pathol ; 225(2): 255-64, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21710691

ABSTRACT

Chronic activation of the gastric mucosal adaptive immune response is a characteristic trait of gastric cancer. It has recently emerged that a new class of T helper (Th) cells, defined by their ability to produce interleukin (IL)-17A (Th17), is associated with a host of inflammatory responses, including gastritis. However, the role of these Th17 cells in the pathogenesis of gastric cancer is less clear. To formally address this, we employed gp130(F/F) mice, which spontaneously develop gastric inflammation-associated tumours akin to human intestinal-type gastric cancer. At the molecular level, these tumours demonstrate hyper-activation of the latent transcription factor signal transducer and activator of transcription (STAT)3 via the IL-6 cytokine family member, IL-11. In gp130(F/F) mice, the generation of Th17 cells, as well as the gastric expression of IL-17a and other Th17-related factors (Rorγt, IL-23), were augmented compared to wild-type gp130(+/+) mice. Consistent with a role for IL-6 and STAT3 in regulating IL-17A, increased Th17 generation and gastric expression of Th17-related factors in gp130(F/F) mice were reduced to wild-type levels in gp130(F/F) :Stat3(-/+) mice displaying normalized STAT3 activity, and also in gp130(F/F) :IL-6(-/-) mice. Importantly, genetic ablation of IL-17A in gp130(F/F) :IL-17a(-/-) mice did not suppress the initiation and growth of gastric tumours. Furthermore, IL-17A and RORC gene expression was strongly increased in human gastric biopsies from patients with gastritis, but not gastric cancer. Collectively, our data suggest that increased expression of Th17-related factors does not correlate with the molecular pathogenesis of gastric tumourigenesis.


Subject(s)
Cell Transformation, Neoplastic/immunology , Interleukin-17/biosynthesis , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/immunology , Animals , Cell Differentiation/immunology , Cell Separation , Cell Transformation, Neoplastic/metabolism , Flow Cytometry , Gastritis/immunology , Gastritis/metabolism , Gene Knock-In Techniques , Humans , Immunohistochemistry , Interleukin-6/immunology , Interleukin-6/metabolism , Mice , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
6.
J Immunol ; 181(3): 2189-95, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18641358

ABSTRACT

The successful resolution of inflammation is dependent upon the coordinated transition from the initial recruitment of neutrophils to a more sustained population of mononuclear cells. IL-6, which signals via the common receptor subunit gp130, represents a crucial checkpoint regulator of neutrophil trafficking during the inflammatory response by orchestrating chemokine production and leukocyte apoptosis. However, the relative contribution of specific IL-6-dependent signaling pathways to these processes remains unresolved. To define the receptor-mediated signaling events responsible for IL-6-driven neutrophil trafficking, we used a series of gp130 knockin mutant mice displaying altered IL-6-signaling capacities in an experimental model of acute peritoneal inflammation. Hyperactivation of STAT1 and STAT3 in gp130(Y757F/Y757F) mice led to a more rapid clearance of neutrophils, and this coincided with a pronounced down-modulation in production of the neutrophil-attracting chemokine CXCL1/KC. By contrast, the proportion of apoptotic neutrophils in the inflammatory infiltrate remained unaffected. In gp130(Y757F/Y757F) mice lacking IL-6, neutrophil trafficking and CXCL1/KC levels were normal, and this corresponded with a reduction in the level of STAT1/3 activity. Furthermore, monoallelic ablation of Stat3 in gp130(Y757F/Y757F) mice specifically reduced STAT3 activity and corrected both the rapid clearance of neutrophils and impaired CXCL1/KC production. Conversely, genetic deletion of Stat1 in gp130(Y757F/Y757F) mice failed to rescue the altered responses observed in gp130(Y757F/Y757F) mice. Collectively, these data genetically define that IL-6-driven signaling via STAT3, but not STAT1, limits the inflammatory recruitment of neutrophils, and therefore represents a critical event for the termination of the innate immune response.


Subject(s)
Cell Movement/immunology , Interleukin-6/immunology , Interleukin-6/metabolism , Neutrophils/cytology , Neutrophils/metabolism , STAT3 Transcription Factor/metabolism , Acute Disease , Animals , Apoptosis , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/immunology , Cytokine Receptor gp130/metabolism , Gene Expression Regulation , Genotype , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/deficiency , Interleukin-6/genetics , Mice , Mice, Transgenic , Neutrophils/immunology , Signal Transduction
7.
J Clin Invest ; 118(5): 1727-38, 2008 May.
Article in English | MEDLINE | ID: mdl-18431520

ABSTRACT

Deregulated activation of STAT3 is frequently associated with many human hematological and epithelial malignancies, including gastric cancer. While exaggerated STAT3 signaling facilitates an antiapoptotic, proangiogenic, and proproliferative environment for neoplastic cells, the molecular mechanisms leading to STAT3 hyperactivation remain poorly understood. Using the gp130(Y757F/Y757F) mouse model of gastric cancer, which carries a mutated gp130 cytokine receptor signaling subunit that cannot bind the negative regulator of cytokine signaling SOCS3 and is characterized by hyperactivation of the signaling molecules STAT1 and STAT3, we have provided genetic evidence that IL-11 promotes chronic gastric inflammation and associated tumorigenesis. Expression of IL-11 was increased in gastric tumors in gp130(Y757F/Y757F) mice, when compared with unaffected gastric tissue in wild-type mice, while gp130(Y757F/Y757F) mice lacking the IL-11 ligand-binding receptor subunit (IL-11Ralpha) showed normal gastric STAT3 activation and IL-11 expression and failed to develop gastric tumors. Furthermore, reducing STAT3 activity in gp130(Y757F/Y757F) mice, either genetically or by therapeutic administration of STAT3 antisense oligonucleotides, normalized gastric IL-11 expression and alleviated gastric tumor burden. Surprisingly, the genetic reduction of STAT1 expression also reduced gastric tumorigenesis in gp130(Y757F/Y757F) mice and coincided with reduced gastric inflammation and IL-11 expression. Collectively, our data have identified IL-11 as a crucial cytokine promoting chronic gastric inflammation and associated tumorigenesis mediated by excessive activation of STAT3 and STAT1.


Subject(s)
Cytokine Receptor gp130/immunology , Inflammation/metabolism , Interleukin-11/immunology , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/immunology , Stomach Neoplasms/metabolism , Animals , Cytokine Receptor gp130/genetics , Gastric Mucosa/metabolism , Humans , Interleukin-11/genetics , Interleukin-6/immunology , Mice , Mice, Knockout , Mice, Transgenic , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/physiology , Stomach/anatomy & histology , Stomach/pathology , Stomach Neoplasms/pathology
8.
Blood ; 109(6): 2380-8, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17082315

ABSTRACT

We have previously demonstrated that STAT3 hyperactivation via the interleukin 6 (IL-6) cytokine family receptor gp130 in gp130 (Y757F/Y757F) mice leads to numerous hematopoietic and lymphoid pathologies, including neutrophilia, thrombocytosis, splenomegaly, and lymphadenopathy. Because IL-6 and IL-11 both signal via a gp130 homodimer, we report here a genetic approach to dissect their individual roles in these pathologies. Neutrophilia and thrombocytosis were absent in gp130 (Y757F/Y757F) mice lacking either IL-6 (gp130 (Y757F/Y757F): IL-6 (-/-)) or the IL-11 receptor alpha subunit (gp130 (Y757F/Y757F): IL-11Ralpha1 (-/-)), and this was associated with a normalized bone marrow compartment. The elevated myelopoiesis and megakaryopoiesis in bone marrow of gp130 (Y757F/Y757F) mice was attributable to an increase by either IL-6 or IL-11 in the STAT3-driven impairment of transforming growth factor beta (TGF-beta) signaling, which is a suppressor of these lineages. In contrast, the absence of IL-6, but not IL-11 signaling, prevented the splenomegaly, abnormal lymphopoiesis, and STAT3 hyperactivation in lymphoid organs of gp130 (Y757F/Y757F) mice. Furthermore, hyperactivation of STAT3 in lymphoid organs was associated with increased expression of IL-6Ralpha, and IL-6Ralpha expression was reduced in gp130 (Y757F/Y757F): Stat3 (+/-) mice displaying normal levels of STAT3 activity. Collectively, these data genetically define distinct roles of IL-6 and IL-11 in driving pathologic hematopoietic and lymphoid responses mediated by STAT3 hyperactivation.


Subject(s)
Hematopoiesis , Interleukin-11/metabolism , Interleukin-6/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , STAT3 Transcription Factor/metabolism , Alleles , Animals , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Gene Expression Regulation , Interleukin-11/pharmacology , Interleukin-6/deficiency , Interleukin-6/genetics , Lymphatic Diseases/genetics , Lymphatic Diseases/metabolism , Lymphatic Diseases/pathology , Mice , Mice, Transgenic , Receptors, Interleukin-11/metabolism , Signal Transduction , Splenomegaly/genetics , Splenomegaly/metabolism , Splenomegaly/pathology , Thrombocytosis/genetics , Thrombocytosis/metabolism , Thrombocytosis/pathology , Transforming Growth Factor beta1/metabolism
9.
Nat Med ; 11(8): 845-52, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16041381

ABSTRACT

The latent transcription factor Stat3 is activated by gp130, the common receptor for the interleukin (IL)-6 cytokine family and other growth factor and cytokine receptors. Ligand-induced dimerization of gp130 leads to activation of the Stat1, Stat3 and Shp2-Ras-Erk signaling pathways. Here we assess genetically the contribution of exaggerated Stat3 activation to the phenotype of gp130 (Y757F/Y757F) mice, in which a knock-in mutation disrupts the negative feedback mechanism on gp130-dependent Stat signaling. Compared to gp130 (Y757F/Y757F) mice, reduced Stat3 activation in gp130 (Y757F/Y757F) Stat3(+/-) mice increased their lifespan, prevented splenomegaly, normalized exaggerated hepatic acute-phase response and lymphocyte trafficking, and suppressed the growth of spontaneously arising gastric adenomas in young mice. These lesions share histological features of gastric polyps in aging mice with monoallelic null mutations in Smad4, which encodes the common transducer for transforming growth factor (TGF)-beta signaling. Indeed, hyperactivation of Stat3 desensitizes gp130 (Y757F/Y757F) cells to the cytostatic effect of TGF-beta through transcriptional induction of inhibitory Smad7, thereby providing a novel link for cross-talk between Stat and Smad signaling in gastric homeostasis.


Subject(s)
Adenoma/metabolism , Cytokine Receptor gp130/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Stomach Neoplasms/metabolism , Transcriptional Activation/physiology , Transforming Growth Factor beta/metabolism , Adenoma/physiopathology , Animals , Cytokine Receptor gp130/genetics , Histological Techniques , Immunoblotting , Mice , Mice, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction , Smad7 Protein/metabolism , Stomach Neoplasms/physiopathology
10.
Blood ; 105(9): 3512-20, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15650055

ABSTRACT

The interleukin-6 (IL-6) cytokine family plays an important role in regulating cellular responses during hematopoiesis. We report here that mice homozygous for a knock-in mutation in the IL-6 cytokine family receptor signaling subunit glycoprotein (gp) 130 (gp130(Y757F/Y757F)) that leads to gp130-dependent signal transducers and activators of transcription (STAT) 1/3 hyperactivation develop a broad spectrum of hematopoietic abnormalities, including splenomegaly, lymphadenopathy, and thrombocytosis. To determine whether STAT3 hyperactivation was responsible for the perturbed hematopoiesis in gp130(Y757F/Y757F) mice, we generated gp130(Y757F/Y757F) mice on a Stat3 heterozygous (Stat3(+/-)) background to specifically reduce gp130-dependent activation of STAT3, but not STAT1. Normal hematopoiesis was observed in gp130(Y757F/Y757F):Stat3(+/-) bone marrow and spleen, with no evidence of the splenomegaly and thrombocytosis displayed by gp130(Y757F/Y757F) mice. The perturbed cellular composition of thymus and lymph nodes in gp130(Y757F/Y757F) mice was also alleviated in gp130(Y757F/Y757F): Stat3(+/-) mice. Furthermore, we show that hematopoietic cells from gp130(Y757F/Y757F) mice exhibited increased survival and proliferation in response to IL-6 family cytokines. Collectively, these data provide genetic evidence that gp130-dependent STAT3 hyperactivation during hematopoiesis has pathological consequences affecting multiple organs, and therefore identify the threshold of STAT3 signaling elicited by IL-6 family cytokines as a critical determinant for hematopoietic homeostasis.


Subject(s)
Antigens, CD/physiology , DNA-Binding Proteins/physiology , Hematopoiesis , Membrane Glycoproteins/physiology , Signal Transduction/physiology , Trans-Activators/physiology , Animals , Antigens, CD/genetics , Cell Proliferation , Cell Survival , Cells, Cultured , Cytokine Receptor gp130 , Genotype , Hematopoietic Stem Cells/cytology , Membrane Glycoproteins/genetics , Mice , Mice, Mutant Strains , Mutation, Missense , STAT3 Transcription Factor , Splenomegaly/genetics , Thrombocytosis/genetics
11.
J Cell Sci ; 117(Pt 25): 6071-84, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15546919

ABSTRACT

ARAP3 is a GTPase activating protein (GAP) for Rho and Arf GTPases that is implicated in phosphoinositide 3-kinase (PI 3-kinase) signalling pathways controlling lamellipodia formation and actin stress fibre assembly. We have identified ARAP3 as a phosphorylated target of protein tyrosine kinases. In cells, ARAP3 was tyrosine phosphorylated when co-expressed with Src-family kinases (SFKs), upon stimulation with growth factors and during adhesion to the extracellular matrix (ECM) substrate fibronectin. Adhesion-induced phosphorylation of ARAP3 was suppressed by selective inhibitors of Src-family kinases and PI 3-kinase and by a Src dominant interfering mutant. Inducible expression of ARAP3 in HEK293 epithelial cells resulted in increased cell rounding, membrane process formation and cell clustering on ECM substrates. In contrast, ARAP3 dramatically slowed the kinetics of cell spreading on fibronectin but had no effect on cell adhesion. These effects of ARAP3 required a functional Rho GAP domain and were associated with reduced cellular levels of active RhoA and Rac1 but did not require the sterile alpha motif (SAM) or Arf GAP domains. Mutation of two phosphorylation sites, Y1399 and Y1404, enhanced some ARAP3 activities, suggesting that ARAP3 may be negatively regulated by phosphorylation on these tyrosine residues. These results implicate ARAP3 in integrin-mediated tyrosine kinase signalling pathways controlling Rho GTPases and cell spreading.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , GTPase-Activating Proteins/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Adhesion , Cell Line , Cell Membrane/metabolism , Cell Movement , DNA/metabolism , DNA, Complementary/metabolism , Extracellular Matrix/metabolism , Fibronectins/metabolism , GTP Phosphohydrolases/metabolism , GTPase-Activating Proteins/metabolism , Gene Expression Regulation , Genes, Dominant , Humans , Immunoblotting , Immunoprecipitation , Kinetics , Mice , Mutation , NIH 3T3 Cells , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Signal Transduction , Time Factors , Tyrosine/chemistry , Tyrosine/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/chemistry , src-Family Kinases/metabolism
12.
J Urol ; 171(2 Pt 1): 907-10, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14713851

ABSTRACT

PURPOSE: The development of hormone refractory prostate cancer marks the onset of the terminal phase of the disease. Despite the use of traditional chemotherapeutic drugs as well as many novel agents life expectancy is not significantly increased beyond palliative care alone. Selenium is a micronutrient that is incorporated into a number of essential enzymes and a minimum intake is necessary for the maintenance of health. In the last few years evidence has accumulated from case-control and limited randomized control data that supranutritional doses of selenium could inhibit the progression of prostate cancer. While much attention has focused on its use as a chemopreventive agent, its use as specific therapy has been limited. We hypothesized that dietary supplementation of selenium would inhibit the progression of hormone refractory prostate cancer in an experimental model. MATERIALS AND METHODS: We established orthotopic PC3 tumors in the prostates of 6-week-old male nude mice and fed them a baseline selenium replete diet (0.07 ppm), supplementing intake with different forms of selenium (sodium selenate, selenomethionine, methylselenocysteine and selenized yeast) at 2 different concentrations (0.3 and 3 ppm) in drinking water. RESULTS: Inorganic selenium (sodium selenate) significantly retarded the growth of primary prostatic tumors and the development of retroperitoneal lymph node metastases, which was associated with a decrease in angiogenesis. CONCLUSIONS: High dose dietary supplementation of inorganic selenium inhibits the progression of hormone refractory prostate cancer, which is due at least in part to a decrease in angiogenesis.


Subject(s)
Neoplasms, Experimental/drug therapy , Prostatic Neoplasms/drug therapy , Sodium Selenite/therapeutic use , Animals , Cell Line, Tumor , Disease Progression , Hormones/therapeutic use , Humans , Male , Mice , Mice, Inbred BALB C , Time Factors , Treatment Failure
SELECTION OF CITATIONS
SEARCH DETAIL
...