Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Gastroenterology ; 147(2): 485-97.e18, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24859161

RESUMO

BACKGROUND & AIMS: Dysregulation of ß-catenin and the transcriptional activator FOXM1 mediate oncogenesis, but it is not clear how these proteins become dysregulated in tumors that do not typically carry mutations in adenomatous polyposis coli (APC) or ß-catenin, such as pancreatic ductal adenocarcinomas (PDACs). We searched for microRNAs that regulate levels of FOXM1 in PDAC cells and samples from patients. METHODS: We identified microRNAs that affect levels of FOXM1 in PDACs using bioinformatic, genetic, and pharmacologic approaches. We altered expression of the microRNA-494 (miR-494) in PDAC cell lines (AsPC-1 and PANC-1) and examined the effects on FOXM1 and ß-catenin signaling and cell proliferation and colony formation. The cells were injected into immunocompromised mice and growth of xenograft tumors and liver metastases were measured. We performed immunohistochemical analyses of 10 paired PDAC and nontumor pancreatic tissue samples collected from untreated patients during surgery. RESULTS: We identified miR-494 as a negative regulator of FOXM1 levels in PDAC cells, and found that levels of this microRNA were reduced in PDAC specimens, compared with nontumor tissues. Loss of response of PDAC cells to transforming growth factor ß, owing to SMAD4 deficiency, reduced expression of miR-494. Transgenic expression of miR-494 in PDAC cells produced the same effects as reducing expression of FOXM1 or blocking nuclear translocation of ß-catenin, reducing cell proliferation, migration, and invasion, and increasing their sensitivity to gemcitabine. Reduced expression of miR-494 correlated with PDAC metastasis and reduced survival times of patients. CONCLUSIONS: Loss of SMAD4 in PDAC cells leads to reduced levels of miR-494, increased levels of FOXM1, and nuclear localization of ß-catenin. miR-494 might be developed as a prognostic marker for patients with PDAC or a therapeutic target.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Fatores de Transcrição Forkhead/metabolismo , MicroRNAs/metabolismo , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais , Proteína Smad4/metabolismo , beta Catenina/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Antibióticos Antineoplásicos/farmacologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/mortalidade , Carcinoma Ductal Pancreático/secundário , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Feminino , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Pessoa de Meia-Idade , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Proteína Smad4/genética , Fatores de Tempo , Transfecção , Regulação para Cima , beta Catenina/genética , Gencitabina
2.
J Immunother Cancer ; 12(1)2024 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-38199607

RESUMO

BACKGROUND: The extracellular matrix (ECM) and cancer-associated fibroblasts (CAFs) play major roles in tumor progression, metastasis, and the poor response of many solid tumors to immunotherapy. CAF-targeted chimeric antigen receptor-T cell therapy cannot infiltrate ECM-rich tumors such as osteosarcoma. METHOD: In this study, we used RNA sequencing to assess whether the recently invented membrane-anchored and tumor-targeted IL-12-armed (attIL12) T cells, which bind cell-surface vimentin (CSV) on tumor cells, could destroy CAFs to disrupt the ECM. We established an in vitro model of the interaction between osteosarcoma CAFs and attIL12-T cells to uncover the underlying mechanism by which attIL12-T cells penetrate stroma-enriched osteosarcoma tumors. RESULTS: RNA sequencing demonstrated that attIL12-T cell treatment altered ECM-related gene expression. Immunohistochemistry staining revealed disruption or elimination of high-density CAFs and ECM in osteosarcoma xenograft tumors following attIL12-T cell treatment, and CAF/ECM density was inversely correlated with T-cell infiltration. Other IL12-armed T cells, such as wild-type IL-12-targeted or tumor-targeted IL-12-T cells, did not disrupt the ECM because this effect depended on the engagement between CSV on the tumor cell and its ligand on the attIL12-T cells. Mechanistic studies found that attIL12-T cell treatment elevated IFNγ production on interacting with CSV+ tumor cells, suppressing transforming growth factor beta secretion and in turn upregulating FAS-mediated CAF apoptosis. CAF destruction reshaped the tumor stroma to favor T-cell infiltration and tumor inhibition. CONCLUSIONS: This study unveiled a novel therapy-attIL12-T cells-for targeting CAFs/ECM. These findings are highly relevant to humans because CAFs are abundant in human osteosarcoma.


Assuntos
Neoplasias Ósseas , Fibroblastos Associados a Câncer , Osteossarcoma , Animais , Humanos , Interleucina-12 , Xenoenxertos , Osteossarcoma/terapia , Membrana Celular , Matriz Extracelular , Modelos Animais de Doenças , Neoplasias Ósseas/terapia , Terapia Baseada em Transplante de Células e Tecidos
3.
Gastroenterology ; 143(3): 799-810.e2, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22677193

RESUMO

BACKGROUND & AIMS: Krüppel-like factor 4 (KLF4) is a transcription factor and putative tumor suppressor. However, little is known about its effects in hepatocellular carcinogenesis. We investigated the clinical significance, biologic effects, and mechanisms of dysregulated KLF4 signaling. METHODS: We performed microarray analysis of hepatocellular carcinoma (HCC) tissues. We used molecular biology analyses and animal models to evaluate activation and function of KLF4-vitamin D receptor (VDR) pathway. RESULTS: Expression of KLF4 protein was decreased or lost in primary HCC samples, in particular, lymph node metastases, compared with normal liver tissues. Loss of KLF4 from primary tumors was significantly associated with reduced survival time and was identified as a prognostic marker. Most human HCC cell lines had losses or substantial decreases in levels of KLF4. Exogenous expression of KLF4 in HCC cells upregulated expression of mesenchymal-epithelial transition (MET) and inhibited their migration, invasion, and proliferation in vitro. When these cells were injected into mice, tumors grew more slowly and metastasis was inhibited, compared with HCC cells that did not express KLF4. VDR is a direct transcriptional target of KLF4; we identified 2 sites in the VDR promoter that bound specifically to KLF4. Increased expression of VDR sensitized tumor cells to the inhibitory effects of vitamin D. CONCLUSIONS: KLF4 binds to the promoter of VDR to regulate its expression; levels of KLF4 are reduced and levels of VDR are increased in HCC cell lines and primary tumor samples. Expression of KLF4 in HCC cells sensitizes them to the anti-proliferative effects of VD3. This pathway might be manipulated to prevent or treat liver cancer.


Assuntos
Carcinoma Hepatocelular/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Neoplasias Hepáticas/metabolismo , Receptores de Calcitriol/metabolismo , Transdução de Sinais , Animais , Sítios de Ligação , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/secundário , Movimento Celular , Proliferação de Células , Progressão da Doença , Transição Epitelial-Mesenquimal , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Imuno-Histoquímica , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , Metástase Linfática , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Regiões Promotoras Genéticas , Receptores de Calcitriol/genética , Fatores de Tempo , Análise Serial de Tecidos/métodos , Transfecção , Carga Tumoral
4.
Gastroenterology ; 142(3): 531-42, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22155367

RESUMO

BACKGROUND & AIMS: Krüppel-like factor 4 (Klf4) is a putative gastric tumor suppressor gene. Rare, villin-positive progenitor cells in the gastric antrum have multilineage potential. We investigated the function of Klf4 in these cells and in gastric carcinogenesis. METHODS: We created mice with disruption of Klf4 in villin-positive antral mucosa cells (Villin-Cre(+);Klf4(fl/fl) mice). Villin-Cre(+);Klf4(fl/fl) and control mice were given drinking water with or without 240 ppm N-methyl-N-nitrosourea at 5 weeks of age and thereafter on alternating weeks for a total of 10 weeks. Gastric mucosa samples were collected at 35, 50, or 80 weeks of age from mice that were and were not given N-methyl-N-nitrosourea, and analyzed by histopathologic and molecular analyses. Findings were compared with those from human gastric tumor specimens. RESULTS: Preneoplasia formed progressively in the antrum in 35- to 80-week-old Villin-Cre(+);Klf4(fl/fl) mice. Gastric tumors developed in 29% of 80-week-old Villin-Cre(+);Klf4(fl/fl) mice, which were located exclusively in the lesser curvature of the antrum. N-methyl-N-nitrosourea accelerated tumor formation, and tumors developed significantly more frequently in Villin-Cre(+);Klf4(fl/fl) mice than in control mice, at 35 and 50 weeks of age. Mouse and human gastric tumors had reduced expression of Krüppel-like factor 4 and increased expression of FoxM1 compared with healthy gastric tissue. Expression of Krüppel-like factor 4 suppressed transcription of FoxM1. CONCLUSIONS: Inactivation of Klf4 in villin-positive gastric progenitor cells induces transformation of the gastric mucosa and tumorigenesis in the antrum in mice. Villin-Cre(+);Klf4(fl/fl) have greater susceptibility to chemical-induced gastric carcinogenesis and increased rates of gastric tumor progression than control mice.


Assuntos
Mucosa Gástrica/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas dos Microfilamentos/genética , Células-Tronco Neoplásicas/metabolismo , Lesões Pré-Cancerosas/metabolismo , Antro Pilórico/metabolismo , Neoplasias Gástricas/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/metabolismo , Mucosa Gástrica/patologia , Genótipo , Humanos , Integrases/genética , Integrases/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/genética , Metilnitrosoureia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Neoplásicas/patologia , Fenótipo , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Regiões Promotoras Genéticas , Antro Pilórico/patologia , Neoplasias Gástricas/induzido quimicamente , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Fatores de Tempo
5.
Nat Commun ; 14(1): 735, 2023 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-36759517

RESUMO

Although tissue-resident memory T (TRM) cells specific for previously encountered pathogens have been characterized, the induction and recruitment of brain TRM cells following immune therapy has not been observed in the context of glioblastoma. Here, we show that T cells expressing fibrinogen-like 2 (FGL2)-specific single-chain variable fragments (T-αFGL2) can induce tumor-specific CD8+ TRM cells that prevent glioblastoma recurrence. These CD8+ TRM cells display a highly expanded T cell receptor repertoire distinct from that found in peripheral tissue. When adoptively transferred to the brains of either immunocompetent or T cell-deficient naïve mice, these CD8+ TRM cells reject glioma cells. Mechanistically, T-αFGL2 cell treatment increased the number of CD69+CD8+ brain-resident memory T cells in tumor-bearing mice via a CXCL9/10 and CXCR3 chemokine axis. These findings suggest that tumor-specific brain-resident CD8+ TRM cells may have promising implications for the prevention of brain tumor recurrence.


Assuntos
Linfócitos T CD8-Positivos , Glioblastoma , Animais , Camundongos , Encéfalo , Glioblastoma/terapia , Memória Imunológica , Células T de Memória , Recidiva Local de Neoplasia , Linfócitos T/imunologia
6.
Front Immunol ; 13: 952231, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36203573

RESUMO

Interleukin 12 (IL-12) is a naturally occurring cytokine that plays a key role in inducing antitumor immune responses, including induction of antitumor immune memory. Currently, no IL-12-based therapeutic products have been approved for clinical application because of its toxicities. On the basis of this review of clinical trials using primarily wild-type IL-12 and different delivery methods, we conclude that the safe utilization of IL-12 is highly dependent on the tumor-specific localization of IL-12 post administration. In this regard, we have developed a cell membrane-anchored and tumor-targeted IL-12-T (attIL12-T) cell product for avoiding toxicity from both IL-12 and T cells-induced cytokine release syndrome in peripheral tissues. A phase I trial using this product which seeks to avoid systemic toxicity and boost antitumor efficacy is on the horizon. Of note, this product also boosts the impact of CAR-T or TCR-T cell efficacy against solid tumors, providing an alternative approach to utilize CAR-T to overcome tumor resistance.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Citocinas , Humanos , Interleucina-12 , Linfócitos T
7.
Clin Cancer Res ; 28(17): 3862-3873, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35727602

RESUMO

PURPOSE: Chimeric antigen receptor (CAR) T-cell therapy has shown great promise for treating hematologic malignancies but requires a long duration of T-cell expansion, is associated with severe toxicity, and has limited efficacy for treating solid tumors. We designed experiments to address those challenges. EXPERIMENTAL DESIGN: We generated a cell membrane-anchored and tumor-targeted IL12 (attIL12) to arm peripheral blood mononuclear cells (PBMC) instead of T cells to omit the expansion phase for required CAR T cells. RESULTS: This IL12-based attIL12-PBMC therapy showed significant antitumor efficacy in both heterogeneous osteosarcoma patient-derived xenograft tumors and metastatic osteosarcoma tumors with no observable toxic effects. Mechanistically, attIL12-PBMC treatment resulted in tumor-restricted antitumor cytokine release and accumulation of attIL12-PBMCs in tumors. It also induced terminal differentiation of osteosarcoma cells into bone-like cells to impede tumor growth. CONCLUSIONS: In summary, attIL12-PBMC therapy is safe and effective against osteosarcoma. Our goal is to move this treatment into a clinical trial. Owing to the convenience of the attIL12-PBMC production process, we believe it will be feasible.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Neoplasias Ósseas/terapia , Linhagem Celular Tumoral , Humanos , Imunoterapia Adotiva/métodos , Interleucina-12 , Leucócitos Mononucleares , Osteossarcoma/tratamento farmacológico , Receptores de Antígenos de Linfócitos T , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Gastroenterology ; 139(6): 2135-45, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20727893

RESUMO

BACKGROUND & AIMS: Krüppel-like factor 4 (KLF4) is a transcription factor associated with tumor suppression and oncogenesis. KLF4 suppresses pancreatic tumorigenesis by unknown mechanisms; we investigated alterations that might affect KLF4 function and lead to tumor formation. METHODS: We identified different isoforms of KLF4 in pancreatic cancer cells by reverse-transcriptase polymerase chain reaction, cloning, and DNA sequence analyses. We constructed vectors to express the isoform KLF4α and characterize its function. Using real-time polymerase chain reaction, immunoprecipitation, and immunohistochemical analyses, we assessed expression of KLF4α in pancreatic cancer cell lines and tumor tissue samples; xenograft models were used to determine the effect of KLF4α on pancreatic tumorigenesis. RESULTS: We identified 4 KLF4 isoforms in human pancreatic cancer cells, designated KLF4α, KLF4ß, KLF4γ, and KLF4δ. KLF4α localized primarily to the cytoplasm; its protein and messenger RNA were up-regulated in pancreatic cancer cell lines with high metastatic potential and human pancreatic tumors compared with normal pancreatic tissue. Transgenic expression of KLF4α reduced expression of p27(Kip1) and p21(Cip1), promoting cell cycle progression and in vivo tumor formation by pancreatic cancer cells. Increased expression of KLF4α in pancreatic tumor tissue was inversely correlated with overall time of survival in patients with stage II pancreatic ductal adenocarcinoma. CONCLUSIONS: We identified a splice variant of KLF4 (KLF4α) that is up-regulated in aggressive pancreatic cancer cells and human pancreatic tumor tissues. Increased expression promotes growth of pancreatic tumors in mice and is associated with reduced survival times of patients.


Assuntos
Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Fatores de Transcrição Kruppel-Like/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Processamento Alternativo/fisiologia , Animais , Carcinoma Ductal Pancreático/mortalidade , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias Pancreáticas/mortalidade , Prognóstico , Análise de Sobrevida , Transplante Heterólogo , Regulação para Cima/fisiologia
9.
Sci Adv ; 7(5)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33571109

RESUMO

The invasiveness and high immune suppression of glioblastoma multiforme (GBM) produce poor survival of afflicted patients. Unfortunately, in the past decades, no therapeutic approach has remarkably improved the survival time of patients with GBM. Our analysis of the TCGA database and brain tumor tissue arrays indicated that CXCL1 and CXCL2 overexpression is closely associated with GBM's aggressiveness. Our results showed that elevation of CXCL1 or CXCL2 facilitated myeloid cell migration and simultaneously disrupted CD8+ T cell accumulation at tumor sites, causing accelerated tumor progression. Yet, blockade of CXCL1/2 significantly prevented myeloid-derived suppressor cell migration and thereby increased CD8+ T cell accumulation in vitro and in vivo. CXCL1/2 also promoted the paracrine factor S100A9 and further activated Erk1/2 and p70S60k, whereas blocking CXCL1/2 down-regulated these prosurvival factors. The combination of targeting CXCL1/2 and standard temozolomide chemotherapy improved upon the antitumor efficacy of chemotherapy alone, extending the overall survival time in GBM.


Assuntos
Neoplasias Encefálicas , Quimiocina CXCL1 , Quimiocina CXCL2 , Glioblastoma , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/imunologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Evasão Tumoral
10.
Clin Cancer Res ; 15(5): 1821-9, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19223493

RESUMO

PURPOSE: We determined the expression of mammalian target of rapamycin (mTOR) and its activated form, p-mTOR, in Chinese patients with gastric cancer and its clinical effects and underlying mechanisms. EXPERIMENTAL DESIGN: Tissue microarray blocks containing gastric cancer tissue and matched noncancer gastric tissue specimens obtained from 1,072 patients were constructed. Expression of total mTOR and p-mTOR in these specimens was analyzed using immunohistochemical studies and confirmed by Western blotting. RESULTS: The overall rates of total mTOR and p-mTOR overexpression were 50.8% (545 of 1,072) and 46.5% (499 of 1,072), respectively. The p-mTOR overexpression was significantly correlated with total mTOR overexpression. Overexpression of total mTOR protein was significantly correlated with tumor differentiation, T1/T2 tumors, and stage I/II/III disease, whereas p-mTOR overexpression was significantly correlated with lymph node metastasis and all stage disease. The Cox proportional hazards model revealed that the overexpression of p-mTOR, but not total mTOR, was an independent prognostic factor for gastric cancer. The overexpression of p-mTOR also predicted the angiogenic phenotype of human gastric cancer and regulated angiogenesis of gastric cancer cells. CONCLUSIONS: Increased activation of mTOR is frequent in human gastric cancer and overexpression of p-mTOR is an independent prognostic factor, suggesting that mTOR pathway could be a potential target for therapy of this malignancy.


Assuntos
Adenocarcinoma Mucinoso/metabolismo , Adenocarcinoma/metabolismo , Carcinoma Papilar/metabolismo , Carcinoma de Células em Anel de Sinete/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Quinases/metabolismo , Neoplasias Gástricas/metabolismo , Adenocarcinoma/secundário , Adenocarcinoma Mucinoso/secundário , Animais , Povo Asiático , Western Blotting , Carcinoma Papilar/secundário , Carcinoma de Células em Anel de Sinete/secundário , Células Cultivadas , Endotélio Vascular , Feminino , Humanos , Técnicas Imunoenzimáticas , Neoplasias Hepáticas/secundário , Metástase Linfática , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neovascularização Patológica , Fosforilação , Prognóstico , Neoplasias Gástricas/patologia , Taxa de Sobrevida , Serina-Treonina Quinases TOR , Análise Serial de Tecidos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cancer Res ; 80(20): 4500-4513, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32699135

RESUMO

Pancreatic cancer is a lethal disease owing to its intrinsic and acquired resistance to therapeutic modalities. The altered balance between pro- and antiapoptosis signals within cancer cells is critical to therapeutic resistance. However, the molecular mechanisms underlying increased antiapoptosis signals remain poorly understood. In this study, we report that PRMT1 expression is increased in pancreatic cancer tissues and is associated with higher tumor grade, increased aggressiveness, and worse prognosis. PRMT1 overexpression increased arginine methylation of HSPs of 70 kDa (HSP70); this methylation enhanced HSP70 binding and stabilization of BCL2 mRNA through AU-rich elements in 3'-untranslated region and consequentially increased BCL2 protein expression and protected cancer cells from apoptosis induced by cellular stresses and therapeutics. RNA binding and regulation function of HSP70 was involved in pancreatic cancer drug resistance and was dependent on protein arginine methylation. These findings not only reveal a novel PRMT1-HSP70-BCL2 signaling axis that is crucial to pancreatic cancer cell survival and therapeutic resistance, but they also provide a proof of concept that targeted inhibition of this axis may represent a new therapeutic strategy. SIGNIFICANCE: This study demonstrates that a PRMT1-mediated stabilization of BCL2 mRNA contributes to therapeutic resistance in pancreatic cancer and that targeting this pathway could overcome said resistance.


Assuntos
Carcinoma Ductal Pancreático/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/fisiologia , Proteínas de Choque Térmico HSP70/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/genética , Idoso , Animais , Arginina/metabolismo , Carcinoma Ductal Pancreático/mortalidade , Carcinoma Ductal Pancreático/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Proteínas de Choque Térmico HSP70/genética , Humanos , Masculino , Metilação , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Ligação Proteica , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Cancer Res ; 67(10): 4878-85, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17510417

RESUMO

The impact of antiangiogenic therapy on the Sp1/vascular endothelial growth factor (VEGF) pathway and that of alteration of Sp1 signaling on the efficacy of antiangiogenic therapy is unclear, yet understanding their interactions has significant clinical implications. Treatment with bevacizumab, a neutralizing antibody against VEGF, suppressed human pancreatic cancer growth in nude mice. Gene expression analyses revealed that this treatment substantially up-regulated the expression of Sp1 and its downstream target genes, including VEGF and epidermal growth factor receptor, in tumor tissues, whereas it did not have this effect on pancreatic cancer cells in culture. Treatment with mithramycin A, an Sp1 inhibitor, suppressed the expression of Sp1 and its downstream target genes in both cell culture and tumors growing in nude mice. Combined treatment with bevacizumab and mithramycin A produced synergistic tumor suppression, which was consistent with suppression of the expression of Sp1 and its downstream target genes. Thus, treatment with bevacizumab may block VEGF function but activate the pathway of its expression via positive feedback. Given the fact that Sp1 is an important regulator of the expression of multiple angiogenic factors, bevacizumab-initiated up-regulation of Sp1 and subsequent overexpression of its downstream target genes may profoundly affect the potential angiogenic phenotype and effectiveness of antiangiogenic strategies for human pancreatic cancer. Therefore, this study is the first to show the significance and clinical implications of alteration of Sp1 signaling in antiangiogenic therapy for pancreatic cancer and other cancers.


Assuntos
Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Anticorpos Monoclonais/farmacologia , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas/tratamento farmacológico , Plicamicina/análogos & derivados , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animais , Anticorpos Monoclonais Humanizados , Bevacizumab , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Plicamicina/farmacologia , Regiões Promotoras Genéticas , Fator de Transcrição Sp1/biossíntese , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Res ; 79(1): 72-85, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30425057

RESUMO

Aberrant activation of ß-catenin signaling is a critical driver for tumorigenesis, but the mechanism underlying this activation is not completely understood. In this study, we demonstrate a critical role of ß-catenin signaling in stabilization of enhancer of zeste homolog 2 (EZH2) and control of EZH2-mediated gene repression in oncogenesis. ß-Catenin/TCF4 activated the transcription of the deubiquitinase USP1, which then interacted with and deubiquitinated EZH2 directly. USP1-mediated stabilization of EZH2 promoted its recruitment to the promoters of CDKN1B, RUNX3, and HOXA5, resulting in enhanced enrichment of histone H3K27me3 and repression of target gene expression. In human glioma specimens, expression levels of nuclear ß-catenin, USP1, and EZH2 correlated with one another. Depletion of ß-catenin/USP1/EZH2 repressed glioma cell proliferation in vitro and tumor formation in vivo. Our findings indicate that a ß-catenin-USP1-EZH2 axis orchestrates the interplay between dysregulated ß-catenin signaling and EZH2-mediated gene epigenetic silencing during glioma tumorigenesis. SIGNIFICANCE: These findings identify the ß-catenin-USP1-EZH2 signaling axis as a critical mechanism for glioma tumorigenesis that may serve as a new therapeutic target in glioblastoma.


Assuntos
Carcinogênese/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/química , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Proteases Específicas de Ubiquitina/metabolismo , beta Catenina/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Proliferação de Células , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Glioma/genética , Glioma/metabolismo , Humanos , Camundongos , Camundongos Nus , Prognóstico , Estabilidade Proteica , Taxa de Sobrevida , Células Tumorais Cultivadas , Proteases Específicas de Ubiquitina/genética , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/genética
14.
Int J Oncol ; 33(1): 161-7, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18575762

RESUMO

Overexpression of the transcription factor Sp1 may play a critical role in human gastric cancer angiogenesis. In the present studies, we determined whether targeting Sp1 has a therapeutic benefit. Treatment with mithramycin A (MIT) suppressed the expression of Sp1 and its downstream target genes in both human gastric cancer cell culture and tumors growing in nude mice. The molecular responses were accompanied by a significant inhibition of gastric cancer angiogenesis, growth and metastasis. Conversely, treatment with bevacizumab (BVZ), a neutralizing antibody against VEGF A, suppressed human gastric cancer growth in nude mice in a dose-dependent manner. Gene expression analyses revealed that treatment with low dose of BVZ substantially upregulated the expression of Sp1 and its downstream target genes, including VEGF and EGFR, in tumor tissues, whereas it did not have this effect on gastric cancer cells in culture. Combined treatment with BVZ and MIT produced synergistic tumor suppression, which was consistent with suppression of the expression of Sp1 and its downstream target genes. Thus, treatment with BVZ may block VEGF function but activate the pathway of its expression via positive feedback. Collectively, Sp1 is an important regulator of the expression of multiple angiogenic factors and functional status of Sp1 signaling pathway may profoundly affect the angiogenic phenotype of and effectiveness of antiangiogenic strategies for human gastric cancer.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Neovascularização Patológica/prevenção & controle , Plicamicina/análogos & derivados , Fator de Transcrição Sp1/antagonistas & inibidores , Neoplasias Gástricas/terapia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Bevacizumab , Linhagem Celular Tumoral , Modelos Animais de Doenças , Sinergismo Farmacológico , Feminino , Humanos , Camundongos , Camundongos Nus , Metástase Neoplásica , Plicamicina/farmacologia , Plicamicina/uso terapêutico , Fator de Transcrição Sp1/fisiologia , Neoplasias Gástricas/irrigação sanguínea , Neoplasias Gástricas/patologia , Transcrição Gênica , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
15.
Int J Oncol ; 33(5): 979-84, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18949360

RESUMO

Recent studies demonstrated an epigenetic inactivation of the runt-related transcription factor 3 (RUNX3) gene in human colon cancer. However, it remains unclear whether RUNX3 is tumor suppressive in colon cancer and, if so, the underlying molecular mechanisms of this activity are still unknown. In the present study, we sought to determine the level of RUNX3 expression in human colon tumor specimens and used an animal model of colon cancer to determine the impact of RUNX3 expression on tumor growth and metastasis. First, we analyzed RUNX3 expression in 83 human colon tumor specimens using immunohistochemical, reverse transcriptase-polymerase chain reaction, and Western blot analysis. RUNX3 mRNA and protein expression levels were consistently lower in tumor tissue specimens than in matched normal colon tissue specimens. Also, restoration of RUNX3 expression in colon cancer cells using gene transfer inhibited colon tumor growth and metastasis in our animal model, which was consistent with inhibition of colon tumor growth in vitro. Collectively, our clinical and experimental data support the notion that RUNX3 is a tumor suppressor in human colon cancer.


Assuntos
Proliferação de Células , Neoplasias do Colo/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Idoso , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Epigênese Genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Metástase Neoplásica , RNA Mensageiro/metabolismo , Fatores de Tempo , Transfecção , Proteínas Supressoras de Tumor/genética
16.
Clin Cancer Res ; 12(21): 6386-94, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17085650

RESUMO

PURPOSE: Recent studies indicated that RUNX3 exhibits potent antitumor activity. However, the underlying molecular mechanisms of this activity remain unclear. In the present study, we used a gastric cancer model to determine the effect of RUNX3 expression on tumor angiogenesis. EXPERIMENTAL DESIGN: The effects of increased RUNX3 expression on vascular endothelial growth factor (VEGF) expression in and angiogenic potential of human gastric cancer cells were determined in vitro and in animal models. RUNX3 and VEGF expression was determined in 120 human gastric cancer specimens and their relationship was analyzed. RESULTS: RUNX3 gene transfer suppressed VEGF expression in human gastric cancer cells. Down-regulation of VEGF expression correlated with a significantly impaired angiogenic potential of human gastric cancer cells. Furthermore, RUNX3 restoration inhibited tumor growth and metastasis in animal models, which was consistent with inhibition of angiogenesis as determined by evaluating VEGF expression and tumor microvessel formation. In gastric cancer specimens, loss or decrease in RUNX3 expression inversely associated with increased VEGF expression and elevated microvessel formation. CONCLUSIONS: Our clinical and experimental data provide a novel molecular mechanism for the antitumor activity of RUNX3 and may help design effective therapy targeting RUNX3 pathway to control gastric cancer growth and metastasis.


Assuntos
Adenocarcinoma/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/biossíntese , Metástase Neoplásica , Neovascularização Patológica/metabolismo , Neoplasias Gástricas/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Adenocarcinoma/irrigação sanguínea , Animais , Western Blotting , Primers do DNA , Modelos Animais de Doenças , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , Neoplasias Gástricas/irrigação sanguínea , Transcrição Gênica , Transdução Genética , Fator A de Crescimento do Endotélio Vascular/genética
17.
Clin Cancer Res ; 12(21): 6395-402, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17085651

RESUMO

PURPOSE: Increasing evidence indicates that the transcription factor, Sp1, regulates the expression of multiple genes involved in tumor development and progression. We have recently reported that Sp1 overexpression is directly correlated with the angiogenic potential of and poor prognosis for human gastric cancer. However, the underlying mechanisms that result in Sp1 overexpression remain unclear. EXPERIMENTAL DESIGN: The expression of Sp1 and Krüppel-like factor 4 (KLF4), a potential tumor suppressor gene, in gastric cancer tissue was analyzed by immunohistochemistry and Western blot analysis. Alterations of Sp1 and KLF4 expression were achieved by gene transfer and verified by Northern and Western blot analyses. Furthermore, Sp1 promoter activity assay, electrophoretic mobility shift assay, and chromatin immunoprecipitation assay were done to identify the KLF4 binding sites on the Sp1 promoter. RESULTS: Mutually exclusive expression of Sp1 and KLF4 was evident in gastric cancer and noncancerous tissue. Specifically, strong Sp1 expression but loss of KLF4 expression was found in cancer tissue, whereas the adjacent noncancerous tissue showed negative Sp1 expression but strong KLF4 expression. Enforced KLF4 expression repressed Sp1 expression at the promoter activity, mRNA, and protein levels. Moreover, a region within the proximal Sp1 promoter was identified to have overlapping KLF4- and Sp1-binding sites, to which KLF4 and Sp1 compete for binding. Sp1 positively regulated its own promoter, whereas KLF4 did the opposite. CONCLUSIONS: Our data suggests that disruption of KLF4-mediated negative regulation contributes to the molecular events of Sp1 overexpression and to the development and progression of human gastric cancer.


Assuntos
Adenocarcinoma/metabolismo , Fatores de Transcrição Kruppel-Like/biossíntese , Fator de Transcrição Sp1/biossíntese , Neoplasias Gástricas/metabolismo , Sequência de Bases , Northern Blotting , Western Blotting , Linhagem Celular Tumoral , Primers do DNA , Ensaio de Desvio de Mobilidade Eletroforética , Expressão Gênica , Humanos , Imuno-Histoquímica , Imunoprecipitação , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Dados de Sequência Molecular , Regiões Promotoras Genéticas , RNA Interferente Pequeno , Fator de Transcrição Sp1/genética
18.
Clin Cancer Res ; 23(3): 687-696, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27449499

RESUMO

PURPOSE: Musashi 2 (MSI2) is reported to be a potential oncoprotein in cases of leukemia and several solid tumors. However, its expression, function, and regulation in pancreatic ductal adenocarcinoma (PDAC) cases have yet to be demonstrated. Therefore, in the current study, we investigated the clinical significance and biologic effects of MSI2 expression in PDAC cases and sought to delineate the clinical significance of the newly identified Krüppel-like factor 4 (KLF4)/MSI2 regulatory pathway. EXPERIMENTAL DESIGN: MSI2 expression and its association with multiple clinicopathologic characteristics in human PDAC specimens were analyzed immunohistochemically. The biological functions of MSI2 regarding PDAC cell growth, migration, invasion, and metastasis were studied using gain- and loss-of-function assays both in vitro and in vivo Regulation of MSI2 expression by KLF4 was examined in several cancer cell lines, and the underlying mechanisms were studied using molecular biologic methods. RESULTS: MSI2 expression was markedly increased in both PDAC cell lines and human PDAC specimens, and high MSI2 expression was associated with poor prognosis for PDAC. Forced MSI2 expression promoted PDAC proliferation, migration, and invasion in vitro and growth and metastasis in vivo, whereas knockdown of MSI2 expression did the opposite. Transcriptional inhibition of MSI2 expression by KLF4 occurred in multiple PDAC cell lines as well as mouse models of PDAC. CONCLUSIONS: Lost expression of KLF4, a transcriptional repressor of MSI2 results in overexpression of MSI2 in PDACs, which may be a biomarker for accurate prognosis. A dysregulated KLF4/MSI2 signaling pathway promotes PDAC progression and metastasis. Clin Cancer Res; 23(3); 687-96. ©2016 AACR.


Assuntos
Carcinoma Ductal Pancreático/patologia , Fatores de Transcrição Kruppel-Like/fisiologia , Metástase Neoplásica/fisiopatologia , Proteínas de Neoplasias/fisiologia , Neoplasias Pancreáticas/patologia , Proteínas de Ligação a RNA/fisiologia , Transdução de Sinais/fisiologia , Animais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/secundário , Linhagem Celular Tumoral , Movimento Celular , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Xenoenxertos , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Fatores de Transcrição Kruppel-Like/genética , Neoplasias Hepáticas/secundário , Camundongos , Camundongos Nus , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/metabolismo , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/genética , Organismos Livres de Patógenos Específicos , Transcrição Gênica , Ensaio Tumoral de Célula-Tronco
19.
Clin Cancer Res ; 23(18): 5585-5597, 2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28659310

RESUMO

Purpose: The dismal prognosis of pancreatic cancer has been linked to poor tumor differentiation. However, molecular basis of pancreatic cancer differentiation and potential therapeutic value of the underlying molecules remain unknown. We investigated the mechanistic underexpression of Krüppel-like factor 4 (KLF4) in pancreatic cancer and defined a novel epigenetic pathway of its activation for pancreatic cancer differentiation and treatment.Experimental Design: Expressions of KLF4 and DNMT1 in pancreatic cancer tissues were determined by IHC and the genetic and epigenetic alterations of KLF4 in and KLF4's impact on differentiation of pancreatic cancer were examined using molecular biology techniques. The function of dietary 3,3'-diindolylmethane (DIM) on miR-152/DNMT1/KLF4 signaling in pancreatic cancer was evaluated using both cell culture and animal models.Results: Overexpression of DNMT1 and promoter hypermethylation contributed to decreased KLF4 expression in and associated with poor differentiation of pancreatic cancer. Manipulation of KLF4 expression significantly affected differentiation marker expressions in pancreatic cancer cells. DIM treatment significantly induced miR-152 expression, which blocked DNMT1 protein expression and its binding to KLF4 promoter region, and consequently reduced promoter DNA methylation and activated KLF4 expression in pancreatic cancer cells. In addition, DIM treatment caused significant inhibition of cell growth in vitro and tumorigenesis in animal models of pancreatic cancer.Conclusions: This is the first demonstration that dysregulated KLF4 expression associates with poor differentiation of pancreatic cancer. Epigenetic activation of miR-152/DNMT1/KLF4 signaling pathway by dietary DIM causes differentiation and significant growth inhibition of pancreatic cancer cells, highlighting its translational implications for pancreatic and other cancers. Clin Cancer Res; 23(18); 5585-97. ©2017 AACR.


Assuntos
Desdiferenciação Celular/genética , DNA (Citosina-5-)-Metiltransferase 1/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Fatores de Transcrição Kruppel-Like/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Animais , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Modelos Animais de Doenças , Epigênese Genética , Feminino , Expressão Gênica , Genes Reporter , Xenoenxertos , Humanos , Indóis/farmacologia , Fator 4 Semelhante a Kruppel , Camundongos , MicroRNAs/genética , Gradação de Tumores , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo
20.
Cancer Res ; 77(24): 6851-6862, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29038348

RESUMO

The serine/threonine kinase STK33 has been implicated in cancer cell proliferation. Here, we provide evidence of a critical role for STK33 in the pathogenesis and metastatic progression of pancreatic ductal adenocarcinoma (PDAC). STK33 expression in PDAC was regulated by the hypoxia-inducible transcription factor HIF1α. In human PDAC specimens, STK33 was overexpressed and associated with poor prognosis. Enforced STK33 expression promoted PDAC proliferation, migration, invasion, and tumor growth, whereas STK33 depletion exerted opposing effects. Mechanistic investigations showed that HIF1α regulated STK33 via direct binding to a hypoxia response element in its promoter. In showing that dysregulated HIF1α/STK33 signaling promotes PDAC growth and progression, our results suggest STK33 as a candidate therapeutic target to improve PDAC treatment. Cancer Res; 77(24); 6851-62. ©2017 AACR.


Assuntos
Carcinoma Ductal Pancreático/patologia , Proliferação de Células/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Neoplasias Pancreáticas/patologia , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Carcinoma Ductal Pancreático/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Transdução de Sinais/genética , Neoplasias Pancreáticas
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa