Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Exp Clin Cancer Res ; 42(1): 203, 2023 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-37563605

RESUMO

BACKGROUND: TP53, encoding the tumor suppressor p53, is frequently mutated in various cancers, producing mutant p53 proteins (mutp53) which can exhibit neomorphic, gain-of-function properties. The latter transform p53 into an oncoprotein that promotes metastatic tumor progression via downstream effectors such as ENTPD5, an endoplasmic reticulum UDPase involved in the calnexin/calreticulin cycle of N-glycoprotein biosynthesis. Elucidating the mechanisms underlying the pro-metastatic functions of the mutp53-ENTPD5 axis is crucial for developing targeted therapies for aggressive metastatic cancer. METHODS: We analyzed pancreatic, lung, and breast adenocarcinoma cells with p53 missense mutations to study the impact of mutp53 and ENTPD5 on the N-glycoproteins integrin-α5 (ITGA5) and integrin-ß1 (ITGB1), which heterodimerize to form the key fibronectin receptor. We assessed the role of the mutp53-ENTPD5 axis in integrin-dependent tumor-stroma interactions and tumor cell motility using adhesion, migration, and invasion assays, identifying and validating therapeutic intervention targets. We employed an orthotopic xenograft model of pancreatic ductal adenocarcinoma to examine in vivo targeting of mutp53-ENTPD5-mediated ITGA5 regulation for cancer therapy. RESULTS: Mutp53 depletion diminished ITGA5 and ITGB1 expression and impaired tumor cell adhesion, migration, and invasion, rescued by ENTPD5. The mutp53-ENTPD5 axis maintained ITGA5 expression and function via the calnexin/calreticulin cycle. Targeting this axis using ITGA5-blocking antibodies, α-glucosidase inhibitors, or pharmacological degradation of mutp53 by HSP90 inhibitors, such as Ganetespib, effectively inhibited ITGA5-mediated cancer cell motility in vitro. In the orthotopic xenograft model, Ganetespib reduced ITGA5 expression and metastasis in an ENTPD5-dependent manner. CONCLUSIONS: The mutp53-ENTPD5 axis fosters ITGA5 and ITGB1 expression and tumor cell motility through the calnexin/calreticulin cycle, contributing to cancer metastasis. ITGA5-blocking antibodies or α-glucosidase inhibitors target this axis and represent potential therapeutic options worth exploring in preclinical models. The pharmacologic degradation of mutp53 by HSP90 inhibitors effectively blocks ENTPD5-ITGA5-mediated cancer cell motility and metastasis in vivo, warranting further clinical evaluation in p53-mutant cancers. This research underscores the significance of understanding the complex interplay between mutp53, ENTPD5, and the calnexin/calreticulin cycle in integrin-mediated metastatic tumor progression, offering valuable insights for the development of potential therapeutic strategies.


Assuntos
Adenocarcinoma , Antineoplásicos , Animais , Humanos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Calnexina/genética , Calnexina/metabolismo , Integrina alfa5/metabolismo , Calreticulina/metabolismo , Anticorpos Bloqueadores/metabolismo , Inibidores de Glicosídeo Hidrolases , Linhagem Celular Tumoral , Chaperonas Moleculares/metabolismo , Modelos Animais de Doenças , Pirofosfatases/metabolismo , Proteínas Oncogênicas/metabolismo
2.
J Ovarian Res ; 15(1): 69, 2022 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-35668504

RESUMO

BACKGROUND: Dysregulation of Ectonucleoside Triphospahate Diphosphohydrolase 5 (ENTPD5) in tumors might be associated with tumor progression, while the role of ENTPD5 in the growth and metastasis of serous ovarian cancer (SOC) is still unclear. METHODS: ENTPD5 expression patterns in ovarian cancer tissues were analyzed by qRT-PCR and immunohistochemistry assay (IHC). Two SOC cell lines, SKOV3 and OVCAR8, were stably transfected with lentivirus to build knockdown and overexpression cell lines. Clone formation assay, collagen gel droplet culture technology, wound healing assay and flow cytometry were used to assess the migration and growth traits of SOC cells. Expression levels of ENTPD5, glucose regulated protein 78 (GRP78), eukaryotic translation initiation factor 2 alpha (eIF-2α), phosphorylated -eIF-2α and, C/EBP homologous protein (CHOP) in SOC cells were detected by Western blot. RESULTS: Compared to fallopian tube tissues, the expression of ENTPD5 was significantly higher in tumor tissues obtained from SOC patients, and positively correlated with clinical stage and metastasis. ENTPD5 knockdown robustly inhibited cell proliferation, migration, whereas ENTPD5 overexpression elicited the opposite effect on SOC cells. ENTPD5 knockdown arrested cell cycle in G0/G1 phase and increased apoptosis. Importantly, ENTPD5 knockdown was associated with significantly decreased protein levels for GRP78, CHOP, and p-eIF-2α, suggesting possible involvement of ENTPD5 in endoplasmic reticulum stress (ERS). CONCLUSIONS: Our study demonstrates that ENTPD5 knockdown inhibited SOC cell proliferation, migration and restrained the activation of the GRP78/p-eIF-2α/CHOP pathway, which provides a potentially effective therapeutic target for the treatment of SOC.


Assuntos
Cistadenocarcinoma Seroso , Neoplasias Ovarianas , Apoptose , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Cistadenocarcinoma Seroso/patologia , Fator de Iniciação 2 em Eucariotos/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 2 em Eucariotos/farmacologia , Feminino , Glucose , Humanos , Proteínas Oncogênicas , Neoplasias Ovarianas/patologia , Proteína C/farmacologia , Pirofosfatases/farmacologia
3.
Purinergic Signal ; 17(3): 467-480, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34075526

RESUMO

NTPDase5 is a nucleotidase of the endoplasmic reticulum that plays an important role in proteostasis as a regulator of protein N-glycosylation. This enzyme was first identified in hamster as a proto-oncogene activated upon a single nucleotide deletion that causes a frameshift leading to a truncated protein. Truncated NTPDase5 proteins were detected in human samples, but an oncogene was never identified. Searching for transcript variants in the GenBank database and using TCGA data, we discovered that splice variants could originate truncated human NTPDase5 proteins. We identified three main splicing events in the ENTPD5 gene: alternative acceptors, exon skipping, and alternative terminators. The analysis of impact of splicing events in cancers showed that skipping of exon 11-the event that leads to truncated proteins similar in size to the hamster oncogene-does not affect the hazard ratio of most tumors and was, in fact, a protective factor in the only two cancer studies where it was significant. We also identified four main patterns of impact of ENTPD5 in cancer and a potential variant-specific regulation by miR-215. Our findings shed light on a two-decade uncertainty about the origin of truncated NTPDase5 and contribute to the characterization of its impacts in cancer.


Assuntos
Variação Genética/genética , Neoplasias/genética , Neoplasias/mortalidade , Proteínas Oncogênicas/genética , Isoformas de Proteínas/genética , Pirofosfatases/genética , Humanos , Taxa de Sobrevida/tendências
4.
Nan Fang Yi Ke Da Xue Xue Bao ; 41(4): 555-561, 2021 Apr 20.
Artigo em Chinês | MEDLINE | ID: mdl-33963715

RESUMO

OBJECTIVE: To investigate the expression of ENTPD5 in epithelial ovarian cancer and explore its clinical implications. OBJECTIVE: The expression level of ENTPD5 in epithelial ovarian cancer was analyzed based on data from Oncomine and TCGA databases. The relationship between the expression level of ENTPD5 and clinical characteristics of the patients was analyzed using UALCAN database. Gene enrichment analysis (GSEA) was performed to explore the possible role of ENTPD5 in the occurrence and progression of epithelial ovarian cancer. CIBERSORT package was used to analyze the relationship between the expression of ENTPD5 and immune infiltration. The expression patterns of ENTPD5 were verified in 23 epithelial ovarian cancer tissues and 15 normal ovarian tissues using RT-qPCR and Western blotting; the expression of ENTPD5 protein was also detected immunohistochemically in 50 paraffin-embedded samples of epithelial ovarian cancer and 6 normal ovarian tissues. OBJECTIVE: Analysis of Oncomine and TCGA databases showed that the expression of ENTPD5 was significantly higher in epithelial ovarian cancer tissues than in normal ovarian tissues (P < 0.05), and its expression level was negatively correlated with the survival rate of the patients (P < 0.05). Data from UALCAN database showed that the expression level of ENTPD5 was related with the age of patients. The results of GSEA suggested that ENTPD5 was involved in ABC transporter, WNT signaling pathway and insulin signaling, and the expression of ENTPD5 was negatively correlated with the contents of NK cells, mast cells and eosinophils (P < 0.05). In clinical samples of epithelial ovarian cancer tissues, the expression of ENTPD5 was significantly higher than that in normal ovarian tissues at both the mRNA (P < 0.01) and protein (P < 0.05) levels. The paraffinembedded samples also showed significantly higher expressions of ENTPD5 in epithelial ovarian cancer than in normal ovarian tissues (P < 0.05). OBJECTIVE: ENTPD5 is highly expressed in epithelial ovarian cancer, which may promote the occurrence and progression of epithelial ovarian cancer by participating in multiple functional processes and cellular immune infiltration.


Assuntos
Neoplasias Ovarianas , Biomarcadores Tumorais/genética , Carcinoma Epitelial do Ovário/genética , Biologia Computacional , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Oncogênicas , Neoplasias Ovarianas/genética , Prognóstico , Pirofosfatases
5.
Acta Neuropathol ; 138(2): 275-293, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31062076

RESUMO

Glioblastomas strongly invade the brain by infiltrating into the white matter along myelinated nerve fiber tracts even though the myelin protein Nogo-A prevents cell migration by activating inhibitory RhoA signaling. The mechanisms behind this long-known phenomenon remained elusive so far, precluding a targeted therapeutic intervention. This study demonstrates that the prevalent activation of AKT in gliomas increases the ER protein-folding capacity and enables tumor cells to utilize a side effect of RhoA activation: the perturbation of the IRE1α-mediated decay of SPARC mRNA. Once translation is initiated, glioblastoma cells rapidly secrete SPARC to block Nogo-A from inhibiting migration via RhoA. By advanced ultramicroscopy for studying single-cell invasion in whole, undissected mouse brains, we show that gliomas require SPARC for invading into white matter structures. SPARC depletion reduces tumor dissemination that significantly prolongs survival and improves response to cytostatic therapy. Our finding of a novel RhoA-IRE1 axis provides a druggable target for interfering with SPARC production and underscores its therapeutic value.


Assuntos
Neoplasias Encefálicas/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Proteínas de Neoplasias/fisiologia , Proteínas Nogo/biossíntese , Osteonectina/biossíntese , Biossíntese de Proteínas , Substância Branca/patologia , Proteína rhoA de Ligação ao GTP/fisiologia , Animais , Ligação Competitiva , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Invasividade Neoplásica , Proteínas Nogo/genética , Osteonectina/genética , Domínios Proteicos , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Receptores de Esfingosina-1-Fosfato/fisiologia , Células Tumorais Cultivadas , Substância Branca/metabolismo
6.
Elife ; 72018 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-29767626

RESUMO

The patterning of the spine of a zebrafish is controlled by the notochord, a rod-like structure that supports and instructs the developing embryo.


Assuntos
Notocorda , Peixe-Zebra , Animais , Padronização Corporal , Osso e Ossos , Coluna Vertebral
7.
Elife ; 72018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29624170

RESUMO

Segmentation of the axial skeleton in amniotes depends on the segmentation clock, which patterns the paraxial mesoderm and the sclerotome. While the segmentation clock clearly operates in teleosts, the role of the sclerotome in establishing the axial skeleton is unclear. We severely disrupt zebrafish paraxial segmentation, yet observe a largely normal segmentation process of the chordacentra. We demonstrate that axial entpd5+ notochord sheath cells are responsible for chordacentrum mineralization, and serve as a marker for axial segmentation. While autonomous within the notochord sheath, entpd5 expression and centrum formation show some plasticity and can respond to myotome pattern. These observations reveal for the first time the dynamics of notochord segmentation in a teleost, and are consistent with an autonomous patterning mechanism that is influenced, but not determined by adjacent paraxial mesoderm. This behavior is not consistent with a clock-type mechanism in the notochord.


Assuntos
Animais Geneticamente Modificados/fisiologia , Relógios Biológicos , Padronização Corporal , Osso e Ossos/fisiologia , Notocorda/fisiologia , Pirofosfatases/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados/embriologia , Animais Geneticamente Modificados/genética , Osso e Ossos/embriologia , Embrião não Mamífero/citologia , Embrião não Mamífero/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/embriologia , Mesoderma/fisiologia , Mutação , Notocorda/embriologia , Pirofosfatases/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
8.
Mol Cell Oncol ; 4(2): e1288678, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28401189

RESUMO

Mutations in cancer abolish normal tumor suppressive functions of tumor protein p53 (TP53, best known as p53) and convert it into an oncogene. We recently reported the identification of ectonucleoside triphosphate diphosphohydrolase 5 (ENTPD5) as a transcriptional target of mutant p53 that enhances folding of N-glycosylated proteins required for cancer cell migration, invasion, and metastasis.

10.
Proc Natl Acad Sci U S A ; 113(52): E8433-E8442, 2016 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-27956623

RESUMO

Mutations in the p53 tumor suppressor gene are the most frequent genetic alteration in cancer and are often associated with progression from benign to invasive stages with metastatic potential. Mutations inactivate tumor suppression by p53, and some endow the protein with novel gain of function (GOF) properties that actively promote tumor progression and metastasis. By comparative gene expression profiling of p53-mutated and p53-depleted cancer cells, we identified ectonucleoside triphosphate diphosphohydrolase 5 (ENTPD5) as a mutant p53 target gene, which functions as a uridine 5'-diphosphatase (UDPase) in the endoplasmic reticulum (ER) to promote the folding of N-glycosylated membrane proteins. A comprehensive pan-cancer analysis revealed a highly significant correlation between p53 GOF mutations and ENTPD5 expression. Mechanistically, mutp53 is recruited by Sp1 to the ENTPD5 core promoter to induce its expression. We show ENTPD5 to be a mediator of mutant p53 GOF activity in clonogenic growth, architectural tissue remodeling, migration, invasion, and lung colonization in an experimental metastasis mouse model. Our study reveals folding of N-glycosylated membrane proteins in the ER as a mechanism underlying the metastatic progression of tumors with mutp53 that could provide new possibilities for cancer treatment.


Assuntos
Retículo Endoplasmático/metabolismo , Metástase Neoplásica , Proteínas Oncogênicas/metabolismo , Pirofosfatases/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose , Calnexina/metabolismo , Calreticulina/metabolismo , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Glicoproteínas/metabolismo , Glicosilação , Humanos , Masculino , Camundongos , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Mutação , Invasividade Neoplásica , Prognóstico , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fator de Transcrição Sp1/metabolismo
11.
J Proteomics ; 108: 171-87, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-24859727

RESUMO

The transcription factor Nrf2 is a master regulator of cellular defence: Nrf2 null mice (Nrf2((-/-))) are highly susceptible to chemically induced toxicities. We report a comparative iTRAQ-based study in Nrf2((-/-)) mice treated with a potent inducer, methyl-2-cyano-3,12-dioxooleana-1,9(11)dien-28-oate (CDDO-me; bardoxolone -methyl), to define both the Nrf2-dependent basal and inducible hepatoproteomes. One thousand five hundred twenty-one proteins were fully quantified (FDR <1%). One hundred sixty-one were significantly different (P<0.05) between WT and Nrf2((-/-)) mice, confirming extensive constitutive regulation by Nrf2. Treatment with CDDO-me (3mg/kg; i.p.) resulted in significantly altered expression of 43 proteins at 24h in WT animals. Six proteins were regulated at both basal and inducible levels exhibiting the largest dynamic range of Nrf2 regulation: cytochrome P4502A5 (CYP2A5; 17.2-fold), glutathione-S-transferase-Mu 3 (GSTM3; 6.4-fold), glutathione-S-transferase Mu 1 (GSTM1; 5.9-fold), ectonucleoside-triphosphate diphosphohydrolase (ENTPD5; 4.6-fold), UDP-glucose-6-dehydrogenase (UDPGDH; 4.1-fold) and epoxide hydrolase (EPHX1; 3.0-fold). These proteins, or their products, thus provide a potential source of biomarkers for Nrf2 activity. ENTPD5 is of interest due to its emerging role in AKT signalling and, to our knowledge, this protein has not been previously shown to be Nrf2-dependent. Only two proteins altered by CDDO-me in WT animals were similarly affected in Nrf2((-/-)) mice, demonstrating the high degree of selectivity of CDDO-me for the Nrf2:Keap1 signalling pathway. BIOLOGICAL SIGNIFICANCE: The Nrf2:Keap1 signalling pathway is attracting considerable interest as a therapeutic target for different disease conditions. For example, CDDO-me (bardoxolone methyl) was investigated in clinical trials for the treatment of acute kidney disease, and dimethyl fumarate, recently approved for reducing relapse rate in multiple sclerosis, is a potent Nrf2 inducer. Such compounds have been suggested to act through multiple mechanisms; therefore, it is important to define the selectivity of Nrf2 inducers to assess the potential for off-target effects that may lead to adverse drug reactions, and to provide biomarkers with which to assess therapeutic efficacy. Whilst there is considerable information on the global action of such inducers at the mRNA level, this is the first study to catalogue the hepatic protein expression profile following acute exposure to CDDO-me in mice. At a dose shown to evoke maximal Nrf2 induction in the liver, CDDO-me appeared highly selective for known Nrf2-regulated proteins. Using the transgenic Nrf2((-/-)) mouse model, it could be shown that 97% of proteins induced in wild type mice were associated with a functioning Nrf2 signalling pathway. This analysis allowed us to identify a panel of proteins that were regulated both basally and following Nrf2 induction. Identification of these proteins, which display a large magnitude of variation in their expression, provides a rich source of potential biomarkers for Nrf2 activity for use in experimental animals, and which may be translatable to man to define individual susceptibility to chemical stress, including that associated with drugs, and also to monitor the pharmacological response to Nrf2 inducers.


Assuntos
Fator 2 Relacionado a NF-E2/metabolismo , Ácido Oleanólico/análogos & derivados , Proteoma/biossíntese , Transdução de Sinais/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch , Fígado , Masculino , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Ácido Oleanólico/toxicidade , Oxirredutases/biossíntese , Oxirredutases/genética , Proteoma/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA