Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Cancer Res ; 20(9): 1379-1390, 2022 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-35612556

RESUMO

Small cell lung cancer (SCLC) is a recalcitrant malignancy defined by subtypes on the basis of differential expression of the ASCL1, NEUROD1, and POU2F3 transcription factors. The MUC1-C protein is activated in pulmonary epithelial cells by exposure to environmental carcinogens and promotes oncogenesis; however, there is no known association between MUC1-C and SCLC. We report that MUC1-C is expressed in classic neuroendocrine (NE) SCLC-A, variant NE SCLC-N and non-NE SCLC-P cells and activates the MYC pathway in these subtypes. In SCLC cells characterized by NE differentiation and DNA replication stress, we show that MUC1-C activates the MYC pathway in association with induction of E2F target genes and dysregulation of mitotic progression. Our studies further demonstrate that the MUC1-C→MYC pathway is necessary for induction of (i) NOTCH2, a marker of pulmonary NE stem cells that are the proposed cell of SCLC origin, and (ii) ASCL1 and NEUROD1. We also show that the MUC1-C→MYC→NOTCH2 network is necessary for self-renewal capacity and tumorigenicity of NE and non-NE SCLC cells. Analyses of datasets from SCLC tumors confirmed that MUC1 expression in single SCLC cells significantly associates with activation of the MYC pathway. These findings demonstrate that SCLC cells are addicted to MUC1-C and identify a potential new target for SCLC treatment. IMPLICATIONS: This work uncovers addiction of SCLC cells to MUC1-C, which is a druggable target that could provide new opportunities for advancing SCLC treatment.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Células Neuroendócrinas , Carcinoma de Pequenas Células do Pulmão , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Mucina-1/genética , Mucina-1/metabolismo , Células Neuroendócrinas/patologia , Proteínas Oncogênicas/genética , Carcinoma de Pequenas Células do Pulmão/genética
2.
Clin Transl Gastroenterol ; 13(5): e00487, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35351843

RESUMO

INTRODUCTION: The ability of carbohydrate antigen 19-9 (CA19-9) to differentiate pancreatic cancer from other benign pancreatic lesions is unsatisfactory. This study explored the diagnostic value of KRAS gene mutations and plasma circulating tumor DNA (ctDNA) in patients with pancreatic cancer. METHODS: The prospective cohort study comprised 149 consecutive patients with solid pancreatic lesions who underwent endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA). KRAS subtype mutations were analyzed by digital droplet PCR (ddPCR) in EUS-FNA histopathology tissue samples, and blood samples were sent for plasma ctDNA analysis. The final diagnosis was based on surgical resection pathology or follow-up for at least 2 years. RESULTS: Adding KRAS mutation ddPCR increased the sensitivity and accuracy of EUS-FNA from 71.4% to 91.6% (P < 0.001) and 75.8% to 88.6% (P < 0.001), respectively. By comparison, the sensitivities of circulating biomarkers ctDNA and CA19-9 were only 35.2% and 71.2%. The area under the curve of the receiver operating characteristic curve (AUC) of EUS-FNA and KRAS ddPCR combination was >0.90 for distinguishing pancreatic cancer from benign lesions, whereas the AUC of EUS-FNA and CA19-9 combination was 0.83. The median survival time was significantly shorter in patients with G12D KRAS mutations than that in patients with other mutations (180 vs 240 days, P < 0.001). DISCUSSION: FNA tissue sample KRAS mutation analysis in tissues significantly improves the diagnostic accuracy of cyto/histopathological evaluation in EUS-FNA samples. The combination of EUS-FNA and tissue sample KRAS ddPCR provided a more accurate method for pancreatic cancer diagnosis, superior to the combination of EUS-FNA and CA19-9/ctDNA. G12D KRAS mutations in pancreatic cancer were independently associated with poor overall survival.


Assuntos
DNA Tumoral Circulante , Neoplasias Pancreáticas , Antígeno CA-19-9 , DNA Tumoral Circulante/genética , Aspiração por Agulha Fina Guiada por Ultrassom Endoscópico , Humanos , Mutação , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Prognóstico , Estudos Prospectivos , Proteínas Proto-Oncogênicas p21(ras)/genética , Neoplasias Pancreáticas
3.
Carcinogenesis ; 43(1): 67-76, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-34657147

RESUMO

Pancreatic ductal adenocarcinomas (PDAC) and poorly differentiated pancreatic neuroendocrine (NE) carcinomas are KRAS mutant malignancies with a potential common cell of origin. PDAC ductal, but not NE, lineage traits have been associated with cell-intrinsic activation of interferon (IFN) pathways. The present studies demonstrate that the MUC1 C-terminal subunit (MUC1-C), which evolved to protect mammalian epithelia from loss of homeostasis, is aberrantly overexpressed in KRAS mutant PDAC tumors and cell lines. We show that MUC1-C is necessary for activation of the type I and II IFN pathways and for expression of the Yamanaka OCT4, SOX2, KLF4 and MYC (OSKM) pluripotency factors. Our results demonstrate that MUC1-C integrates IFN signaling and pluripotency with NE dedifferentiation by forming a complex with MYC and driving the (i) achaete-scute homolog 1 and BRN2/POU3F2 neural, and (ii) NOTCH1/2 stemness transcription factors. Of translational relevance, targeting MUC1-C genetically and pharmacologically in PDAC cells (i) suppresses OSKM, NE dedifferentiation and NOTCH1/2, and (ii) inhibits self-renewal capacity and tumorigenicity. In PDAC tumors, we show that MUC1 significantly associates with activation of IFN signaling, MYC and NOTCH, and that upregulation of the MUC1-C → MYC pathway confers a poor prognosis. These findings indicate that MUC1-C dictates PDAC NE lineage specification and is a potential target for the treatment of recalcitrant pancreatic carcinomas with NE dedifferentiation.


Assuntos
Adenocarcinoma/genética , Carcinoma Ductal Pancreático/genética , Mucina-1/genética , Células Neuroendócrinas/patologia , Neoplasias Pancreáticas/genética , Adenocarcinoma/patologia , Animais , Carcinogênese/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos Nus , Células-Tronco Neoplásicas/patologia , Neoplasias Pancreáticas/patologia , Transdução de Sinais/genética , Neoplasias Pancreáticas
4.
Cancer Cell Int ; 21(1): 670, 2021 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-34906136

RESUMO

BACKGROUND: Owing to the lack of effective treatment options, early metastasis remains the major cause of pancreatic ductal adenocarcinoma (PDAC) recurrence and mortality. However, the molecular mechanism of early metastasis is largely unknown. We characterized the function of eukaryotic translation initiation factors (eIFs) in epithelial-mesenchymal-transition (EMT) and metastasis in pancreatic cancer cells to investigate whether eIFs and downstream c-MYC affect EMT and metastasis by joint interference. METHODS: We used The Cancer Genome Atlas (TCGA) and Genome Tissue Expression (GTEx) databases to analyze eIF4A1 expression in PDAC tissues and further validated the findings with a microarray containing 53 PDAC samples. Expression regulation and pharmacological inhibition of eIF4A1 and c-MYC were performed to determine their role in migration, invasion, and metastasis in pancreatic cancer cells in vitro and in vivo. RESULTS: Elevated eIF4A1 expression was positively correlated with lymph node infiltration, tumor size, and indicated a poor prognosis. eIF4A1 decreased E-cadherin expression through the c-MYC/miR-9 axis. Loss of eIF4A1 and c-MYC decreased the EMT and metastasis capabilities of pancreatic cancer cells, whereas upregulation of eIF4A1 attenuated the inhibition of EMT and metastasis induced by c-MYC downregulation. Treatment with the eIF4A1 inhibitor rocaglamide (RocA) or the c-MYC inhibitor Mycro3 either alone or in combination significantly decreased the expression level of EMT markers in pancreatic cancer cells in vitro. However, the efficiency and safety of RocA alone were not inferior to those of the combination treatment in vivo. CONCLUSION: Overexpression of eIF4A1 downregulated E-cadherin expression through the c-MYC/miR-9 axis, which promoted EMT and metastasis of pancreatic cancer cells. Despite the potential feedback loop between eIF4A1 and c-MYC, RocA monotherapy is a promising treatment inhibiting eIF4A1-induced PDAC metastasis.

5.
Oncol Rep ; 45(1): 230-238, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33416145

RESUMO

Induction of the apoptosis of tumor cells is a promising therapeutic approach for the treatment of cancer. Tumor necrosis factor­related apoptosis­inducing ligand (TRAIL) is a novel type of anticancer drug. However, gallbladder cancer cells (GBC) exhibit strong resistance to TRAIL. The aim of the present study was to assess the effect of rocaglate CR­1­31B (CR­31), an inhibitor of eukaryotic translation initiation factor 4A (eIF4A), on the sensitization of cells to TRAIL­induced apoptosis in TRAIL­resistant GBC. eIF4A was highly abundant in GBC tissues and cell lines (GBC­SD and SGC­996). GBC cells were treated using TRAIL and/or CR­31 and then apoptosis and TRAIL signaling were detected in vitro. CR­31 enhanced the sensitivity of TRAIL­resistant GBC cells, due to the CR­31­mediated eIF4A translational downregulation of c­FLIP and the subsequent activation of the caspase cascade. Furthermore, GBC­SD tumor xenografts models were established and the effects of CR­31 in vivo were assessed. CR­31 significantly reduced the growth and initiated the apoptosis of tumor cells, suggesting that CR­31 also increased sensitivity in vivo. Taken together, the results of the present study show that CR­31 treatment countered the resistance to TRAIL in GBC cells in vitro and in vivo. Therefore, eIF4A may serve as a novel therapeutic target and its combination with TRAIL­CR­31 as a therapy may serve as a novel strategy for GBC treatment.


Assuntos
Benzofuranos/farmacologia , Fator de Iniciação 4A em Eucariotos/antagonistas & inibidores , Neoplasias da Vesícula Biliar/tratamento farmacológico , Ácidos Hidroxâmicos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Benzofuranos/uso terapêutico , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Regulação para Baixo/efeitos dos fármacos , Fator de Iniciação 4A em Eucariotos/genética , Fator de Iniciação 4A em Eucariotos/metabolismo , Neoplasias da Vesícula Biliar/genética , Neoplasias da Vesícula Biliar/patologia , Técnicas de Silenciamento de Genes , Humanos , Ácidos Hidroxâmicos/uso terapêutico , Masculino , Camundongos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Exp Ther Med ; 14(5): 4839-4845, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29201188

RESUMO

Obstructive jaundice, owing to biliary obstruction, has been illustrated to trigger various biochemical, histological and immunological changes, leading to liver damage or even failure. The detailed molecular mechanism of simvastatin (Sim) involvement in liver regeneration during obstructive jaundice progression remains poorly elucidated. In the present study, an acute obstructive jaundice rat model was established by ligation and division of common bile duct, which was used to investigate the effects of Sim as a hepatoprotective treatment. Male Sprague-Dawley rats were randomly divided into four groups: Sham-operated, bile duct ligation (BDL) plus saline treatment [0.02 mg/kg/d, intraperitoneally (i.p.)], BDL plus low-dose Sim treatment (0.02 mg/kg, i.p.) and BDL plus high-dose Sim treatment (0.2 mg/kg, i.p.). During this experiment, the BDL+normal saline (NS) group demonstrated increased levels of transforming growth factor-ß1 (TGF-ß1) expression. Furthermore, Sim-treated animals demonstrated significantly downregulated TGF-ß1 expression and improved liver function vs. the BDL+NS group, indicating a TGF-ß1 antagonizing function. Additionally, Sim increased hepatocyte DNA synthesis in BDL rats compared to both the BDL+NS and Sham group. Apoptosis was increased in BDL+NS compared to the Sham group, and Sim markedly reduced hepatocyte apoptosis in the BDL group. Moreover, analysis of TGF-ß1 signaling pathways demonstrated that there was an increased hepatic TGF-ß1 and Smad3 expression in the BDL group, which was attenuated in the presence of Sim. In contrast to TGF-ß1, Sim induced the activity of the Smad7 (an inhibitor of TGF-ß1 signaling) mRNA and Smad7 protein expression. Sim displays hepatoprotective effects in liver cells via the upregulation of Smad7 expression and impaired TGF-ß signaling. Furthermore, the observations of the present study may provide evidence on the mechanism behind Sim blunting TGF-ß1 signaling, which is used to ameliorate the complication of liver damage and reduce the mortality rates associated with obstructive jaundice.

7.
Org Lett ; 19(14): 3931-3934, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28696714

RESUMO

A novel braided hetero[2](3)rotaxane is demonstrated by integrating the braided structure and mechanically interlocked rotaxane, in which a heterotritopic linear tris(dialkylammonium) guest penetrates a heterotritopic tris(crown ether) host, resulting in the formation of braided pseudohetero[2](3)rotaxane with different crossing and threading points. Then a braided hetero[2](3)rotaxane is constructed through the "CuAAC" click reaction.

8.
Org Lett ; 19(7): 1618-1621, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28304173

RESUMO

The successful triggering of ring-shuttling motion between two stations in a [2]rotaxane is demonstrated by employing a photochemical bond-cleavage strategy. A photolabile bulk barrier is covalently introduced into two identical stations of the thread to prevent dynamic shuttling of the macrocycle, resulting in a "gated" state. Irradiation of UV light (λ = 365 nm) results in the complete removal of the bulk barrier and the balanced shuttling motion of the macrocycle, indicating an "open" state of the rotaxane. In addition, the process from the "open" rotaxane to the "gated" rotaxane was executed by a chemical-rebonding method.

9.
J Exp Clin Cancer Res ; 35: 68, 2016 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-27084680

RESUMO

BACKGROUND: Prohibitin (PHB), a pleiotropic protein overexpressed in several tumor types, has been implicated in the regulation of cell proliferation, invasive migration and survival. However, PHB expression and its biological function in gallbladder cancer (GBC) remain largely unknown. METHODS: PHB and p-ERK protein expressions were determined in human GBC tissues by immunohistochemistry (IHC). The effects of PHB knockdown on GBC cell proliferation and invasiveness were evaluated using Cell Counting Kit-8 (CCK-8) cell viability, cell cycle analysis, transwell invasion and gelatin zymography assays. Subcutaneous xenograft and tail vein-lung metastasis tumor models in nude mice were employed to further substantiate the role of PHB in GBC progression. RESULTS: PHB protein was overexpressed in GBC tissues and was significantly associated with histological grade, tumor stage and perineural invasion. Furthermore, PHB expression was negatively associated with overall survival in GBC patients. In vitro experimental studies demonstrated that the downregulation of PHB expression by lentivirus-mediated shRNA interference not only inhibited the ERK pathway activation but also reduced the proliferative and invasive capacities of GBC cells. Moreover, PD0325901, a specific inhibitor of MEK, markedly impaired PHB- mediated phosphorylation of ERK protein. IHC statistical analyses further validated that PHB expression was positively correlated with ERK protein phosphorylation levels in GBC tissue samples. In vivo, PHB depletion also resulted in dramatic reductions in the growth and metastasis of GBC cells. CONCLUSION: Our findings demonstrate that PHB overexpression predicts poor survival in GBC patients. PHB could serve as a novel prognostic biomarker and a potential therapeutic target for GBCs.


Assuntos
Neoplasias da Vesícula Biliar/patologia , Sistema de Sinalização das MAP Quinases , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Regulação para Cima , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Neoplasias da Vesícula Biliar/genética , Neoplasias da Vesícula Biliar/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Transplante de Neoplasias , Fosforilação , Prognóstico , Proibitinas , Análise de Sobrevida
10.
Int J Mol Med ; 37(3): 631-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26846204

RESUMO

Ectopic apoptosis of vascular cells plays a critical role in the early stage development of diabetic retinopathy (DR). Uncoupling protein 2 (UCP2) is a mitochondrial modulator which protects against endothelial dysfunction. However, the role which UCP2 plays in endothelial apoptosis and its association with DR was unclear. In the present study, we investigated whether UCP2 functioned as an inhibitor of DR in endothelial cells. Firstly, we noted that in UCP2­knockout mice retinal cell death and damage in vivo was similar to that of db/db diabetic mice. Additionally, UCP2 knockdown induced caspase-3 activation and exaggerated high glucose (HG)-induced apoptosis of human umbilical vein endothelial cells (HUVECs). Conversely, adenovirus-mediated UCP2 overexpression inhibited the apoptosis of HUVECs and HG-induced caspase-3 activation. Furthermore, HG treatment resulted in the opening of the permeability transition pore (PTP) and liberation of cytochrome c from mitochondria to the cytosol in HUVECs. Notably, UCP2 overexpression inhibited these processes. Furthermore, adenovirus-mediated UCP2 overexpression led to a significant increase in intracellular nitric oxide (NO) levels and a decrease in reactive oxygen species (ROS) generation in HUVECs. Collectively, these data suggest that UCP2 plays an anti-apoptotic role in endothelial cells. Thus, we suggest that approaches which augment UCP2 expression in vascular endothelial cells aid in preventing the early stage development and progression of DR.


Assuntos
Glucose/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteína Desacopladora 2/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Retinopatia Diabética/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Óxido Nítrico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína Desacopladora 2/genética
11.
Mol Med Rep ; 11(1): 203-11, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25333816

RESUMO

The enhancement of apoptosis is a therapeutic strategy used in the treatment of cancer. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising antitumor agent. However, hepatocellular carcinoma (HCC) cells exhibit marked resistance to the induction of cell death by TRAIL. The present study investigated whether rocaglamide, a naturally occurring product isolated from the genus Aglaia, is able to sensitize resistant HCC cells to TRAIL-mediated apoptosis. Two HCC cell lines, HepG2 and Huh-7, were treated with rocaglamide and/or TRAIL and the induction of apoptosis and effects on the TRAIL signaling pathway were investigated. The in vivo efficacy of rocaglamide was determined in TRAIL-resistant Huh-7-derived tumor xenografts. Rocaglamide significantly sensitized the TRAIL-resistant HCC cells to apoptosis by TRAIL, which resulted from the rocaglamide-mediated downregulation of cellular FLICE-like inhibitory protein and subsequent caspase-8 activation. Furthermore, rocaglamide markedly inhibited tumor growth from Huh-7 cells propagated in severe combined immunodeficient mice, suggesting that chemosentization also occurred in vivo. These data suggest that rocaglamide acted synergistically with TRAIL against the TRAIL-resistant HCC cells. Thus, it is concluded that rocaglamide as an adjuvant to TRAIL-based therapy may present a promising therapeutic approach for the treatment of HCC.


Assuntos
Apoptose/efeitos dos fármacos , Benzofuranos/farmacologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Carcinoma Hepatocelular/metabolismo , Caspase 8/metabolismo , Neoplasias Hepáticas/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Apoptose/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Feminino , Técnicas de Silenciamento de Genes , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Camundongos , Camundongos SCID
12.
Eur J Cardiothorac Surg ; 46(6): 967-73, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24722942

RESUMO

OBJECTIVES: Development of intimal hyperplasia (IH) is the main pathology underlying graft failure following coronary artery bypass graft surgeries for ischaemic heart diseases, especially for great saphenous vein grafts which have a lower patency rate than internal mammary arteries. Neuropilin 1 (NRP1), which is a co-receptor for vascular endothelial growth factor found in vascular endothelial and smooth muscle cells, affects the development of IH. We examined the difference in NRP1 expression and distribution in human coronaries, aortas, mammary arteries and saphenous veins to detect a possible relation to their susceptibility to IH. METHODS: Ninety-five human vascular segments obtained from 40 patients were used for the comparison of NRP1 expression between different groups of blood vessels by western blot and real-time PCR. Additionally, staining scores were generated by computerized analysis of the microscopic images obtained after immunofluorescence and immunohistochemical staining to compare NRP1 expression patterns in endothelium, smooth muscles and adventitia in each vessel type. RESULTS: NRP1 expression in the aorta (2.03 ± 1.44) was more than twice as high as mammary artery expression (0.85 ± 0.75; n = 16, P = 0.0004); NRP1 of the latter (0.99 ± 0.91) was more than 30% greater than that of the corresponding saphenous vein (0.73 ± 0.69; n = 20, P = 0.0085). In adventitia, NRP1 receptor staining of the saphenous vein was higher (22.96 ± 8.73) than in the mammary artery (15.83 ± 7.13; n = 7, P = 0.049). Variations in NRP1 protein levels were accompanied by parallel variations in its mRNA levels (n = 15, P = 0.34). CONCLUSIONS: Autologous saphenous vein grafts, unlike internal mammary artery grafts, have lower NRP1 expression and abundant adventitial distribution of NRP1 within their walls; this may correlate with higher susceptibility to IH development.


Assuntos
Aorta/metabolismo , Prótese Vascular , Ponte de Artéria Coronária/métodos , Vasos Coronários/metabolismo , Neuropilina-1/biossíntese , Adulto , Idoso , Western Blotting , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neuropilina-1/genética , Especificidade de Órgãos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Túnica Íntima/metabolismo
13.
Mol Cancer ; 13: 38, 2014 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-24568222

RESUMO

BACKGROUND: Robust ERK1/2 activity, which frequently results from KRAS mutation, invariably occurs in pancreatic ductal adenocarcinoma (PDAC). However, direct interference of KRAS signaling has not led to clinically successful drugs. Correct localization of RAF is regulated by the scaffold protein prohibitin (PHB) that ensures the spatial organization between RAS and RAF in plasma membranes, thus leading to activation of downstream effectors. METHODS: PHB expression was analyzed in human pancreatic cancer cell lines, normal pancreas, and PDAC tissue. Furthermore, genetic ablation or pharmacological inhibition of PHB was performed to determine its role in growth, migration, and signaling of pancreatic cancer cells in vitro and in vivo. RESULTS: The level of PHB expression was crucial for maintenance of oncogenic ERK-driven pancreatic tumorigenesis. Additionally, rocaglamide (RocA), a small molecular inhibitor, selectively bound to PHB with nanomolar affinity to disrupt the PHB-CRAF interaction by altering its localization to the plasma membrane. Consequently, there was an impairment of oncogenic RAS-ERK signaling, thereby blocking in vitro and in vivo growth and metastasis of pancreatic cancer cells that were addicted to RAS-ERK signaling. More importantly, RocA treatment resulted in a significant increase of the lifespan of tumor-bearing mice without any detectable toxicity. CONCLUSIONS: Blockade of the PHB scaffold-CRAF kinase interaction, which is distinct from direct kinase inhibition, may be a new therapeutic strategy to target oncogenic ERK-driven pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Repressoras/metabolismo , Animais , Linhagem Celular Tumoral , Citoesqueleto/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Xenoenxertos , Humanos , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos SCID , Microscopia Confocal , Proibitinas , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas p21(ras) , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA