Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Front Oncol ; 12: 1043538, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36439467

RESUMO

Integrin α6ß4 is highly expressed in triple negative breast cancer (TNBC) and drives its most aggressive traits; however, its impact on chemotherapeutic efficacy remains untested. We found that integrin α6ß4 signaling promoted sensitivity to cisplatin and carboplatin but not to other chemotherapies tested. Mechanistic investigations revealed that integrin α6ß4 stimulated the activation of ATM, p53, and 53BP1, which required the integrin ß4 signaling domain. Genetic manipulation of gene expression demonstrated that mutant p53 cooperated with integrin α6ß4 for cisplatin sensitivity and was necessary for downstream phosphorylation of 53BP1 and enhanced ATM activation. Additionally, we found that in response to cisplatin-induced DNA double strand break (DSB), integrin α6ß4 suppressed the homologous recombination (HR) activity and enhanced non-homologous end joining (NHEJ) repair activity. Finally, we discovered that integrin α6ß4 preferentially activated DNA-PK, facilitated DNA-PK-p53 and p53-53BP1 complex formation in response to cisplatin and required DNA-PK to enhance ATM, 53BP1 and p53 activation as well as cisplatin sensitivity. In summary, we discovered a novel function of integrin α6ß4 in promoting cisplatin sensitivity in TNBC through DNA damage response pathway.

2.
J Cell Sci ; 135(2)2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34897465

RESUMO

Integrin α6ß4 binds plectin to associate with vimentin; however, the biological function remains unclear. Here, we utilized various integrin ß4 mutants and CRISPR-Cas9 editing to investigate this association. Upon laminin binding, integrin α6ß4 distinctly distributed peripherally as well as centrally, proximal to the nucleus. Upon fibronectin addition, integrin α6ß4 was centrally recruited to large focal adhesions (FAs) and enhanced Fak (also known as PTK2) phosphorylation. Integrin ß4 plectin-binding mutants or genetic deletion of plectin inhibited ß4 recruitment to FAs and integrin α6ß4-enhanced cell spreading, migration and three-dimensional invasive growth. Loss of the ß4 signaling domain (but retaining plectin binding) blocked migration and invasiveness but not cell spreading, recruitment to FAs or colony growth. Immunostaining revealed that integrin α6ß4 redistributed vimentin perinuclearly, where it colocalized with plectin and FAs. Depletion of vimentin completely blocked integrin ß4-enhanced invasive growth, Fak phosphorylation and proliferation in three dimensions but not two dimensions. In summary, we demonstrate the essential roles of plectin and vimentin in promoting an invasive phenotype downstream of integrin α6ß4. This article has an associated First Person interview with the first author of the paper.


Assuntos
Integrina alfa6beta4 , Plectina , Adesão Celular , Humanos , Integrina alfa6beta4/genética , Integrina beta4/genética , Filamentos Intermediários , Plectina/genética , Vimentina/genética
3.
Bio Protoc ; 10(7): e3579, 2020 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-33659549

RESUMO

Metastasis accounts for the majority of cancer related deaths. The genetically engineered mouse (GEM) models and cell line-based subcutaneous and orthotopic mouse xenografts have been developed to study the metastatic process. By using lung cancer cell line A549 as an example, we present a modified protocol to establish the cell line-based xenograft. Our protocol ensures sufficient establishment of the mouse xenografts and allows us to monitor tumor growth and spontaneous metastasis. This protocol could be adapted to other types of established cancer cell lines or primary cancer cells to study the mechanism of metastatic process as well as to test the effect of the potential anti-cancer agents on tumor growth and metastatic capacity.

4.
J Cancer Educ ; 35(1): 205-209, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-30887313

RESUMO

In 2017, the National Cancer Institute (NCI) announced new guidelines for the Cancer Center Support Grant (CCSG) application that requires reporting on education and career development activities in a section entitled, "Cancer Research Career Enhancement and Related Activities" (CRCERA). While these cancer education and training programs previously have been a required part of the CCSG application, the new guidelines require greater rigor in conducting and reporting these endeavors. In this commentary, I give my perspective of how I have built a cancer education program, first as a program director and then as an associate director of cancer education and mentoring, the critical role leadership plays in the process, and how I have recruited contributors to this educational "stone soup."


Assuntos
Liderança , Oncologia/educação , Tutoria/métodos , Mentores/educação , Neoplasias/terapia , Pesquisadores/educação , Humanos , Neoplasias/diagnóstico
5.
Nat Commun ; 10(1): 3708, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31420553

RESUMO

Neuropilin-1 (NRP1) is an essential transmembrane receptor with a variety of cellular functions. Here, we identify two human NRP1 splice variants resulting from the skipping of exon 4 and 5, respectively, in colorectal cancer (CRC). Both NRP1 variants exhibit increased endocytosis/recycling activity and decreased levels of degradation, leading to accumulation on endosomes. This increased endocytic trafficking of the two NRP1 variants, upon HGF stimulation, is due to loss of N-glycosylation at the Asn150 or Asn261 site, respectively. Moreover, these NRP1 variants enhance interactions with the Met and ß1-integrin receptors, resulting in Met/ß1-integrin co-internalization and co-accumulation on endosomes. This provides persistent signals to activate the FAK/p130Cas pathway, thereby promoting CRC cell migration, invasion and metastasis. Blocking endocytosis or endosomal Met/ß1-integrin/FAK signaling profoundly inhibits the oncogenic effects of both NRP1 variants. These findings reveal an important role for these NRP1 splice variants in the regulation of endocytic trafficking for cancer cell dissemination.


Assuntos
Neoplasias Colorretais/genética , Endossomos/metabolismo , Neuropilina-1/genética , Processamento Alternativo/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Neoplasias Colorretais/patologia , Proteína Substrato Associada a Crk/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Glicosilação , Células HCT116 , Células HT29 , Humanos , Integrina beta1/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Nus , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Neuropilina-1/metabolismo , Transporte Proteico , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais
7.
J Biol Chem ; 294(18): 7516-7527, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-30885944

RESUMO

It is generally accepted that alterations in metabolism are critical for the metastatic process; however, the mechanisms by which these metabolic changes are controlled by the major drivers of the metastatic process remain elusive. Here, we found that S100 calcium-binding protein A4 (S100A4), a major metastasis-promoting protein, confers metabolic plasticity to drive tumor invasion and metastasis of non-small cell lung cancer cells. Investigating how S100A4 regulates metabolism, we found that S100A4 depletion decreases oxygen consumption rates, mitochondrial activity, and ATP production and also shifts cell metabolism to higher glycolytic activity. We further identified that the 49-kDa mitochondrial complex I subunit NADH dehydrogenase (ubiquinone) Fe-S protein 2 (NDUFS2) is regulated in an S100A4-dependent manner and that S100A4 and NDUFS2 exhibit co-occurrence at significant levels in various cancer types as determined by database-driven analysis of genomes in clinical samples using cBioPortal for Cancer Genomics. Importantly, we noted that S100A4 or NDUFS2 silencing inhibits mitochondrial complex I activity, reduces cellular ATP level, decreases invasive capacity in three-dimensional growth, and dramatically decreases metastasis rates as well as tumor growth in vivo Finally, we provide evidence that cells depleted in S100A4 or NDUFS2 shift their metabolism toward glycolysis by up-regulating hexokinase expression and that suppressing S100A4 signaling sensitizes lung cancer cells to glycolysis inhibition. Our findings uncover a novel S100A4 function and highlight its importance in controlling NDUFS2 expression to regulate the plasticity of mitochondrial metabolism and thereby promote the invasive and metastatic capacity in lung cancer.


Assuntos
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , NADH Desidrogenase/metabolismo , Invasividade Neoplásica , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Regulação para Cima , Trifosfato de Adenosina/biossíntese , Linhagem Celular Tumoral , Inativação Gênica , Glicólise , Humanos , NADH Desidrogenase/genética , Metástase Neoplásica , Transdução de Sinais
8.
Sci Rep ; 7(1): 6174, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28733611

RESUMO

Aberrant DNA methylation patterns are a common theme across all cancer types. Specific DNA demethylation of regulatory sequences can result in upregulation of genes that are critical for tumor development and progression. Integrin α6ß4 is highly expressed in pancreatic carcinoma and contributes to cancer progression, in part, through the specific DNA demethylation and upregulation of epidermal growth factor receptor (EGFR) ligands amphiregulin (AREG) and epiregulin (EREG). Whole genome bisulfite sequencing (WGBS) revealed that integrin α6ß4 signaling promotes an overall hypomethylated state and site specific DNA demethylation of enhancer elements within the proximal promoters of AREG and EREG. Additionally, we find that the base excision repair (BER) pathway is required to maintain expression of AREG and EREG, as blocking DNA repair molecules, TET1 GADD45A, TDG, or PARP-1 decreased gene expression. Likewise, we provide the novel finding that integrin α6ß4 confers an enhanced ability on cells to repair DNA lesions and survive insult. Therefore, while many known signaling functions mediated by integrin α6ß4 that promote invasive properties have been established, this study demonstrates that integrin α6ß4 can dramatically impact the epigenome of cancer cells, direct global DNA methylation levels toward a hypomethylated state, and impact DNA repair and subsequent cell survival.


Assuntos
Anfirregulina/genética , Epirregulina/genética , Integrina alfa6beta4/metabolismo , Neoplasias Pancreáticas/metabolismo , Regulação para Cima , Linhagem Celular Tumoral , Sobrevivência Celular , Metilação de DNA , Reparo do DNA , Elementos Facilitadores Genéticos , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neoplasias Pancreáticas/genética , Sequenciamento Completo do Genoma
10.
Hum Pathol ; 54: 174-83, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27107458

RESUMO

Lung cancer carries a poor prognosis and is the most common cause of cancer-related death worldwide. The integrin α6ß4, a laminin receptor, promotes carcinoma progression in part by cooperating with various growth factor receptors to facilitate invasion and metastasis. In carcinoma cells with mutant TP53, the integrin α6ß4 promotes cell survival. TP53 mutations and integrin α6ß4 overexpression co-occur in many aggressive malignancies. Because of the high frequency of TP53 mutations in lung squamous cell carcinoma (SCC), we sought to investigate the association of integrin ß4 expression with clinicopathologic features and survival in non-small cell lung cancer (NSCLC). We constructed a lung cancer tissue microarray and stained sections for integrin ß4 subunit expression using immunohistochemistry. We found that integrin ß4 expression is elevated in SCC compared with adenocarcinoma (P<.0001), which was confirmed in external gene expression data sets (P<.0001). We also determined that integrin ß4 overexpression associates with the presence of venous invasion (P=.0048) and with reduced overall patient survival (hazard ratio, 1.46; 95% confidence interval, 1.01-2.09; P=.0422). Elevated integrin ß4 expression was also shown to associate with reduced overall survival in lung cancer gene expression data sets (hazard ratio, 1.49; 95% confidence interval, 1.31-1.69; P<.0001). Using cBioPortal, we generated a network map demonstrating the 50 most highly altered genes neighboring ITGB4 in SCC, which included laminins, collagens, CD151, genes in the EGFR and PI3K pathways, and other known signaling partners. In conclusion, we demonstrate that integrin ß4 is overexpressed in NSCLC where it is an adverse prognostic marker.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma Pulmonar de Células não Pequenas/química , Integrina alfa6/análise , Integrina beta4/análise , Neoplasias Pulmonares/química , Veias Pulmonares/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Biologia Computacional , Mineração de Dados , Bases de Dados Genéticas , Feminino , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Imuno-Histoquímica , Integrina alfa6/genética , Integrina beta4/genética , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Prognóstico , RNA Mensageiro/genética , Fatores de Tempo , Análise Serial de Tecidos , Regulação para Cima
11.
Oncotarget ; 7(23): 34630-42, 2016 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-27127879

RESUMO

S100A4 (metastasin-1), a metastasis-associated protein and marker of the epithelial to mesenchymal transition, contributes to several hallmarks of cancer and has been implicated in the progression of several types of cancer. However, the impacts of S100A4 signaling in lung cancer progression and its potential use as a target for therapy in lung cancer have not been properly explored. Using established lung cancer cell lines, we demonstrate that S100A4 knockdown reduces cell proliferation, invasion and three-dimensional invasive growth, while overexpression of S100A4 increases invasive potential. In patient-derived tissues, S100A4 is preferentially elevated in lung adenocarcinoma. This elevation is associated with lymphovascular invasion and decreased overall survival. In addition, depletion of S100A4 by shRNA inhibits NF-κB activity and decreases TNFα-induced MMP9 expression. Furthermore, inhibition of the NF-κB/MMP9 axis decreases lung carcinoma invasive potential. Niclosamide, a reported inhibitor of S100A4, blocks expression and function of S100A4 with a reduction in proliferation, invasion and NF-κB-mediated MMP9 expression. Collectively, this study highlights the importance of the S100A4/NF-κB/MMP9 axis in lung cancer invasion and provides a rationale for targeting S100A4 to combat lung cancer.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Niclosamida/farmacologia , Proteína A4 de Ligação a Cálcio da Família S100/antagonistas & inibidores , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Células A549 , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Humanos , Neoplasias Pulmonares/patologia , Metaloproteinase 9 da Matriz/biossíntese , NF-kappa B/metabolismo , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Prognóstico , Interferência de RNA , RNA Interferente Pequeno/genética , Proteína A4 de Ligação a Cálcio da Família S100/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
J Biol Chem ; 290(45): 27228-27238, 2015 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-26381405

RESUMO

Integrin α6ß4 is up-regulated in pancreatic adenocarcinomas where it contributes to carcinoma cell invasion by altering the transcriptome. In this study, we found that integrin α6ß4 up-regulates several genes in the epidermal growth factor receptor (EGFR) pathway, including amphiregulin (AREG), epiregulin (EREG), and ectodomain cleavage protease MMP1, which is mediated by promoter demethylation and NFAT5. The correlation of these genes with integrin α6ß4 was confirmed in The Cancer Genome Atlas Pancreatic Cancer Database. Based on previous observations that integrin α6ß4 cooperates with c-Met in pancreatic cancers, we examined the impact of EGFR signaling on hepatocyte growth factor (HGF)-stimulated migration and invasion. We found that AREG and EREG were required for autocrine EGFR signaling, as knocking down either ligand inhibited HGF-mediated migration and invasion. We further determined that HGF induced secretion of AREG, which is dependent on integrin-growth factor signaling pathways, including MAPK, PI3K, and PKC. Moreover, matrix metalloproteinase activity and integrin α6ß4 signaling were required for AREG secretion. Blocking EGFR signaling with EGFR-specific antibodies or an EGFR tyrosine kinase inhibitor hindered HGF-stimulated pancreatic carcinoma cell chemotaxis and invasive growth in three-dimensional culture. Finally, we found that EGFR was phosphorylated in response to HGF stimulation that is dependent on EGFR kinase activity; however, c-Met phosphorylation in response to HGF was unaffected by EGFR signaling. Taken together, these data illustrate that integrin α6ß4 stimulates invasion by promoting autocrine EGFR signaling through transcriptional up-regulation of key EGFR family members and by facilitating HGF-stimulated EGFR ligand secretion. These signaling events, in turn, promote pancreatic carcinoma migration and invasion.


Assuntos
Receptores ErbB/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Integrina alfa6beta4/metabolismo , Anfirregulina , Linhagem Celular Tumoral , Movimento Celular , Família de Proteínas EGF/genética , Família de Proteínas EGF/metabolismo , Epirregulina/genética , Epirregulina/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Técnicas de Silenciamento de Genes , Humanos , Integrina alfa6beta4/genética , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/metabolismo , Modelos Biológicos , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Transdução de Sinais , Microambiente Tumoral , Regulação para Cima
13.
Lab Invest ; 95(9): 976-86, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26121317

RESUMO

Integrin α6ß4 is a cellular adhesion molecule that binds to laminins in the extracellular matrix and nucleates the formation of hemidesmosomes. During carcinoma progression, integrin α6ß4 is released from hemidesmosomes, where it can then signal to facilitate multiple aspects of tumor progression including sustaining proliferative signaling, tumor invasion and metastasis, evasion of apoptosis, and stimulation of angiogenesis. The integrin achieves these ends by cooperating with growth factor receptors including EGFR, ErbB-2, and c-Met to amplify downstream pathways such as PI3K, AKT, MAPK, and the Rho family small GTPases. Furthermore, it dramatically alters the transcriptome toward a more invasive phenotype by controlling promoter DNA demethylation of invasion and metastasis-associated proteins, such as S100A4 and autotaxin, and upregulates and activates key tumor-promoting transcription factors such as the NFATs and NF-κB. Expression of integrin α6ß4 has been studied in many human malignancies where its overexpression is associated with aggressive behavior and a poor prognosis. This review provides an assessment of integrin α6ß4 expression patterns and their prognostic significance in human malignancies, and describes key signaling functions of integrin α6ß4 that contribute to tumor progression.


Assuntos
Carcinoma/metabolismo , Carcinoma/fisiopatologia , Metilação de DNA/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Integrina alfa6beta4/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Transdução de Sinais/fisiologia , Hemidesmossomos/metabolismo , Humanos
14.
PLoS One ; 9(2): e89892, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24587105

RESUMO

Dysregulation of Ras and Rho family small GTPases drives the invasion and metastasis of multiple cancers. For their biological functions, these GTPases require proper subcellular localization to cellular membranes, which is regulated by a series of post-translational modifications that result in either farnesylation or geranylgeranylation of the C-terminal CAAX motif. This concept provided the rationale for targeting farnesyltransferase (FTase) and geranylgeranyltransferases (GGTase) for cancer treatment. However, the resulting prenyl transferase inhibitors have not performed well in the clinic due to issues with alternative prenylation and toxicity. As an alternative, we have developed a unique class of potential anti-cancer therapeutics called Prenyl Function Inhibitors (PFIs), which are farnesol or geranyl-geraniol analogs that act as alternate substrates for FTase or GGTase. Here, we test the ability of our lead PFIs, anilinogeraniol (AGOH) and anilinofarnesol (AFOH), to block the invasion of breast cancer cells. We found that AGOH treatment effectively decreased invasion of MDA-MB-231 cells in a two-dimensional (2D) invasion assay at 100 µM while it blocked invasive growth in three-dimensional (3D) culture model at as little as 20 µM. Notably, the effect of AGOH on 3D invasive growth was phenocopied by electroporation of cells with C3 exotransferase. To determine if RhoA and RhoC were direct targets of AGOH, we performed Rho activity assays in MDA-MB-231 and MDA-MB-468 cells and found that AGOH blocked RhoA and RhoC activation in response to LPA and EGF stimulation. Notably, the geranylgeraniol analog AFOH was more potent than AGOH in inhibiting RhoA and RhoC activation and invasive growth. Interestingly, neither AGOH nor AFOH impacted 3D growth of MCF10A cells. Collectively, this study demonstrates that AGOH and AFOH dramatically inhibit breast cancer invasion, at least in part by blocking Rho function, thus, suggesting that targeting prenylation by using PFIs may offer a promising mechanism for treatment of invasive breast cancer.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Invasividade Neoplásica/prevenção & controle , Prenilação de Proteína/efeitos dos fármacos , Terpenos/farmacologia , Proteínas rho de Ligação ao GTP/metabolismo , Monoterpenos Acíclicos , Antineoplásicos/metabolismo , Técnicas de Cultura de Células , Farneseno Álcool/farmacologia , Feminino , Imunofluorescência , Humanos , Immunoblotting , Estrutura Molecular , Terpenos/metabolismo
15.
Carcinogenesis ; 35(6): 1341-51, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24510238

RESUMO

Upregulation of fatty acid synthase (FASN), a key enzyme of de novo lipogenesis, is associated with metastasis in colorectal cancer (CRC). However, the mechanisms of regulation are unknown. Since angiogenesis is crucial for metastasis, we investigated the role of FASN in the neovascularization of CRC. The effect of FASN on tumor vasculature was studied in orthotopic CRCs, the chick embryo chorioallantoic membrane (CAM) and Matrigel plug models using immunohistochemistry, immunofluorescent staining and confocal microscopy. Cell secretion was evaluated by ELISA and antibody arrays. Proliferation, migration and tubulogenesis of endothelial cells (ECs) were assessed in CRC-EC coculture models. In this study, we found that stable knockdown of FASN decreased microvessel density in HT29 and HCT116 orthotopic CRCs and resulted in 'normalization' of tumor vasculature in both orthotopic and CAM models. Furthermore, FASN regulated secretion of pro- and antiangiogenic factors, including vascular endothelial growth factor-A (VEGF-A). Mechanisms associated with the antiangiogenic activity noted with knockdown of FASN included: downregulation of VEGF(189), upregulation of antiangiogenic isoform VEGF(165b) and a decrease in expression and activity of matrix metalloproteinase-9. Furthermore, conditioned medium from FASN knockdown CRC cells inhibited activation of vascular endothelial growth factor receptor-2 and its downstream signaling and decreased proliferation, migration and tubulogenesis of ECs as compared with control medium. Together, these results suggest that cancer cell-associated FASN regulates tumor vasculature through alteration of the profile of secreted angiogenic factors and regulation of their bioavailability. Inhibition of FASN upstream of VEGF-A and other angiogenic pathways can be a novel therapeutic strategy to prevent or inhibit metastasis in CRC.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Células Endoteliais/metabolismo , Ácido Graxo Sintases/genética , Neovascularização Patológica/genética , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Modelos Animais de Doenças , Ácido Graxo Sintases/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Patológica/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
16.
BMC Cancer ; 13: 501, 2013 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-24160245

RESUMO

BACKGROUND: Various pathways impinge on the actin-myosin pathway to facilitate cell migration and invasion including members of the Rho family of small GTPases and MAPK. However, the signaling components that are considered important for these processes vary substantially within the literature with certain pathways being favored. These distinctions in signaling pathways utilized are often attributed to differences in cell type or physiological conditions; however, these attributes have not been systematically assessed. METHODS: To address this question, we analyzed the migration and invasion of MDA-MB-231 breast carcinoma cell line in response to various stimuli including lysophosphatidic acid (LPA), hepatocyte growth factor (HGF) and epidermal growth factor (EGF) and determined the involvement of select signaling pathways that impact myosin light chain phosphorylation. RESULTS: LPA, a potent stimulator of the Rho-ROCK pathway, surprisingly did not require the Rho-ROCK pathway to stimulate migration but instead utilized Rac and MAPK. In contrast, LPA-stimulated invasion required Rho, Rac, and MAPK. Of these three major pathways, EGF-stimulated MDA-MB-231 migration and invasion required Rho; however, Rac was essential only for invasion and MAPK was dispensable for migration. HGF signaling, interestingly, utilized the same pathways for migration and invasion, requiring Rho but not Rac signaling. Notably, the dependency of HGF-stimulated migration and invasion as well as EGF-stimulated invasion on MAPK was subject to the inhibitors used. As expected, myosin light chain kinase (MLCK), a convergence point for MAPK and Rho family GTPase signaling, was required for all six conditions. CONCLUSIONS: These observations suggest that, while multiple signaling pathways contribute to cancer cell motility, not all pathways operate under all conditions. Thus, our study highlights the plasticity of cancer cells to adapt to multiple migratory cues.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Movimento Celular , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Lisofosfolipídeos/metabolismo , Transdução de Sinais , Actinas/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Miosinas/metabolismo , Invasividade Neoplásica , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
17.
Oncotarget ; 3(12): 1576-87, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23211542

RESUMO

Neuroblastomas express increased levels of gastrin-releasing peptide receptor (GRP-R). However, the exact molecular mechanisms involved in GRP-R-mediated cell signaling in neuroblastoma growth and metastasis are unknown. Here, we report that focal adhesion kinase (FAK), as a critical downstream target of GRP-R, is an important regulator of neuroblastoma tumorigenicity. We found that FAK expression correlates with GRP-R expression in human neuroblastoma sections and cell lines. GRP-R overexpression in SK-N-SH cells increased FAK, integrin α3 and ß1 expressions and cell migration. These cells demonstrated flatter cell morphology with broad lamellae, in which intense FAK expression was localized to the leading edges of lamellipodia. Interestingly, FAK activation was, in part, dependent on integrin α3 and ß1 expression. Conversely, GRP-R silencing decreased FAK as well as Mycn levels in BE(2)-C cells, which displayed a denser cellular morphology. Importantly, rescue experiments in GRP-R silenced BE(2)-C cells showed FAK overexpression significantly enhanced cell viability and soft agar colony formation; similarly, FAK overexpression in SK-N-SH cells also resulted in increased cell growth. These effects were reversed in FAK silenced BE(2)-C cells in vitro as well as in vivo. Moreover, we evaluated the effect of FAK inhibition in vivo. FAK inhibitor (Y15) suppressed GRP-induced neuroblastoma growth and metastasis. Our results indicate that FAK is a critical downstream regulator of GRP-R, which mediates tumorigenesis and metastasis in neuroblastoma.


Assuntos
Movimento Celular , Quinase 1 de Adesão Focal/metabolismo , Integrina alfa3/metabolismo , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/secundário , Neuroblastoma/enzimologia , Neuroblastoma/secundário , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Forma Celular , Ativação Enzimática , Quinase 1 de Adesão Focal/antagonistas & inibidores , Quinase 1 de Adesão Focal/genética , Humanos , Integrina beta1/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Masculino , Camundongos , Camundongos Nus , Proteína Proto-Oncogênica N-Myc , Invasividade Neoplásica , Neuroblastoma/tratamento farmacológico , Neuroblastoma/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Pseudópodes/enzimologia , Interferência de RNA , Receptores da Bombesina/genética , Receptores da Bombesina/metabolismo , Transdução de Sinais , Fatores de Tempo , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Am J Physiol Cell Physiol ; 302(3): C605-14, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22049212

RESUMO

The α(6)ß(4) integrin promotes carcinoma invasion through its ability to promote directed migration and polarization of carcinoma cells. In this study, we explore how the α(6)ß(4) integrin cooperates with lysophosphatidic acid (LPA) to activate Rho and Rac small GTPases. Through the use of dominant negative Rho constructs, C3 exotransferase, and Rho kinase inhibitor, we find that Rho is critical for LPA-dependent chemotaxis and lamellae formation. However, utilization of specific Rho isoforms depends on integrin α(6)ß(4) expression status. Integrin α(6)ß(4)-negative MDA-MB-435 cells utilize only RhoC for motility, whereas integrin α(6)ß(4)-expressing cells utilize RhoC but additionally activate and utilize RhoA for LPA-dependent cell motility and lamellae formation. Notably, the activation of RhoA by cooperative LPA and integrin α(6)ß(4) signaling requires the Rho guanine nucleotide exchange factor AKAP-Lbc. We also determine that integrin α(6)ß(4) cannot activate Rac1 directly but promotes LPA-mediated Rac1 activation that is dependent on RhoA activity and de novo ß(1) integrin ligation. Finally, we find that the regulation of Rac1 and RhoA in response to LPA is differentially regulated by phosphodiesterases, PKA, and phosphatidylinositol 3-kinase, thus supporting their spatially distinct compartmentalization. In summary, signaling from integrin α(6)ß(4) facilitates LPA-stimulated chemotaxis through preferential activation of RhoA, which, in turn, facilitates activation of Rac1.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Integrina alfa6beta4/metabolismo , Lisofosfolipídeos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Linhagem Celular Tumoral , Quimiotaxia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Integrina alfa6beta4/biossíntese , Fosfatidilinositol 3-Quinases , Isoformas de Proteínas , Interferência de RNA , RNA Interferente Pequeno , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais , Quinases Associadas a rho/antagonistas & inibidores
19.
Cancer Res ; 71(9): 3246-56, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21430067

RESUMO

Activation of phosphoinositide 3-kinase (PI3K)/Akt signaling is associated with growth and progression of colorectal cancer (CRC). We have previously shown that the mTOR kinase, a downstream effector of PI3K/Akt signaling, regulates tumorigenesis of CRC. However, the contribution of mTOR and its interaction partners toward regulating CRC progression and metastasis remains poorly understood. We found that increased expression of mTOR, Raptor, and Rictor mRNA was noted with advanced stages of CRC, suggesting that mTOR signaling may be associated with CRC progression and metastasis. mTOR, Raptor, and Rictor protein levels were also significantly elevated in primary CRCs (stage IV) and their matched distant metastases compared with normal colon. Inhibition of mTOR signaling, using rapamycin or stable inhibition of mTORC1 (Raptor) and mTORC2 (Rictor), attenuated migration and invasion of CRCs. Furthermore, knockdown of mTORC1 and mTORC2 induced a mesenchymal-epithelial transition (MET) and enhanced chemosensitivity of CRCs to oxaliplatin. We observed increased cell-cell contact and decreased actin cytoskeletal remodeling concomitant with decreased activation of the small GTPases, RhoA and Rac1, upon inhibition of both mTORC1 and mTORC2. Finally, establishment of CRC metastasis in vivo was completely abolished with targeted inhibition of mTORC1 and mTORC2 irrespective of the site of colonization. Our findings support a role for elevated mTORC1 and mTORC2 activity in regulating epithelial-mesenchymal transition (EMT), motility, and metastasis of CRCs via RhoA and Rac1 signaling. These findings provide the rationale for including mTOR kinase inhibitors, which inhibit both mTORC1 and mTORC2, as part of the therapeutic regimen for CRC patients.


Assuntos
Movimento Celular/fisiologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Proteínas/metabolismo , Fatores de Transcrição/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Actinas/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/enzimologia , Citoesqueleto/metabolismo , Transição Epitelial-Mesenquimal , Células HCT116 , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Metástase Neoplásica , Transdução de Sinais , Serina-Treonina Quinases TOR
20.
Am J Physiol Cell Physiol ; 300(5): C1155-63, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21289293

RESUMO

S100A4 is associated with the progression of many types of cancers as well as several nonmalignant conditions. However, how it is regulated by intracellular signaling and/or at the transcriptional level has not been extensively studied. We recently demonstrated that S100A4 is partially regulated by nuclear factor in activated T cell 5 (NFAT5) downstream of integrin α6ß4. NFAT5 is a mammalian osmotic regulator. To study the regulation of S100A4 by NFAT5 in a more readily inducible model, colon cancer cells were subjected to hyperosmotic stress. We found that S100A4 is induced in a subset of colon cancer cell lines, and the ability to induce S100A4 depends on the methylation status of S100A4. The osmotic stress response elements were identified in the first intron region of S100A4 by S100A4 luciferase reporter assays. Depletion of NFAT5 by small interfering RNA abolished S100A4 induction. Furthermore, chromatin immunoprecipitation assays showed that NFAT5 is induced to bind to the first intron region. Inhibition of Src kinase pathways reduced S100A4 induction by affecting NFAT5 transactivation and protein levels. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to study the function of S100A4 induction in colon cancer cells under the condition of hyperosmotic stress; the results suggest that S100A4 induction contributes to cell survival. In conclusion, this study demonstrates that hyperosmotic stress induces S100A4 through NFAT5, and Src and chromatin remodeling are involved. In addition, the induction of S100A4 contributes to cell survival. Given that the gastrointestinal tract is periodically exposed to hyperosmotic stress, this study may uncover a novel signaling pathway that could contribute to GI cancer progression.


Assuntos
Neoplasias do Colo/metabolismo , Proteínas S100/metabolismo , Fatores de Transcrição/metabolismo , Quinases da Família src/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Neoplasias do Colo/patologia , Humanos , Integrina alfa6beta4/metabolismo , Íntrons , Pressão Osmótica , RNA Interferente Pequeno/farmacologia , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA