Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Proc Natl Acad Sci U S A ; 108(17): 7022-7, 2011 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-21482783

RESUMO

Abl interactor 1 (Abi1) plays a critical function in actin cytoskeleton dynamics through participation in the WAVE2 complex. To gain a better understanding of the specific role of Abi1, we generated a conditional Abi1-KO mouse model and MEFs lacking Abi1 expression. Abi1-KO cells displayed defective regulation of the actin cytoskeleton, and this dysregulation was ascribed to altered activity of the WAVE2 complex. Changes in motility of Abi1-KO cells were manifested by a decreased migration rate and distance but increased directional persistence. Although these phenotypes did not correlate with peripheral ruffling, which was unaffected, Abi1-KO cells exhibited decreased dorsal ruffling. Western blotting analysis of Abi1-KO cell lysates indicated reduced levels of the WAVE complex components WAVE1 and WAVE2, Nap1, and Sra-1/PIR121. Although relative Abi2 levels were more than doubled in Abi1-KO cells, the absolute Abi2 expression in these cells amounted only to a fifth of Abi1 levels in the control cell line. This finding suggests that the presence of Abi1 is critical for the integrity and stability of WAVE complex and that Abi2 levels are not sufficiently increased to compensate fully for the loss of Abi1 in KO cells and to restore the integrity and function of the WAVE complex. The essential function of Abi1 in WAVE complexes and their regulation might explain the observed embryonic lethality of Abi1-deficient embryos, which survived until approximately embryonic day 11.5 and displayed malformations in the developing heart and brain. Cells lacking Abi1 and the conditional Abi1-KO mouse will serve as critical models for defining Abi1 function.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Encéfalo/embriologia , Proteínas do Citoesqueleto/metabolismo , Embrião de Mamíferos/embriologia , Coração/embriologia , Complexos Multiproteicos/metabolismo , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular , Movimento Celular/fisiologia , Proteínas do Citoesqueleto/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Complexos Multiproteicos/genética , Família de Proteínas da Síndrome de Wiskott-Aldrich/genética
2.
Gynecol Oncol ; 128(1): 101-106, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23103931

RESUMO

OBJECTIVE: This study aims to test the hypothesis that targeted nanoparticle delivery of DNA encoding HPV16-regulated diphtheria toxin (DT-A) will result in the death of HPV16-infected cells. MATERIALS AND METHODS: Plasmid constructs containing a HPV16 Long Control Region (LCR) DNA sequence upstream of DT-A or luciferase reporter (Luc) DNA sequences were used to formulate poly(ß-amino ester) nanoparticles. The effect on tumor growth of HPV/DT-A-nanoparticle injection directly into HPV16(+) CaSki human cervical cancer cell-derived xenografts in mice was determined. To evaluate the ability of the HPV16 LCR regulatory sequence to activate gene expression specifically in HPV16-infected cells, mice underwent bioluminescent optical imaging following intraperitoneal injection of HPV/Luc-nanoparticles. The use of Lutrol F127, a thermal-sensitive gel, to target delivery of nanoparticles and subsequent gene expression to cervical epithelial cells was evaluated in ex vivo cultures of mouse cervix and following intravaginal delivery of nanoparticle/gel in mice, as well as in ex vivo cultures of surgical LEEP samples. RESULTS: The selected HPV16 LCR regulatory sequence activates gene expression in both HPV16-infected cells and non-infected cells. However, in the cervix, it is specifically active in epithelial cells. Following exposure of cervical cells to HPV/DT-A-nanoparticles mixed with Lutrol F127 gel, DT-A is expressed and cells die. CONCLUSIONS: An HPV16 DNA sequence that targets gene expression specifically to HPV16-infected cells remains to be discovered. Topical application of a Lutrol F127 thermal gel/nanoparticle mix is illustrative of how to restrict exposure of cells to therapeutic nanoparticles, thereby allowing for targeted DNA delivery to cervical pre-cancerous lesions.


Assuntos
DNA/administração & dosagem , Toxina Diftérica/genética , Papillomavirus Humano 16/genética , Nanopartículas/administração & dosagem , Fragmentos de Peptídeos/genética , Lesões Pré-Cancerosas/terapia , Neoplasias do Colo do Útero/terapia , Animais , Feminino , Humanos , Camundongos
3.
Proc Natl Acad Sci U S A ; 106(9): 3426-30, 2009 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-19208807

RESUMO

Claudin-3 (CLDN3) is a tight junction protein that is overexpressed in 90% of ovarian tumors. Previous in vitro studies have indicated that CLDN3 overexpression promotes the migration, invasion, and survival of ovarian cancer cells. Here, we investigated the efficacy of lipidoid-formulated CLDN3 siRNA in 3 different ovarian cancer models. Intratumoral injection of lipidoid/CLDN3 siRNA into OVCAR-3 xenografts resulted in dramatic silencing of CLDN3, significant reduction in cell proliferation, reduction in tumor growth, and a significant increase in the number of apoptotic cells. Intraperitoneal injection of lipidoid-formulated CLDN3 siRNA resulted in a substantial reduction in tumor burden in MISIIR/TAg transgenic mice and mice bearing tumors derived from mouse ovarian surface epithelial cells. Ascites development was reduced in CLDN3 siRNA-treated mice, suggesting the treatment effectively suppressed metastasis. Toxicity was not observed after multiple i.p. injections. Importantly, treatment of mice with nonimmunostimulatory 2'-OMe modified CLDN3 siRNA was as effective in suppressing tumor growth as unmodifed siRNA. These results suggest that lipidoid-formulated CLDN3 siRNA has potential as a therapeutic for ovarian cancer.


Assuntos
Proteínas de Membrana/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , RNA Interferente Pequeno/genética , Animais , Linhagem Celular Tumoral , Claudina-3 , Modelos Animais de Doenças , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica/patologia , Neoplasias Ovarianas/genética , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Transgenic Res ; 20(5): 1073-86, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21222155

RESUMO

To establish a method for efficient and relatively easy isolation of a cell population containing epithelial prostate stem cells, we developed two transgenic mouse models, K5/CFP and K18/RFP. In these models, promoters of the cytokeratin 5 (Krt5) and the cytokeratin 18 (Krt18) genes regulate cyan and red fluorescent proteins (CFP and RFP), respectively. CFP and RFP reporter protein fluorescence allows for visualization of K5(+) and K18(+) epithelial cells within the cellular spatial context of the prostate gland and for their direct isolation by FACS. Using these models, it is possible to test directly the stem cell properties of prostate epithelial cell populations that are positively selected based on expression of cytoplasmic proteins, K5 and K18. After validating appropriate expression of the K5/CFP and K18/RFP transgenes in the developing and adult prostate, we demonstrate that a subset of CFP-expressing prostate cells exhibits stem cell proliferation potential and differentiation capabilities. Then, using prostate cells sorted from double transgenic mice (K5/CFP + K18/RFP), we compare RNA microarrays of sorted K5(+)K18(+) basal and K5(-)K18(+) luminal epithelial cells, and identify genes that are differentially expressed. Several genes that are over-expressed in K5(+) cells have previously been identified as potential stem cell markers. These results suggest that FACS isolation of prostate cells from these mice based on combining reporter gene fluorescence with expression of potential stem cell surface marker proteins will yield populations of cells enriched for stem cells to a degree that has not been attained by using cell surface markers alone.


Assuntos
Células Epiteliais/metabolismo , Queratina-18/genética , Queratina-18/metabolismo , Queratina-5/genética , Queratina-5/metabolismo , Próstata/metabolismo , Células-Tronco/metabolismo , Transgenes/genética , Animais , Diferenciação Celular , Proliferação de Células , Citometria de Fluxo , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Análise de Célula Única
5.
Mol Ther ; 15(7): 1306-12, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17375071

RESUMO

Poly(beta-amino ester)s are cationic degradable polymers that have significant potential as gene delivery vectors. Here we present a generalized method to modify poly(beta-amino ester)s at the chain ends to improve their delivery performance. End-chain coupling reactions were developed so that polymers could be synthesized and tested in a high-throughput manner, without the need for purification. In this way, many structural variations at the polymer terminus could be rapidly evaluated. End-modification of the terminal amine structure of a previously optimized poly(beta-amino ester), C32, significantly enhanced its in vitro transfection efficiency. In vivo, intraperitoneal (IP) gene delivery using end-modified C32 polymers resulted in expression levels over one order of magnitude higher than unmodified C32 and jet-polyethylenimine (jet-PEI) levels in several abdominal organs. The rapid end-modification strategy presented here has led to the discovery of many effective polymers for gene delivery and may be a useful method to develop and optimize cationic polymers for gene therapy.


Assuntos
Ésteres , Técnicas de Transferência de Genes , Polímeros , Aminas/química , Animais , Células COS , Chlorocebus aethiops , Ésteres/síntese química , Ésteres/química , Expressão Gênica , Genes Reporter/genética , Injeções Intraperitoneais , Injeções Intravenosas , Masculino , Camundongos , Estrutura Molecular , Polímeros/síntese química , Polímeros/química , Fatores de Tempo
6.
Mol Ther ; 15(7): 1306-1312, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28182918

RESUMO

Poly(ß-amino ester)s are cationic degradable polymers that have significant potential as gene delivery vectors. Here we present a generalized method to modify poly(ß-amino ester)s at the chain ends to improve their delivery performance. End-chain coupling reactions were developed so that polymers could be synthesized and tested in a high-throughput manner, without the need for purification. In this way, many structural variations at the polymer terminus could be rapidly evaluated. End-modification of the terminal amine structure of a previously optimized poly(ß-amino ester), C32, significantly enhanced its in vitro transfection efficiency. In vivo, intraperitoneal (IP) gene delivery using end-modified C32 polymers resulted in expression levels over one order of magnitude higher than unmodified C32 and jet-polyethylenimine (jet-PEI) levels in several abdominal organs. The rapid end-modification strategy presented here has led to the discovery of many effective polymers for gene delivery and may be a useful method to develop and optimize cationic polymers for gene therapy.

7.
Adv Exp Med Biol ; 622: 209-19, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18546630

RESUMO

Intraperitoneal administration of polymeric nanoparticles to deliver DNA encoding suicide genes holds much promise as an effective therapy for advanced epithelial ovarian cancer. Poly(beta-amino ester)s, a class of cationic, biodegradable polymers complex to DNA to form nanoparticles that deliver DNA to cells in ovarian tumors. Modifications to poly(beta-amino ester)s can improve both the efficiency and specificity with which DNA is delivered to tumor cells. Preclinical studies to test therapeutic efficacy of gene therapy strategies that are under development make use of mouse models for epithelial ovarian cancer and new imaging technologies.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Genes Transgênicos Suicidas , Terapia Genética/métodos , Nanopartículas/uso terapêutico , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Animais , DNA/uso terapêutico , Feminino , Humanos
8.
Mol Cell Biol ; 38(3)2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29133460

RESUMO

Human antigen R (ELAVL1; HuR) is perhaps the best-characterized RNA-binding protein. Through its overexpression in various tumor types, HuR promotes posttranscriptional regulation of target genes in multiple core signaling pathways associated with tumor progression. The role of HuR overexpression in pancreatic tumorigenesis is unknown and led us to explore the consequences of HuR overexpression using a novel transgenic mouse model that has a >2-fold elevation of pancreatic HuR expression. Histologically, HuR-overexpressing pancreas displays a fibroinflammatory response and other pathological features characteristic of chronic pancreatitis. This pathology is reflected in changes in the pancreatic gene expression profile due, in part, to genes whose expression changes as a consequence of direct binding of their respective mRNAs to HuR. Older mice develop pancreatic steatosis and severe glucose intolerance. Elevated HuR cooperated with mutant K-rasG12D to result in a 3.4-fold increase in pancreatic ductal adenocarcinoma (PDAC) incidence compared to PDAC presence in K-rasG12D alone. These findings implicate HuR as a facilitator of pancreatic tumorigenesis, especially in the setting of inflammation, and a novel therapeutic target for pancreatitis treatment.


Assuntos
Proteína Semelhante a ELAV 1/genética , Neoplasias Pancreáticas/etiologia , Pancreatite/patologia , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Citoplasma/genética , Citoplasma/metabolismo , Proteína Semelhante a ELAV 1/metabolismo , Feminino , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pâncreas/patologia , Pâncreas/fisiologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Pancreatite/genética , Papiloma/etiologia , Papiloma/genética , Papiloma/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Neoplasias Pancreáticas
9.
Nat Biotechnol ; 22(4): 411-7, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15024388

RESUMO

The hair follicle bulge possesses putative epithelial stem cells. Characterization of these cells has been hampered by the inability to target bulge cells genetically. Here, we use a Keratin1-15 (Krt1-15, also known as K15) promoter to target mouse bulge cells with an inducible Cre recombinase construct or with the gene encoding enhanced green fluorescent protein (EGFP), which allow for lineage analysis and for isolation of the cells. We show that bulge cells in adult mice generate all epithelial cell types within the intact follicle and hair during normal hair follicle cycling. After isolation, adult Krt1-15-EGFP-positive cells reconstituted all components of the cutaneous epithelium and had a higher proliferative potential than Krt1-15-EGFP-negative cells. Genetic profiling of hair follicle stem cells revealed several known and unknown receptors and signaling pathways important for maintaining the stem cell phenotype. Ultimately, these findings provide potential targets for the treatment of hair loss and other disorders of skin and hair.


Assuntos
Células Epiteliais/citologia , Técnicas Genéticas , Folículo Piloso/fisiologia , Células-Tronco/citologia , Células 3T3 , Animais , Divisão Celular , Linhagem Celular , Linhagem da Célula , Separação Celular , DNA Complementar/metabolismo , Fibroblastos/metabolismo , Citometria de Fluxo , Proteínas de Fluorescência Verde , Folículo Piloso/metabolismo , Queratinócitos/metabolismo , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , RNA/metabolismo , RNA Mensageiro/metabolismo , Recombinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Regulação para Cima
10.
Cancer Res ; 76(6): 1549-59, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26921342

RESUMO

Growing evidence shows that cancer cells use mRNA-binding proteins and miRNAs to posttranscriptionally regulate signaling pathways to adapt to harsh tumor microenvironments. In ovarian cancer, cytoplasmic accumulation of mRNA-binding protein HuR (ELAVL1) is associated with poor prognosis. In this study, we observed high HuR expression in ovarian cancer cells compared with ovarian primary cells, providing a rationale for targeting HuR. RNAi-mediated silencing of HuR in ovarian cancer cells significantly decreased cell proliferation and anchorage-independent growth, and impaired migration and invasion. In addition, HuR-depleted human ovarian xenografts were smaller than control tumors. A biodistribution study showed effective tumor-targeting by a novel Cy3-labeled folic acid (FA)-derivatized DNA dendrimer nanocarrier (3DNA). We combined siRNAs against HuR with FA-3DNA and found that systemic administration of the resultant FA-3DNA-siHuR conjugates to ovarian tumor-bearing mice suppressed tumor growth and ascites development, significantly prolonging lifespan. NanoString gene expression analysis identified multiple HuR-regulated genes that function in many essential cellular and molecular pathways, an attractive feature of candidate therapeutic targets. Taken together, these results are the first to demonstrate the versatility of the 3DNA nanocarrier for in vivo-targeted delivery of a cancer therapeutic and support further preclinical investigation of this system adapted to siHuR-targeted therapy for ovarian cancer.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proteína Semelhante a ELAV 1/administração & dosagem , Nanopartículas/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , RNA Mensageiro/administração & dosagem , Proteínas de Ligação a RNA/administração & dosagem , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Sistemas de Liberação de Medicamentos/métodos , Proteína Semelhante a ELAV 1/genética , Feminino , Células HEK293 , Heterozigoto , Humanos , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/administração & dosagem , MicroRNAs/genética , Neoplasias Ovarianas/genética , Interferência de RNA/efeitos dos fármacos , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Distribuição Tecidual/genética
11.
Oncotarget ; 7(16): 21812-24, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-26943573

RESUMO

This retrospective study aimed to investigate the role that an RNA-binding protein, HuR, plays in the response of high-grade serous ovarian tumors to chemotherapeutics. We immunohistochemically stained sections of 31 surgically-debulked chemo-naïve ovarian tumors for HuR and scored the degree of HuR cytoplasmic staining. We found no correlation between HuR intracellular localization in tumor sections and progression free survival (PFS) of these patients, 29 of whom underwent second-line gemcitabine/platin combination therapy for recurrent disease. Ribonucleoprotein immunoprecipitation (RNP-IP) analysis of ovarian cancer cells in culture showed that cytoplasmic HuR increases deoxycytidine kinase (dCK), a metabolic enzyme that activates gemcitabine. The effects of carboplatin treatment on HuR and WEE1 (a mitotic inhibitor) expression, and on cell cycle kinetics, were also examined. Treatment of ovarian cancer cells with carboplatin results in increased HuR cytoplasmic expression and elevated WEE1 expression, arresting cell cycle G2/M transition. This may explain why HuR cytoplasmic localization in chemo-naïve tumors is not predictive of therapeutic response and PFS following second-line gemcitabine/platin combination therapy. These results suggest treatment of recurrent ovarian tumors with a combination of gemcitabine, carboplatin, and a WEE1 inhibitor may be potentially advantageous as compared to current clinical practices.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteína Semelhante a ELAV 1/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Idoso , Carboplatina/administração & dosagem , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Desoxicitidina Quinase/metabolismo , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Proteínas Nucleares/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Transporte Proteico/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Estudos Retrospectivos , Gencitabina
12.
Mol Cancer Res ; 14(7): 599-611, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27053682

RESUMO

UNLABELLED: Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal cancers, in part, due to resistance to both conventional and targeted therapeutics. TRAIL directly induces apoptosis through engagement of cell surface Death Receptors (DR4 and DR5), and has been explored as a molecular target for cancer treatment. Clinical trials with recombinant TRAIL and DR-targeting agents, however, have failed to show overall positive outcomes. Herein, we identify a novel TRAIL resistance mechanism governed by Hu antigen R (HuR, ELAV1), a stress-response protein abundant and functional in PDA cells. Exogenous HuR overexpression in TRAIL-sensitive PDA cell lines increases TRAIL resistance whereas silencing HuR in TRAIL-resistant PDA cells, by siRNA oligo-transfection, decreases TRAIL resistance. PDA cell exposure to soluble TRAIL induces HuR translocation from the nucleus to the cytoplasm. Furthermore, it is demonstrated that HuR interacts with the 3'-untranslated region (UTR) of DR4 mRNA. Pre-treatment of PDA cells with MS-444 (Novartis), an established small molecule inhibitor of HuR, substantially increased DR4 and DR5 cell surface levels and enhanced TRAIL sensitivity, further validating HuR's role in affecting TRAIL apoptotic resistance. NanoString analyses on the transcriptome of TRAIL-exposed PDA cells identified global HuR-mediated increases in antiapoptotic processes. Taken together, these data extend HuR's role as a key regulator of TRAIL-induced apoptosis. IMPLICATIONS: Discovery of an important new HuR-mediated TRAIL resistance mechanism suggests that tumor-targeted HuR inhibition increases sensitivity to TRAIL-based therapeutics and supports their re-evaluation as an effective treatment for PDA patients. Mol Cancer Res; 14(7); 599-611. ©2016 AACR.


Assuntos
Carcinoma Ductal Pancreático/tratamento farmacológico , Proteína Semelhante a ELAV 1/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Proteínas Recombinantes/farmacologia , Transfecção
13.
Oncotarget ; 6(29): 27312-31, 2015 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-26314962

RESUMO

Post-transcriptional regulation is a powerful mediator of gene expression, and can rapidly alter the expression of numerous transcripts involved in tumorigenesis. We have previously shown that the mRNA-binding protein HuR (ELAVL1) is elevated in human pancreatic ductal adenocarcinoma (PDA) specimens compared to normal pancreatic tissues, and its cytoplasmic localization is associated with increased tumor stage. To gain a better insight into HuR's role in PDA biology and to assess it as a candidate therapeutic target, we altered HuR expression in PDA cell lines and characterized the resulting phenotype in preclinical models. HuR silencing by short hairpin and small interfering RNAs significantly decreased cell proliferation and anchorage-independent growth, as well as impaired migration and invasion. In comparison, HuR overexpression increased migration and invasion, but had no significant effects on cell proliferation and anchorage-independent growth. Importantly, two distinct targeted approaches to HuR silencing showed marked impairment in tumor growth in mouse xenografts. NanoString nCounter® analyses demonstrated that HuR regulates core biological processes, highlighting that HuR inhibition likely thwarts PDA viability through post-transcriptional regulation of diverse signaling pathways (e.g. cell cycle, apoptosis, DNA repair). Taken together, our study suggests that targeted inhibition of HuR may be a novel, promising approach to the treatment of PDA.


Assuntos
Adenocarcinoma/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Proteína Semelhante a ELAV 1/metabolismo , Neoplasias Pancreáticas/metabolismo , Adenocarcinoma/tratamento farmacológico , Animais , Carcinoma Ductal Pancreático/tratamento farmacológico , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Citoplasma/metabolismo , Doxiciclina/química , Proteína Semelhante a ELAV 1/antagonistas & inibidores , Feminino , Inativação Gênica , Humanos , Camundongos , Nanopartículas/química , Invasividade Neoplásica , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Pancreáticas/tratamento farmacológico , Fenótipo , Análise de Componente Principal , RNA/metabolismo , Processamento Pós-Transcricional do RNA , Transdução de Sinais
14.
Mol Cancer Res ; 13(3): 439-48, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25336517

RESUMO

UNLABELLED: Mucin1 (MUC1) is overexpressed in pancreatic ductal adenocarcinoma (PDA) and is associated with tumor aggressiveness, suggesting that MUC1 is a promising therapeutic target for promoter-driven diphtheria toxin A (DTA). Endogenous MUC1 transcript levels were analyzed by quantitative PCR (qPCR) in multiple PDA cells (Capan1, HPAFII, Su.86.86, Capan2, Hs766T, MiaPaCa2, and Panc1). Expression levels were correlated with luciferase activity and cell death after transfection with MUC1 promoter-driven luciferase and DTA constructs. MUC1-positive (+) cells had significantly elevated MUC1 mRNA expression compared with MUC1-negative (-) cells. Luciferase activity was significantly higher in MUC1(+) cells when transfected with MUC1 promoter-driven luciferase and MUC1(+) cells underwent enhanced cell death after transfection with a single dose of MUC1 promoter-driven DTA. IFNγ pretreatment enhanced MUC1 expression in MUC1(-) cells and induced sensitivity to MUC1-DTA therapy. Matched primary and metastatic tumor lesions from clinical specimens revealed similar MUC1 IHC labeling patterns, and a tissue microarray of human PDA biopsies revealed increased immunolabeling with a combination of MUC1 and mesothelin (MSLN) antibodies, compared with either antibody alone. Combining MUC1 with MSLN-targeted DTA enhanced drug efficacy in an in vitro model of heterogeneous PDA. These data demonstrate that MUC1 promoter-driven DTA preferentially kills MUC1-expressing PDA cells and drugs that enhance MUC1 expression sensitize PDA cells with low MUC1 expression. IMPLICATIONS: MUC1 expression in primary and metastatic lesions provides a rationale for the development of a systemic MUC1 promoter-driven DTA therapy that may be further enhanced by combination with other promoter-driven DTA constructs.


Assuntos
Carcinoma Ductal Pancreático/terapia , Toxina Diftérica/farmacologia , Terapia de Alvo Molecular/métodos , Mucina-1/genética , Neoplasias Pancreáticas/terapia , Fragmentos de Peptídeos/farmacologia , Regiões Promotoras Genéticas , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Morte Celular , Linhagem Celular Tumoral , Toxina Diftérica/genética , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos/farmacologia , Humanos , Interferon gama/farmacologia , Mesotelina , Mucina-1/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fragmentos de Peptídeos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia
15.
Cancer Res ; 74(4): 1128-40, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24536047

RESUMO

HuR (ELAV1), an RNA-binding protein abundant in cancer cells, primarily resides in the nucleus, but under specific stress (e.g., gemcitabine), HuR translocates to the cytoplasm in which it tightly modulates the expression of mRNA survival cargo. Here, we demonstrate for the first time that stressing pancreatic ductal adenocarcinoma (PDA) cells by treatment with DNA-damaging anticancer agents (mitomycin C, oxaliplatin, cisplatin, carboplatin, and a PARP inhibitor) results in HuR's translocation from the nucleus to the cytoplasm. Importantly, silencing HuR in PDA cells sensitized the cells to these agents, whereas overexpressing HuR caused resistance. HuR's role in the efficacy of DNA-damaging agents in PDA cells was, in part, attributed to the acute upregulation of WEE1 by HuR. WEE1, a mitotic inhibitor kinase, regulates the DNA damage repair pathway, and therapeutic inhibition of WEE1 in combination with chemotherapy is currently in early phase trials for the treatment of cancer. We validate WEE1 as a HuR target in vitro and in vivo by demonstrating (i) direct binding of HuR to WEE1's mRNA (a discrete 56-bp region residing in the 3' untranslated region) and (ii) HuR siRNA silencing and overexpression directly affects the protein levels of WEE1, especially after DNA damage. HuR's positive regulation of WEE1 increases γ-H2AX levels, induces Cdk1 phosphorylation, and promotes cell-cycle arrest at the G2-M transition. We describe a novel mechanism that PDA cells use to protect against DNA damage in which HuR posttranscriptionally regulates the expression and downstream function of WEE1 upon exposure to DNA-damaging agents.


Assuntos
Carcinoma Ductal Pancreático/genética , Proteínas de Ciclo Celular/genética , Dano ao DNA/fisiologia , Proteínas ELAV/fisiologia , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Proteínas Tirosina Quinases/genética , Interferência de RNA , Animais , Antineoplásicos/farmacologia , Carcinoma Ductal Pancreático/metabolismo , Proteínas de Ciclo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/metabolismo , Transporte Proteico/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Células Tumorais Cultivadas
16.
Mol Cancer Res ; 11(8): 901-11, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23696131

RESUMO

UNLABELLED: Pancreatic ductal adenocarcinoma (PDA) is the fourth leading cause of cancer-related death in the United States, with a 95% five-year mortality rate. For over a decade, gemcitabine (GEM) has been the established first-line treatment for this disease despite suboptimal response rates. The development of PARP inhibitors that target the DNA damage repair (DDR) system in PDA cells has generated encouraging results. Ubiquitin-specific peptidase 11 (USP11), an enzyme that interacts with the DDR protein BRCA2, was recently discovered to play a key role in DNA double-strand break repair and may be a novel therapeutic target. A systematic high-throughput approach was used to biochemically screen 2,000 U.S. Food and Drug Administration (FDA)-approved compounds for inhibition of USP11 enzymatic activity. Six pharmacologically active small molecules that inhibit USP11 enzymatic activity were identified. An in vitro drug sensitivity assay demonstrated that one of these USP11 inhibitors, mitoxantrone, impacted PDA cell survival with an IC50 of less than 10 nM. Importantly, across six different PDA cell lines, two with defects in the Fanconi anemia/BRCA2 pathway (Hs766T and Capan-1), mitoxantrone is 40- to 20,000-fold more potent than GEM, with increased endogenous USP11 mRNA levels associated with increased sensitivity to mitoxantrone. Interestingly, USP11 silencing in PDA cells also enhanced sensitivity to GEM. These findings establish a preclinical model for the rapid discovery of FDA-approved compounds and identify USP11 as a target of mitoxantrone in PDA. IMPLICATIONS: This high-throughput approach provides a strong rationale to study mitoxantrone in an early-phase clinical setting for the treatment of PDA.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Desoxicitidina/análogos & derivados , Mitoxantrona/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Tioléster Hidrolases/antagonistas & inibidores , Proteína BRCA2/genética , Benzimidazóis/uso terapêutico , Carcinoma Ductal Pancreático/enzimologia , Linhagem Celular Tumoral , Dano ao DNA/genética , Desoxicitidina/uso terapêutico , Ensaios de Seleção de Medicamentos Antitumorais , Inativação Gênica , Ensaios de Triagem em Larga Escala , Humanos , Neoplasias Pancreáticas/enzimologia , Poli(ADP-Ribose) Polimerase-1 , Inibidores de Poli(ADP-Ribose) Polimerases , Tioléster Hidrolases/metabolismo , Gencitabina
17.
Cancer J ; 18(6): 665-73, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23187855

RESUMO

The current treatment options for pancreatic ductal adenocarcinoma fall exceedingly short of a cure, providing a sobering 5-year survival rate of only 5% for all patients, and the disease will be responsible for more than 37,000 deaths in the United States alone this year. These numbers continue to grow, and it was recently predicted that, within decades, pancreatic ductal adenocarcinoma will become the second most lethal cancer in this country. Beyond conventional oncologic-based therapies, researchers are working hard, albeit with increasingly limited federal support, to develop multiple novel therapeutic options focusing primarily on targeting specific, disrupted core signaling pathways within pancreatic cancer cells. In line with the history of medical oncology and medical paradigms, many pharmaceutical companies and large academic institutions have been focused on searching for compounds (biologic and chemicals) in an effort to find that unique "magic bullet" that will extend pancreatic cancer patients' lives (e.g., K-ras inhibitors). This magic bullet has been difficult to find in a haystack full of molecular pathways and mutated genes because the challenge is defined by identifying a therapeutic window that kills the tumor, yet spares the host. This therapeutic window has been hard to discover in the backdrop of the heterogeneous cell populations that make up a pancreatic tumor together with a heterogeneous patient population that has multiple, undefined tumor subtypes. Thus, to date, efforts have had limited success. Perhaps the best recent example of limited success is the discovery of the classic combination of 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX), extending life by only 4 months when compared with gemcitabine (note that this drug combination does not directly target one single pathway or pancreatic cancer subtype).


Assuntos
Carcinoma Ductal Pancreático/terapia , Terapias Complementares/métodos , Neoplasias Pancreáticas/terapia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/patologia , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Taxa de Sobrevida , Resultado do Tratamento
18.
Cancer Biol Ther ; 10(9): 878-84, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20798601

RESUMO

Gene therapy protocols for the treatment of cancer often employ gene promoter sequences that are known to be over-expressed in specific tumor cell types relative to normal cells. These promoters, while specific, are often weakly active. It would be desirable to increase the activity of such promoters, while at the same time retain specificity, so that the therapeutic gene is more robustly expressed. Using a luciferase reporter DNA construct in both in vitro cell transfection assays and in vivo mouse tumor models, we have determined that in the absence of any other DNA sequence, a previously identified 18-base pair enhancer sequence called CanScript, lying upstream of the MSLN gene, has ~25% of the promoter activity of CAG, a very strong non-specific promoter/enhancer, in tumor cells in which MSLN is highly expressed. Furthermore, tandem repeat copies of CanScript enhance transcription in a dose-dependent manner and, when coupled with promoter sequences that are active in tumor cells, increase promoter activity. These findings suggest that the incorporation of CanScript into gene constructs may have application in enhancing activity of promoters used in cancer-targeting gene therapy strategies, thereby improving therapeutic efficacy.


Assuntos
Elementos Facilitadores Genéticos , Proteínas Ligadas por GPI/genética , Terapia Genética/métodos , Terapia de Alvo Molecular/métodos , Neoplasias Experimentais/genética , Neoplasias Experimentais/terapia , Regiões Promotoras Genéticas , Animais , Sequência de Bases , Linhagem Celular Tumoral , DNA/genética , Genes Reporter , Luciferases/genética , Luciferases/metabolismo , Mesotelina , Camundongos , Reação em Cadeia da Polimerase , Sequências de Repetição em Tandem , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas
19.
Cancer Res ; 69(15): 6184-91, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19643734

RESUMO

There is currently no effective therapy for patients with advanced ovarian cancer. To address the need for a more effective treatment for this deadly disease, we conducted preclinical tests in ovarian tumor-bearing mice to evaluate the therapeutic efficacy of using a cationic biodegradable poly(beta-amino ester) polymer as a vector for nanoparticulate delivery of DNA encoding a diphtheria toxin suicide protein (DT-A). The promoter sequences of two genes that are highly active in ovarian tumor cells, MSLN and HE4, were used to target DT-A expression to tumor cells. Administration of DT-A nanoparticles directly to s.c. xenograft tumors and to the peritoneal cavity of mice bearing primary and metastatic ovarian tumors resulted in a significant reduction in tumor mass and a prolonged life span compared to control mice. Minimal nonspecific tissue and blood chemistry toxicity was observed following extended treatment with nanoparticles. DT-A nanoparticle therapy suppressed tumor growth more effectively than treatment with clinically relevant doses of cisplatin and paclitaxel. Our findings suggest that i.p. administration of polymeric nanoparticles to deliver DT-A encoding DNA, combined with transcriptional regulation to target gene expression to ovarian tumor cells, holds promise as an effective therapy for advanced-stage ovarian cancer.


Assuntos
DNA/administração & dosagem , Terapia Genética/métodos , Nanopartículas/administração & dosagem , Neoplasias Ovarianas/terapia , Polímeros/administração & dosagem , Animais , DNA/genética , Proteínas Secretadas pelo Epidídimo/genética , Feminino , Proteínas Ligadas por GPI , Vetores Genéticos/administração & dosagem , Humanos , Glicoproteínas de Membrana/genética , Mesotelina , Camundongos , Nanopartículas/química , Neoplasias Ovarianas/genética , Polímeros/química , Regiões Promotoras Genéticas , Ensaios Antitumorais Modelo de Xenoenxerto , beta-Defensinas
20.
Cancer Res ; 68(23): 9567-9, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19047129

RESUMO

The persistence of fetal stem cells with multilineage potential in women who have been pregnant, a phenomenon known as fetal microchimerism, is emerging as a potential contributing factor in certain diseases, including cancer. For example, fetal microchimerism has been implicated in autoimmune disease, wound healing, and cancer. Studies of this phenomenon may provide a novel perspective on cancer in women, including in breast, ovarian, and lung cancers.


Assuntos
Transformação Celular Neoplásica/patologia , Quimerismo/embriologia , Células-Tronco Fetais/patologia , Neoplasias/patologia , Neoplasias da Mama/etiologia , Neoplasias da Mama/patologia , Feminino , Células-Tronco Fetais/fisiologia , Humanos , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/patologia , Neoplasias/etiologia , Neoplasias Ovarianas/etiologia , Neoplasias Ovarianas/patologia , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA