Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Immunother Cancer ; 12(1)2024 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-38199607

RESUMEN

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.


Asunto(s)
Neoplasias Óseas , Fibroblastos Asociados al Cáncer , Osteosarcoma , Animales , Humanos , Interleucina-12 , Xenoinjertos , Osteosarcoma/terapia , Membrana Celular , Matriz Extracelular , Modelos Animales de Enfermedad , Neoplasias Óseas/terapia , Tratamiento Basado en Trasplante de Células y Tejidos
2.
Nat Commun ; 14(1): 735, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36759517

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos , Glioblastoma , Animales , Ratones , Encéfalo , Glioblastoma/terapia , Memoria Inmunológica , Células T de Memoria , Recurrencia Local de Neoplasia , Linfocitos T/inmunología
3.
Front Immunol ; 13: 952231, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36203573

RESUMEN

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.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Citocinas , Humanos , Interleucina-12 , Linfocitos T
4.
Clin Cancer Res ; 28(17): 3862-3873, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35727602

RESUMEN

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.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Neoplasias Óseas/terapia , Línea Celular Tumoral , Humanos , Inmunoterapia Adoptiva/métodos , Interleucina-12 , Leucocitos Mononucleares , Osteosarcoma/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Sci Adv ; 7(5)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33571109

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas , Quimiocina CXCL1 , Quimiocina CXCL2 , Glioblastoma , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/inmunología , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Temozolomida/farmacología , Temozolomida/uso terapéutico , Escape del Tumor
6.
Cancer Res ; 80(20): 4500-4513, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32699135

RESUMEN

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.


Asunto(s)
Carcinoma Ductal Pancreático/tratamiento farmacológico , Resistencia a Antineoplásicos/fisiología , Proteínas HSP70 de Choque Térmico/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/genética , Anciano , Animales , Arginina/metabolismo , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Proteínas HSP70 de Choque Térmico/genética , Humanos , Masculino , Metilación , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Unión Proteica , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Res ; 79(1): 72-85, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30425057

RESUMEN

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.


Asunto(s)
Carcinogénesis/patología , Proteína Potenciadora del Homólogo Zeste 2/química , Regulación Neoplásica de la Expresión Génica , Glioma/patología , Proteasas Ubiquitina-Específicas/metabolismo , beta Catenina/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética , Glioma/genética , Glioma/metabolismo , Humanos , Ratones , Ratones Desnudos , Pronóstico , Estabilidad Proteica , Tasa de Supervivencia , Células Tumorales Cultivadas , Proteasas Ubiquitina-Específicas/genética , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
8.
Cancer Res ; 77(24): 6851-6862, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29038348

RESUMEN

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.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Proliferación Celular/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neoplasias Pancreáticas/patología , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Carcinoma Ductal Pancreático/genética , Línea Celular Tumoral , Movimiento Celular/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Invasividad Neoplásica , Neoplasias Pancreáticas/genética , Transducción de Señal/genética , Neoplasias Pancreáticas
9.
Clin Cancer Res ; 23(18): 5585-5597, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28659310

RESUMEN

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.


Asunto(s)
Desdiferenciación Celular/genética , ADN (Citosina-5-)-Metiltransferasa 1/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Animales , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Modelos Animales de Enfermedad , Epigénesis Genética , Femenino , Expresión Génica , Genes Reporteros , Xenoinjertos , Humanos , Indoles/farmacología , Factor 4 Similar a Kruppel , Ratones , MicroARNs/genética , Clasificación del Tumor , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo
10.
Clin Cancer Res ; 23(3): 687-696, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27449499

RESUMEN

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.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Factores de Transcripción de Tipo Kruppel/fisiología , Metástasis de la Neoplasia/fisiopatología , Proteínas de Neoplasias/fisiología , Neoplasias Pancreáticas/patología , Proteínas de Unión al ARN/fisiología , Transducción de Señal/fisiología , Animales , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/secundario , Línea Celular Tumoral , Movimiento Celular , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Xenoinjertos , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Hepáticas/secundario , Ratones , Ratones Desnudos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/metabolismo , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Organismos Libres de Patógenos Específicos , Transcripción Genética , Ensayo de Tumor de Célula Madre
11.
Cancer Res ; 77(4): 874-885, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27923832

RESUMEN

The ETS family transcription factor ESE3 is a crucial element in differentiation and development programs for many epithelial tissues. Here we report its role as a tumor suppressor in pancreatic cancer. We observed drastically lower ESE3 expression in pancreatic ductal adenocarcinomas (PDAC) compared with adjacent normal pancreatic tissue. Reduced expression of ESE3 in PDAC correlated closely with an increase in lymph node metastasis and vessel invasion and a decrease in relapse-free and overall survival in patients. In functional experiments, downregulating the expression of ESE3 promoted PDAC cell motility and invasiveness along with metastasis in an orthotopic mouse model. Mechanistic studies in PDAC cell lines, the orthotopic mouse model, and human PDAC specimens demonstrated that ESE3 inhibited PDAC metastasis by directly upregulating E-cadherin expression at the level of its transcription. Collectively, our results establish ESE3 as a negative regulator of PDAC progression and metastasis by enforcing E-cadherin upregulation. Cancer Res; 77(4); 874-85. ©2016 AACR.


Asunto(s)
Cadherinas/fisiología , Carcinoma Ductal Pancreático/patología , Metástasis de la Neoplasia/prevención & control , Neoplasias Pancreáticas/patología , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Antígenos CD , Azacitidina/análogos & derivados , Azacitidina/farmacología , Cadherinas/genética , Carcinoma Ductal Pancreático/mortalidad , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Decitabina , Humanos , Ratones , Invasividad Neoplásica , Neoplasias Pancreáticas/mortalidad , Regiones Promotoras Genéticas , Factores de Transcripción/análisis , Factores de Transcripción/genética , Regulación hacia Arriba
12.
Cancer Cell ; 29(3): 324-338, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26977883

RESUMEN

Understanding the molecular mechanisms of tumor initiation has significant impact on early cancer detection and intervention. To define the role of KLF4 in pancreatic ductal adenocarcinoma (PDA) initiation, we used molecular biological analyses and mouse models of klf4 gain- and loss-of-function and mutant Kras. KLF4 is upregulated in and required for acinar-to-ductal metaplasia. Klf4 ablation drastically attenuates the formation of pancreatic intraepithelial neoplasia induced by mutant Kras(G12D), whereas upregulation of KLF4 does the opposite. Mutant KRAS and cellular injuries induce KLF4 expression, and ectopic expression of KLF4 in acinar cells reduces acinar lineage- and induces ductal lineage-related marker expression. These results demonstrate that KLF4 induces ductal identity in PanIN initiation and may be a potential target for prevention of PDA initiation.


Asunto(s)
Células Acinares/patología , Carcinogénesis/genética , Carcinogénesis/patología , Factores de Transcripción de Tipo Kruppel/metabolismo , Páncreas/patología , Conductos Pancreáticos/patología , Lesiones Precancerosas/genética , Células Acinares/metabolismo , Animales , Carcinogénesis/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Genes ras/genética , Humanos , Factor 4 Similar a Kruppel , Ratones , Páncreas/metabolismo , Conductos Pancreáticos/metabolismo , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Regulación hacia Arriba/genética
13.
Cancer Res ; 76(8): 2419-31, 2016 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-26880805

RESUMEN

KLF4 and CD44 regulate cancer cell stemness, but their precise functions and roles in metastatic progression are not well understood. In this study, we used both inducible and genetic engineering approaches to assess whether the activities of these two factors intersect in pancreatic cancer. We found that genetic ablation of Klf4 in pancreatic cancer cells isolated from Klf4(flox/flox) mice drastically increased CD44 expression and promoted the acquisition of stem-like properties, whereas tetracycline-inducible expression of KLF4 suppressed these properties in vitro and in vivo Further mechanistic investigation revealed that KLF4 bound to the CD44 promoter to negatively regulate transcription and also the expression of the CD44 variant. Moreover, in human pancreatic ductal adenocarcinoma (PDAC) tissues, the expression patterns of KLF4 and CD44 were mutually exclusive, and this inverse relationship was particularly striking in human metastatic pancreatic tumors and in autochthonous mouse models of PDAC. Taken together, our findings demonstrate that KLF4 acts as a tumor suppressor in PDAC cells that restricts metastatic behaviors through direct negative regulation of CD44, providing support for the clinical investigation of therapeutic approaches focusing on targeted KLF4 activation in advanced tumors. Cancer Res; 76(8); 2419-31. ©2016 AACR.


Asunto(s)
Receptores de Hialuranos/metabolismo , Factores de Transcripción de Tipo Kruppel/fisiología , Metástasis de la Neoplasia , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/patología , Transducción de Señal , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Receptores de Hialuranos/genética , Factor 4 Similar a Kruppel , Ratones , Transcripción Genética
14.
Clin Cancer Res ; 22(2): 502-12, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26338995

RESUMEN

PURPOSE: Tumor differentiation is a behavioral index for hepatocellular carcinoma (HCC) and a prognostic factor for patients with HCC who undergo orthotopic liver transplantation (OLT). However, the molecular basis for HCC differentiation and prognostic value of the underlying molecules that regulate HCC differentiation are unclear. In this study, we defined a potential driver pathway for HCC differentiation and prognostication. EXPERIMENTAL DESIGN: The regulation and function of Krüppel-like factor 4 (KLF4) and hepatocyte nuclear factor-6 (HNF-6) in HCC differentiation was evaluated using human tissues, molecular and cell biology, and animal models, and its prognostic significance was determined according to its impact on patient survival. RESULTS: There was a direct relationship between the expression levels of KLF4 and HNF6 in HCC. Reduced KLF4 or HNF6 expression correlated with high HCC grade. Poorly differentiated HCC cells had lower expression of KLF4 or HNF6 and differentiation-associated markers than did well-differentiated cells. Elevated KLF4 of HNF6 expression induced differentiation of poorly differentiated hepatoma cells. Mechanistically, KLF4 trans-activated HNF-6 expression. Restored HNF-6 expression upregulated expression of differentiation-associated markers and inhibited HCC cell migration and invasion, whereas HNF-6 knockdown did the opposite. Loss of KLF4 expression in primary HCC correlated with reduced overall survival and shortened relapse-free survival durations after OLT. Combination of KLF4 expression and the Milan criteria improved prognostication for HCC after OLT. CONCLUSIONS: The dysregulated KLF4/HNF-6 pathway drives dedifferentition and progression of HCC, and KLF4 is a biomarker for accurate prognostication of patients with HCC treated by OLT when integrated with the Milan Criteria.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Diferenciación Celular/genética , Factor Nuclear 6 del Hepatocito/genética , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Células Hep G2 , Humanos , Factor 4 Similar a Kruppel , Ratones , Recurrencia Local de Neoplasia/genética , Pronóstico , Regulación hacia Arriba/genética
15.
Cancer Res ; 75(22): 4778-4789, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26483206

RESUMEN

Merlin, the protein encoded by the NF2 gene, is a member of the band 4.1 family of cytoskeleton-associated proteins and functions as a tumor suppressor for many types of cancer. However, the roles and mechanism of Merlin expression in pancreatic cancer have remained unclear. In this study, we sought to determine the impact of Merlin expression on pancreatic cancer development and progression using human tissue specimens, cell lines, and animal models. Decreased expression of Merlin was pronounced in human pancreatic tumors and cancer cell lines. Functional analysis revealed that restored expression of Merlin inhibited pancreatic tumor growth and metastasis in vitro and in vivo. Furthermore, Merlin suppressed the expression of Wnt/ß-catenin signaling downstream genes and the nuclear expression of ß-catenin protein, and overexpression of Forkhead box M1 (FOXM1) attenuated the suppressive effect of Merlin on Wnt/ß-catenin signaling. Mechanistically, Merlin decreased the stability of FOXM1 protein, which plays critical roles in nuclear translocation of ß-catenin. Collectively, these findings demonstrated that Merlin critically regulated pancreatic cancer pathogenesis by suppressing FOXM1/ß-catenin signaling, suggesting that targeting novel Merlin/FOXM1/ß-catenin signaling is an effective therapeutic strategy for pancreatic cancer.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Neurofibromina 2/metabolismo , Neoplasias Pancreáticas/patología , Vía de Señalización Wnt/fisiología , Animales , Western Blotting , Femenino , Proteína Forkhead Box M1 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Ratones , Ratones Desnudos , Invasividad Neoplásica/patología , Neoplasias Pancreáticas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares , Transfección
16.
Oncotarget ; 6(34): 35949-63, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26416426

RESUMEN

Transcriptional co-activator with PDZ binding motif (TAZ) is a transducer of the Hippo pathway and promotes cancer development and progression. In the present study, we sought to determine the roles and underlying mechanisms of elevated expression and activation of TAZ in pancreatic cancer development and progression. The mechanistic role of TAZ and Hippo signaling in promotion of pancreatic cancer development and progression was examined using cell culture, molecular biology, and mouse models. The relevance of our experimental and mechanistic findings was validated using human pancreatic tumor specimens. We found that TAZ expression was markedly higher in pancreatic tumors than in normal pancreatic tissue. Further analysis of the correlation of TAZ expression with tissue microarray clinicopathologic parameters revealed that this expression was positively associated with tumor differentiation. Also, TAZ expression was higher in pancreatic cancer cell lines than in pancreatic ductal epithelial cells. TAZ activation in pancreatic cancer cells promoted their proliferation, migration, invasion, and epithelial-mesenchymal transition. Further mechanistic studies demonstrated that aberrant expression and activation of TAZ in pancreatic cancer cells resulted from suppression of the expression of Merlin, a positive regulator upstream of the Hippo pathway, and that the oncogenic function of TAZ in pancreatic cancer cells was mediated by TEA/ATTS domain transcription factors. Therefore, TAZ functioned as an oncogene and promoted pancreatic cancer epithelial-mesenchymal transition and progression. TAZ thus may be a target for effective therapeutic strategies for pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pancreáticas/patología , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Diferenciación Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Xenoinjertos , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Transducción de Señal , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Transfección
17.
Clin Cancer Res ; 21(4): 844-53, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25501129

RESUMEN

PURPOSE: Dysregulated signaling of nuclear transcription factors vitamin D receptor (VDR) and Forkhead box M1 (FOXM1) plays important roles in transformation and tumorigenesis. In this study, we sought to determine whether VDR signaling causally affected FOXM1 signaling in and pathogenesis of pancreatic ductal adenocarcinoma (PDAC). EXPERIMENTAL DESIGN: Genetic and pharmacologic approaches were used to manipulate VDR signaling. The impacts of altered VDR signaling on FOXM1 expression and function in PDAC cells were determined using molecular and biochemical methods, whereas those on PDAC cell biology and tumorigenicity were determined using in vitro and in vivo experimental systems. The clinical relevance of our findings was validated by analyzing human PDAC specimens. RESULTS: There was a striking inverse correlation between reduced expression of VDR and increased expression of FOXM1 in human PDAC cells and tissues. Treatment of PDAC cells with 1,25-dihydroxyvitamin D3 (1,25D), its synthetic analogue EB1089 (EB), and VDR transgenics drastically inhibited FOXM1 signaling and markedly suppressed tumor stemness, growth, and metastasis. Mechanistically, 1,25D and EB repressed FOXM1 transcription and reduced the expression level of nuclear FOXM1 protein. CONCLUSION: Inactivation of Vitamin D/VDR signaling is a critical contributor to PDAC development and progression via elevated expression and function of FOXM1 and enhanced PDAC cell stemness, invasion, and metastasis.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/patología , Receptores de Calcitriol/metabolismo , Animales , Células 3T3 BALB , Western Blotting , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación hacia Abajo , Citometría de Flujo , Proteína Forkhead Box M1 , Xenoinjertos , Humanos , Inmunohistoquímica , Ratones , Neoplasias Pancreáticas/metabolismo , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología , Análisis de Matrices Tisulares , Transfección
18.
Clin Cancer Res ; 20(16): 4370-80, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24947925

RESUMEN

PURPOSE: Krüppel-like factor 4 (KLF4) is a transcription factor and putative tumor suppressor. However, little is known about its effect on aerobic glycolysis in pancreatic tumors. Therefore, we investigated the clinical significance, biologic effects, and mechanisms of dysregulated KLF4 signaling in aerobic glycolysis in pancreatic cancer cells. EXPERIMENTAL DESIGN: Expression of KLF4 and lactate dehydrogenase A (LDHA) in 70 primary pancreatic tumors and 10 normal pancreatic tissue specimens was measured. Also, the underlying mechanisms of altered KLF4 expression and its impact on aerobic glycolysis in pancreatic cancer cells were investigated. RESULTS: We found a negative correlation between KLF4 and LDHA expression in pancreatic cancer cells and tissues and that their expression was associated with clinicopathologic features of pancreatic cancer. KLF4 underexpression and LDHA overexpression were correlated with disease stage and tumor differentiation. Experimentally, KLF4 overexpression significantly attenuated the aerobic glycolysis in and growth of pancreatic cancer cells both in vitro and in orthotopic mouse models, whereas knockdown of KLF4 expression had the opposite effect. Enforced KLF4 expression decreased LDHA expression, whereas small interfering RNA-mediated knockdown of KLF4 expression had the opposite effect. Mechanistically, KLF4 bound directly to the promoter regions of the LDHA gene and negatively regulated its transcription activity. CONCLUSIONS: Dysregulated signaling in this novel KLF4/LDHA pathway significantly impacts aerobic glycolysis in and development and progression of pancreatic cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Glucólisis , Factores de Transcripción de Tipo Kruppel/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Progresión de la Enfermedad , Femenino , Humanos , Técnicas para Inmunoenzimas , Isoenzimas/genética , Isoenzimas/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , L-Lactato Deshidrogenasa/genética , Lactato Deshidrogenasa 5 , Ratones , Ratones Desnudos , Estadificación de Neoplasias , Páncreas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Pronóstico , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
19.
Gastroenterology ; 147(2): 485-97.e18, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24859161

RESUMEN

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.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Factores de Transcripción Forkhead/metabolismo , MicroARNs/metabolismo , Neoplasias Pancreáticas/metabolismo , Transducción de Señal , Proteína Smad4/metabolismo , beta Catenina/metabolismo , Transporte Activo de Núcleo Celular , Animales , Antibióticos Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/secundario , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Femenino , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Persona de Mediana Edad , Invasividad Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Proteína Smad4/genética , Factores de Tiempo , Transfección , Regulación hacia Arriba , beta Catenina/genética , Gemcitabina
20.
Clin Cancer Res ; 20(10): 2595-606, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24634381

RESUMEN

PURPOSE: The transcription factor Forkhead box protein M1 (FOXM1) plays critical roles in cancer development and progression. However, the regulatory role and underlying mechanisms of FOXM1 in cancer metabolism are unknown. In this study, we characterized the regulation of aerobic glycolysis by FOXM1 and its impact on pancreatic cancer metabolism. EXPERIMENTAL DESIGN: The effect of altered expression of FOXM1 on expression of glycolytic enzymes and tumor development and progression was examined using animal models of pancreatic cancer. Also, the underlying mechanisms of altered pancreatic cancer glycolysis were analyzed using in vitro molecular biology. The clinical relevance of aberrant metabolism caused by dysregulated FOXM1 signaling was determined using pancreatic tumor and normal pancreatic tissue specimens. RESULTS: We found that FOXM1 did not markedly change the expression of most glycolytic enzymes except for phosphoglycerate kinase 1 (PGK-1) and lactate dehydrogenase A (LDHA). FOXM1 and LDHA were overexpressed concomitantly in pancreatic tumors and cancer cell lines. Increased expression of FOXM1 upregulated the expression of LDHA at both the mRNA and protein level and elevated LDH activity, lactate production, and glucose utilization, whereas reduced expression of FOXM1 did the opposite. Further studies demonstrated that FOXM1 bound directly to the LDHA promoter region and regulated the expression of the LDHA gene at the transcriptional level. Also, elevated FOXM1-LDHA signaling increased the pancreatic cancer cell growth and metastasis. CONCLUSIONS: Dysregulated expression and activation of FOXM1 play important roles in aerobic glycolysis and tumorigenesis in patients with pancreatic cancer via transcriptional regulation of LDHA expression.


Asunto(s)
Factores de Transcripción Forkhead/genética , Glucólisis/genética , L-Lactato Deshidrogenasa/genética , Neoplasias Pancreáticas/genética , Activación Transcripcional , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Ratones Desnudos , Metástasis de la Neoplasia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA