Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Mol Cancer Ther ; 18(2): 235-244, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30446586

RESUMO

BET inhibitors (BETi), which target transcription of key oncogenic genes, are currently being evaluated in early-phase clinical trials. However, because BETis show limited single-agent activity, there is increasing interest in identifying signaling pathways to enhance the efficacy of BETis. Here, we demonstrate increased MNK kinase-dependent eIF4E phosphorylation following treatment with BETis, indicating activation of a prosurvival feedback mechanism in response to BETis. BET PROTACs, which promote degradation of BET proteins, also induced eIF4E phosphorylation in cancer cells. Mechanistically, we show that the effect of BETis on MNK-eIF4E phosphorylation was mediated by p38 MAPKs. We also show that BETis suppressed RacGAP1 to induce Rac signaling-mediated eIF4E phosphorylation. Significantly, MNK inhibitors and MNK1/2 knockdown enhanced the efficacy of BETis in suppressing proliferation of cancer cells in vitro and in a syngeneic mouse model. Together, these results demonstrate a novel prosurvival feedback signaling induced by BETis, providing a mechanistic rationale for combination therapy with BET and MNK inhibitors for synergistic inhibition of cancer cells.


Assuntos
Acetanilidas/administração & dosagem , Compostos de Anilina/administração & dosagem , Azepinas/administração & dosagem , Fator de Iniciação 4E em Eucariotos/metabolismo , Compostos Heterocíclicos com 3 Anéis/administração & dosagem , Proteínas Serina-Treonina Quinases/metabolismo , Purinas/administração & dosagem , Neoplasias da Glândula Tireoide/tratamento farmacológico , Triazóis/administração & dosagem , Acetanilidas/farmacologia , Compostos de Anilina/farmacologia , Animais , Azepinas/farmacologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Fator de Iniciação 4E em Eucariotos/antagonistas & inibidores , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Camundongos , Fosforilação/efeitos dos fármacos , Purinas/farmacologia , Transdução de Sinais , Neoplasias da Glândula Tireoide/metabolismo , Triazóis/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Sci Rep ; 8(1): 13225, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30185888

RESUMO

The fibrotic reaction is a characteristic feature of human pancreatic ductal adenocarcinoma (PDAC) tumors. It is associated with activation and proliferation of pancreatic stellate cells (PSCs), which are key regulators of fibrosis in vivo. While there is increasing interest in the regulation of PD-L1 expression in cancer and immune cells, the expression and regulation of PD-L1 in other stromal cells, such as PSCs, has not been fully evaluated. Here we show that PSCs in vitro express higher PD-L1 mRNA and protein levels compared to the levels present in PDAC cells. We show that inhibitors targeting bromodomain and extra-terminal (BET) proteins and BRD4 knockdown decrease interferon-γ (IFN-γ)-induced PD-L1 expression in PSCs. We also show that c-MYC, one of the well-established targets of BET inhibitors, does not mediate IFN-γ-regulated PD-L1 expression in PSCs. Instead we show that interferon regulatory factor 1 (IRF1) mediates IFN-γ-induced PD-L1 expression in PSCs. Finally, while we show that BET inhibitors do not regulate IFN-γ-induced IRF1 expression in PSCs, BET inhibitors decrease binding of IRF1 and BRD4 to the PD-L1 promoter. Together, these results demonstrate the interplay between IRF1 and BRD4 in the regulation of PD-L1 in PSCs.


Assuntos
Antígeno B7-H1/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Fator Regulador 1 de Interferon/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/metabolismo , Células Estreladas do Pâncreas/metabolismo , Fatores de Transcrição/metabolismo , Antígeno B7-H1/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Fator Regulador 1 de Interferon/genética , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Células Estreladas do Pâncreas/patologia , Fatores de Transcrição/genética , Neoplasias Pancreáticas
3.
Nutrients ; 10(9)2018 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-30213082

RESUMO

Pancreatic cancer remains a daunting foe despite a vast number of accumulating molecular analyses regarding the mutation and expression status of a variety of genes. Indeed, most pancreatic cancer cases uniformly present with a mutation in the KRAS allele leading to enhanced RAS activation. Yet our understanding of the many epigenetic/environmental factors contributing to disease incidence and progression is waning. Epidemiologic data suggest that diet may be a key factor in pancreatic cancer development and potentially a means of chemoprevention at earlier stages. While diets high in ω3 fatty acids are typically associated with tumor suppression, diets high in ω6 fatty acids have been linked to increased tumor development. Thus, to better understand the contribution of these polyunsaturated fatty acids to pancreatic carcinogenesis, we modeled early stage disease by targeting mutant KRAS to the exocrine pancreas and administered diets rich in these fatty acids to assess tumor formation and altered cell-signaling pathways. We discovered that, consistent with previous reports, the ω3-enriched diet led to reduced lesion penetrance via repression of proliferation associated with reduced phosphorylated AKT (pAKT), whereas the ω6-enriched diet accelerated tumor formation. These data provide a plausible mechanism underlying previously observed effects of fatty acids and suggest that administration of ω3 fatty acids can reduce the pro-survival, pro-growth functions of pAKT. Indeed, counseling subjects at risk to increase their intake of foods containing higher amounts of ω3 fatty acids could aid in the prevention of pancreatic cancer.


Assuntos
Anticarcinógenos/administração & dosagem , Transformação Celular Neoplásica/metabolismo , Dieta , Ácidos Graxos Ômega-3/administração & dosagem , Neoplasias Experimentais/prevenção & controle , Ductos Pancreáticos/enzimologia , Neoplasias Pancreáticas/prevenção & controle , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Animais , Apoptose , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Dieta/efeitos adversos , Regulação para Baixo , Humanos , Camundongos Transgênicos , Mutação , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Ductos Pancreáticos/patologia , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Fosforilação , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo
4.
JCI Insight ; 2(3): e88032, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28194432

RESUMO

The fibrotic reaction, which can account for over 70%-80% of the tumor mass, is a characteristic feature of human pancreatic ductal adenocarcinoma (PDAC) tumors. It is associated with activation and proliferation of pancreatic stellate cells (PSCs), which are key regulators of collagen I production and fibrosis in vivo. In this report, we show that members of the bromodomain and extraterminal (BET) family of proteins are expressed in primary PSCs isolated from human PDAC tumors, with BRD4 positively regulating, and BRD2 and BRD3 negatively regulating, collagen I expression in primary cancer-associated PSCs. We show that the inhibitory effect of pan-BET inhibitors on collagen I expression in primary cancer-associated PSCs is through blocking of BRD4 function. Importantly, we show that FOSL1 is repressed by BRD4 in primary cancer-associated PSCs and negatively regulates collagen I expression. While BET inhibitors do not affect viability or induce PSC apoptosis or senescence, BET inhibitors induce primary cancer-associated PSCs to become quiescent. Finally, we show that BET inhibitors attenuate stellate cell activation, fibrosis, and collagen I production in the EL-KrasG12D transgenic mouse model of pancreatic tumorigenesis. Our results demonstrate that BET inhibitors regulate fibrosis by modulating the activation and function of cancer-associated PSCs.


Assuntos
Azepinas/administração & dosagem , Carcinoma Ductal Pancreático/tratamento farmacológico , Colágeno Tipo I/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/administração & dosagem , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Células Estreladas do Pâncreas/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Triazóis/administração & dosagem , Animais , Azepinas/farmacologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Colágeno Tipo I/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Camundongos , Proteínas Nucleares/antagonistas & inibidores , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Células Estreladas do Pâncreas/citologia , Células Estreladas do Pâncreas/metabolismo , Células Estreladas do Pâncreas/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Triazóis/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
5.
Oncotarget ; 8(54): 92667-92681, 2017 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-29190947

RESUMO

Peroxiredoxin-1 (Prdx1), a member of the thioredoxin (Txn) system, is overexpressed and correlates with poor prognosis in pancreatic cancer patients and can suppress Kras signaling through redox-mediated inhibition of ERK and AKT in lung and breast cancer. Its redox function is maintained by Txn and sulfiredoxin (Srxn), and its tumor promoting functions are activated by post-translational modification. We studied the role of the Txn system in pancreatic neoplasia and cancer by determining how it regulates the phosphorylation of Kras effectors and by determining its association with patient survival. We found that elevated Prdx1 nuclear localization significantly correlated with better patient survival. Our data also demonstrate that the expression of the Txn system is dysregulated, with elevated Prdx1 expression and significantly decreased Txn and Srxn expression in pancreatic lesions of targeted mutant Kras mouse models. This correlated with distinct differences in the interconversion of Prdx1 oligomers that affect its ability to regulate ERK and AKT phosphorylation. Our data also suggest that Prdx1 post-translational modification and oligomerization suppress Prdx1 mediated redox regulation of ERK phosphorylation. We observed distinct differences in Txn expression and in the ability of pTyr-Prdx1 to bind to pERK in a PanIN model of pancreatic neoplasia as compared to an IPMN model, indicating a distinct difference in the function of post-translationally modified Prdx1 in cells with less Txn expression. Modified Txn system function and post-translational regulation may therefore play a significant role in pancreatic tumorigenesis by altering Kras effector phosphorylation and inhibiting the tumor suppressive redox functions of Prdx1.

6.
Oncotarget ; 8(3): 3826-3839, 2017 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-27270652

RESUMO

TGFß has both tumor suppressive and tumor promoting effects in colon cancer. Also, TGFß can affect the extent and composition of inflammatory cells present in tumors, contextually promoting and inhibiting inflammation. While colon tumors display intratumoral inflammation, the contributions of TGFß to this process are poorly understood. In human patients, we found that epithelial loss of TGFß signaling was associated with increased inflammatory burden; yet overexpression of TGFß was also associated with increased inflammation. These findings were recapitulated in mutant APC models of murine tumorigenesis, where epithelial truncation of TGFBR2 led to lethal inflammatory disease and invasive colon cancer, mediated by IL8 and TGFß1. Interestingly, mutant APC mice with global suppression of TGFß signals displayed an intermediate phenotype, presenting with an overall increase in IL8-mediated inflammation and accelerated tumor formation, yet with a longer latency to the onset of disease observed in mice with epithelial TGFBR-deficiency. These results suggest that the loss of TGFß signaling, particularly in colon epithelial cells, elicits a strong inflammatory response and promotes tumor progression. This implies that treating colon cancer patients with TGFß inhibitors may result in a worse outcome by enhancing inflammatory responses.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Neoplasias do Colo/patologia , Citocinas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Neoplasias Experimentais , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad4/metabolismo
7.
Sci Rep ; 6: 29133, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27364947

RESUMO

Cells in the pancreas that have undergone acinar-ductal metaplasia (ADM) can transform into premalignant cells that can eventually become cancerous. Although the epithelial-mesenchymal transition regulator Snail (Snai1) can cooperate with Kras in acinar cells to enhance ADM development, the contribution of Snail-related protein Slug (Snai2) to ADM development is not known. Thus, transgenic mice expressing Slug and Kras in acinar cells were generated. Surprisingly, Slug attenuated Kras-induced ADM development, ERK1/2 phosphorylation and proliferation. Co-expression of Slug with Kras also attenuated chronic pancreatitis-induced changes in ADM development and fibrosis. In addition, Slug attenuated TGF-α-induced acinar cell metaplasia to ductal structures and TGF-α-induced expression of ductal markers in ex vivo acinar explant cultures. Significantly, blocking the Rho-associated protein kinase ROCK1/2 in the ex vivo cultures induced expression of ductal markers and reversed the effects of Slug by inducing ductal structures. In addition, blocking ROCK1/2 activity in Slug-expressing Kras mice reversed the inhibitory effects of Slug on ADM, ERK1/2 phosphorylation, proliferation and fibrosis. Overall, these results increase our understanding of the role of Slug in ADM, an early event that can eventually lead to pancreatic cancer development.


Assuntos
Metaplasia/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Fatores de Transcrição da Família Snail/genética , Células Acinares/patologia , Animais , Transformação Celular Neoplásica/genética , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/genética , Humanos , Metaplasia/patologia , Camundongos , Camundongos Transgênicos , Ductos Pancreáticos/metabolismo , Ductos Pancreáticos/patologia , Neoplasias Pancreáticas/patologia , Pancreatite Crônica/genética , Pancreatite Crônica/patologia , Transdução de Sinais/genética
8.
Cancer Res ; 76(9): 2525-39, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26980767

RESUMO

In early pancreatic carcinogenesis, TGFß acts as a tumor suppressor due to its growth-inhibitory effects in epithelial cells. However, in advanced disease, TGFß appears to promote tumor progression. Therefore, to better understand the contributions of TGFß signaling to pancreatic carcinogenesis, we generated mouse models of pancreatic cancer with either epithelial or systemic TGFBR deficiency. We found that epithelial suppression of TGFß signals facilitated pancreatic tumorigenesis, whereas global loss of TGFß signaling protected against tumor development via inhibition of tumor-associated fibrosis, stromal TGFß1 production, and the resultant restoration of antitumor immune function. Similarly, TGFBR-deficient T cells resisted TGFß-induced inactivation ex vivo, and adoptive transfer of TGFBR-deficient CD8(+) T cells led to enhanced infiltration and granzyme B-mediated destruction of developing tumors. These findings paralleled our observations in human patients, where TGFß expression correlated with increased fibrosis and associated negatively with expression of granzyme B. Collectively, our findings suggest that, despite opposing the proliferation of some epithelial cells, TGFß may promote pancreatic cancer development by affecting stromal and hematopoietic cell function. Therefore, the use of TGFBR inhibition to target components of the tumor microenvironment warrants consideration as a potential therapy for pancreatic cancer, particularly in patients who have already lost tumor-suppressive TGFß signals in the epithelium. Cancer Res; 76(9); 2525-39. ©2016 AACR.


Assuntos
Carcinogênese/metabolismo , Neoplasias Pancreáticas/patologia , Fator de Crescimento Transformador beta/metabolismo , Evasão Tumoral/fisiologia , Microambiente Tumoral/fisiologia , Transferência Adotiva , Animais , Western Blotting , Carcinogênese/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Fibrose/metabolismo , Fibrose/patologia , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Transdução de Sinais/fisiologia
9.
Oncotarget ; 7(19): 28218-34, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-27058416

RESUMO

Pancreatic cancer is characterized by a pronounced fibro-inflammatory reaction that has been shown to contribute to cancer progression. Previous reports have demonstrated that pigment epithelium-derived factor (PEDF) has potent tumor suppressive effects in pancreatic cancer, though little is known about the mechanisms by which PEDF limits pancreatic tumorigenesis. We therefore employed human specimens, as well as mouse and in vitro models, to explore the effects of PEDF upon the pancreatic microenvironment. We found that PEDF expression is decreased in human pancreatic cancer samples compared to non-malignant tissue. Furthermore, PEDF-deficient patients displayed increased intratumoral inflammation/fibrosis. In mice, genetic ablation of PEDF increased cerulein-induced inflammation and fibrosis, and similarly enhanced these events in the background of oncogenic KRAS. In vitro, recombinant PEDF neutralized macrophage migration as well as inhibited macrophage-induced proliferation of tumor cells. Additionally, recombinant PEDF suppressed the synthesis of pro-inflammatory/pro-fibrotic cytokines both in vivo and in vitro, and reduced collagen I deposition and TGFß synthesis by pancreatic stellate cells, consistent with reduced fibrosis. Combined, our results demonstrate that PEDF limits pancreatic cancer progression by attenuating the fibro-inflammatory reaction, and makes restoration of PEDF signaling a potential therapeutic approach to study in pancreatic cancer.


Assuntos
Carcinogênese/metabolismo , Proteínas do Olho/metabolismo , Fatores de Crescimento Neural/metabolismo , Neoplasias Pancreáticas/patologia , Serpinas/metabolismo , Animais , Carcinogênese/patologia , Progressão da Doença , Fibrose/metabolismo , Fibrose/patologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Pancreáticas/metabolismo , Microambiente Tumoral/fisiologia
10.
Pancreas ; 44(6): 882-7, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26166469

RESUMO

OBJECTIVE: We describe the first mouse model of pancreatic intraepithelial neoplasia (PanIN) lesions induced by alcohol in the presence and absence of chronic pancreatitis. METHODS: Pdx1-Cre;LSL-K-ras mice were exposed to Lieber-DeCarli alcohol diet for 6 weeks with cerulein injections. The PanIN lesions and markers of fibrosis, inflammation, histone deacetylation, epithelial-to-mesenchymal transition (EMT), and cancer stemness were measured by immunohistochemistry and Western. RESULTS: Exposure of Pdx1-Cre;LSL-K-ras mice to an alcohol diet significantly stimulated fibrosis and slightly but not significantly increased the level of PanIN lesions associated with an increase in tumor-promoting M2 macrophages. Importantly, the alcohol diet did not increase activation of stellate cells. Alcohol diet and cerulein injections resulted in synergistic and additive effects on PanIN lesion and M2 macrophage phenotype induction, respectively. Cerulein pancreatitis caused stellate cell activation, EMT, and cancer stemness in the pancreas. Pancreatitis caused histone deacetylation, which was promoted by the alcohol diet. Pancreatitis increased EMT and cancer stemness markers, which were not further affected by the alcohol diet. CONCLUSIONS: The results suggest that alcohol has independent effects on promotion of PDAC associated with fibrosis formed through a stellate cell-independent mechanism and that it further promotes early PDAC and M2 macrophage induction in the context of chronic pancreatitis.


Assuntos
Carcinoma in Situ/patologia , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Pancreatite Alcoólica/patologia , Pancreatite Crônica/patologia , Pancreatite/patologia , Acetilação , Doença Aguda , Animais , Carcinoma in Situ/induzido quimicamente , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Ceruletídeo , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Etanol , Fibrose , Histonas/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos Transgênicos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Pâncreas/metabolismo , Neoplasias Pancreáticas/induzido quimicamente , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Células Estreladas do Pâncreas/metabolismo , Células Estreladas do Pâncreas/patologia , Pancreatite/induzido quimicamente , Pancreatite/genética , Pancreatite/metabolismo , Pancreatite Alcoólica/genética , Pancreatite Alcoólica/metabolismo , Pancreatite Crônica/induzido quimicamente , Pancreatite Crônica/genética , Pancreatite Crônica/metabolismo , Fatores de Tempo
11.
Front Physiol ; 5: 464, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25538623

RESUMO

The study of pancreatic cancer has prompted the development of numerous mouse models that aim to recapitulate the phenotypic and mechanistic features of this deadly malignancy. This review accomplishes two tasks. First, it provides an overview of the models that have been used as representations of both the neoplastic and carcinoma phenotypes. Second, it presents new modeling schemes that ultimately will serve to more faithfully capture the temporal and spatial progression of the human disease, providing platforms for improved understanding of the role of non-epithelial compartments in disease etiology as well as evaluating therapeutic approaches.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA