Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Biochem Funct ; 38(5): 651-659, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32196701

RESUMO

It has been shown that overexpression of activated leukocyte cell adhesion molecule (ALCAM) is involved in development of resistance to tamoxifen therapy and promotion of cell invasion, migration and metastasis in ER+ breast cancer cells. Thus, we hypothesized that blockade of ALCAM interconnections with antibodies could be an effective approach for reversing mentioned negative events associated with ALCAM overexpression in breast cancer cells. Here, an anti-ALCAM scFv was recombinantly expressed and used throughout study for examination of the putative anticancer effects of ALCAM blockade. The anti-ALCAM scFv coding sequence was obtained from GenBank database and after addition of a 6× His-tag moiety, signal peptide and flanking sequences, the whole construct was expressed in Escherichia coli. Tamoxifen resistant MCF7 cells were then pretreat for 24 hours with purified recombinant anti-ALCAM scFv prior to administration of tamoxifen. In parallel, the cytotoxicity profile of anti-ALCAM scFv and tamoxifen co-treatments against tamoxifen resistant and sensitive MCF7 cell lines was also evaluated using CompuSyn software. The invasion/migration inhibitory effects of anti-ALCAM scFv on MDA-MB-231 cells were also evaluated. Pretreatment with anti-ALCAM scFv could successfully enhance anti-proliferative effects of tamoxifen against resistant MCF-7 cell lines. Furthermore, the combination of 19.2:1 of tamoxifen to anti-ALCAM scFv demonstrated synergistic cell inhibitory effect against tamoxifen resistant MCF7 cell lines. Also, incubating MDA-MB-231 cell lines with anti-ALCAM scFv resulted in a 30% and 25% reduction in number of invaded and migrated cells respectively. Overall, application of anti-ALCAM scFv could significantly suppress cancer cells metastasis in vitro and modulate tamoxifen resistant ER+ MCF7 cell line's sensitivity to tamoxifen. SIGNIFICANCE OF THE STUDY: Acquisition of resistance to tamoxifen therapy is one of the major challenges associated with cancer chemotherapy, gradually turning a responsive tumour into a refractory more invasive one which ultimately ends in disease progression and relapse. Here, we reported expression of an anti-ALCAM scFv, capable of increasing the sensitivity of tamoxifen resistant ER+ MCF-7 cells to tamoxifen therapy following a 24-hour pretreatment period. In addition, we demonstrated that the anti-ALCAM scFv monotherapy was also capable of suppressing invasion and migration of MDA-MB-231 cells in Boyden chamber assays.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Moléculas de Adesão Celular Neuronais/antagonistas & inibidores , Proteínas Fetais/antagonistas & inibidores , Tamoxifeno/farmacologia , Antígenos CD/genética , Antígenos CD/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
2.
Carcinogenesis ; 35(7): 1491-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24445144

RESUMO

Recent evidence suggests that the expression of brachyury is necessary for chordoma growth. However, the mechanism associated with brachyury-regulated cell growth is poorly understood. Fibroblast growth factor (FGF), a regulator of brachyury expression in normal tissue, may also play an important role in chordoma pathophysiology. Using a panel of chordoma cell lines, we explored the role of FGF signaling and brachyury in cell growth and survival. Western blots showed that all chordoma cell lines expressed fibroblast growth factor receptor 2 (FGFR2), FGFR3, mitogen-activated protein kinase kinase (MEK) and extracellular signal-regulated kinase (ERK), whereas no cell lines expressed FGFR1 and FGFR4. Results of enzyme-linked immunosorbent assay indicated that chordoma cells produced FGF2. Neutralization of FGF2 inhibited MEK/ERK phosphorylation, decreased brachyury expression and induced apoptosis while reducing cell growth. Activation of the FGFR/MEK/ERK/brachyury pathway by FGF2-initiated phosphorylation of FGFR substrate 2 (FRS2)-α (Tyr196) prevented apoptosis while promoting cell growth and epithelial-mesenchymal transition (EMT). Immunofluorescence staining showed that FGF2 promoted the translocation of phosphorylated ERK to the nucleus and increased brachyury expression. The selective inhibition of FGFR, MEK and ERK phosphorylation by PD173074, PD0325901 and PD184352, respectively, decreased brachyury expression, induced apoptosis, and inhibited cell growth and EMT. Moreover, knockdown of brachyury by small hairpin RNA reduced FGF2 secretion, inhibited FGFR/MEK/ERK phosphorylation and blocked the effects of FGF2 on cell growth, apoptosis and EMT. Those findings highlight that FGFR/MEK/ERK/brachyury pathway coordinately regulates chordoma cell growth and survival and may represent a novel chemotherapeutic target for chordoma.


Assuntos
Apoptose , Proliferação de Células , Cordoma/patologia , Proteínas Fetais/metabolismo , MAP Quinase Quinase 1/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Proteínas com Domínio T/metabolismo , Western Blotting , Movimento Celular , Células Cultivadas , Cordoma/genética , Cordoma/metabolismo , Transição Epitelial-Mesenquimal , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Imunofluorescência , Humanos , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/genética , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Fosforilação , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Proteínas com Domínio T/antagonistas & inibidores , Proteínas com Domínio T/genética
3.
Biochem Biophys Res Commun ; 451(2): 334-8, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-25094049

RESUMO

Cluster of differentiation 166 (CD166) is critical for liver cancer cell survival. Our previously study demonstrated that CD166 exerts its anti-apoptotic role through interaction with YAP in liver cancer. However, the interaction between CD166 and other cell surface molecules remains unclear in liver cancer cells. In the current study, we found that both mRNA and protein of CD44 expression was significantly inhibited by knocking-down CD166. Moreover, CD166 affected-CD44 expression is dependent of transcription via blocking NF-κB pathway. On the contrary, CD44 promoted up-regulation of CD166 mRNA and protein. And it may be through E3 ubiquitin ligases COP1 and UBC3 to regulate CD166 protein degradation. Collectively, these results suggest that CD166 and CD44 play important roles in liver cancer development. Therefore, CD166 may develop as a potential therapeutic molecule target for the treatment of liver cancer.


Assuntos
Antígenos CD/genética , Antígenos CD/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , NF-kappa B/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose , Carcinoma Hepatocelular/patologia , Moléculas de Adesão Celular Neuronais/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células , Proteínas Fetais/antagonistas & inibidores , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Hepáticas/patologia , Fosfoproteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Transdução de Sinais , Fatores de Transcrição , Regulação para Cima , Proteínas de Sinalização YAP
4.
Eur J Med Chem ; 216: 113247, 2021 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-33652355

RESUMO

As a newly emerged technology, PROTAC (proteolysis targeting chimera) is a promising therapeutic strategy for varieties of diseases. Unlike small molecule inhibitors, PROTACs catalytically induce target proteins degradation, including currently "undruggable" target proteins. In addition, PROTACs can be a potentially successful strategy to overcome drug resistance. IAPs can inhibit apoptosis by inhibiting caspase, and also exhibits the activity of E3 ubiquitin ligase. Specific and nongenetic IAP-based protein erasers (SNIPERs) are hybrid molecules that designed based on IAPs, and used to degrade the target proteins closely associated with diseases. Their structures consist of three parts, including target protein ligand, E3 ligase ligand and the linker between them. SNIPERs (PROTACs) degrade diseases-associated proteins through human inherent ubiquitin-proteasome system. So far, many SNIPERs have been developed to treat diseases that difficult to handle by traditional methods, such as radiotherapy, chemotherapy and small molecule inhibitors, and showed promising prospects in application. In this paper, the recent advances of SNIPERs were summarized, and the chances and challenges associated with this area were also highlighted.


Assuntos
Proteínas Inibidoras de Apoptose/metabolismo , Ligantes , Antineoplásicos/química , Antineoplásicos/metabolismo , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/metabolismo , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Proteínas de Fusão bcr-abl/metabolismo , Humanos , Proteína Huntingtina/antagonistas & inibidores , Proteína Huntingtina/metabolismo , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Receptor Notch1/antagonistas & inibidores , Receptor Notch1/metabolismo , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo
5.
Nat Commun ; 12(1): 5337, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34504101

RESUMO

TNK1 is a non-receptor tyrosine kinase with poorly understood biological function and regulation. Here, we identify TNK1 dependencies in primary human cancers. We also discover a MARK-mediated phosphorylation on TNK1 at S502 that promotes an interaction between TNK1 and 14-3-3, which sequesters TNK1 and inhibits its kinase activity. Conversely, the release of TNK1 from 14-3-3 allows TNK1 to cluster in ubiquitin-rich puncta and become active. Active TNK1 induces growth factor-independent proliferation of lymphoid cells in cell culture and mouse models. One unusual feature of TNK1 is a ubiquitin-association domain (UBA) on its C-terminus. Here, we characterize the TNK1 UBA, which has high affinity for poly-ubiquitin. Point mutations that disrupt ubiquitin binding inhibit TNK1 activity. These data suggest a mechanism in which TNK1 toggles between 14-3-3-bound (inactive) and ubiquitin-bound (active) states. Finally, we identify a TNK1 inhibitor, TP-5801, which shows nanomolar potency against TNK1-transformed cells and suppresses tumor growth in vivo.


Assuntos
Proteínas 14-3-3/genética , Proteínas Fetais/genética , Linfócitos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Proteínas Tirosina Quinases/genética , Ubiquitina/genética , Proteínas 14-3-3/metabolismo , Células A549 , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/metabolismo , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/patologia , Camundongos , Fosfolipase C gama/genética , Fosfolipase C gama/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/mortalidade , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Pirimidinas/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Análise de Sobrevida , Carga Tumoral/efeitos dos fármacos , Ubiquitina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Biochemistry ; 48(40): 9327-9, 2009 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-19764708

RESUMO

Formins are potent actin assembly factors. Diaphanous formins, including mDia1, mDia2, and mDia3 in mammals, are implicated in mitosis and cytokinesis, but no chemical interactors have been reported. We developed an in vitro screen for inhibitors of actin assembly by mDia1 and identified an inhibitor of mDia1 and mDia2 that does not inhibit mDia3 at the concentrations tested. These results establish the druggability of mDia formins and introduce a first-generation inhibitor.


Assuntos
Actinas/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas de Transporte/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , NADPH Desidrogenase/antagonistas & inibidores , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Proteínas Fetais/antagonistas & inibidores , Forminas , Humanos , Camundongos , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/fisiologia , NADPH Desidrogenase/química , NADPH Desidrogenase/fisiologia , Proteínas Nucleares/antagonistas & inibidores , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiologia , Estrutura Terciária de Proteína/fisiologia , Pirenos/química , Homologia de Sequência de Aminoácidos
8.
Front Immunol ; 10: 759, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31031759

RESUMO

Activated leukocyte cell adhesion molecule (ALCAM, CD166) is a cell adhesion molecule of the immunoglobulin superfamily and has been implicated in diverse pathophysiological processes including T cell activation, leukocyte trafficking, and (lymph)angiogenesis. However, exploring the therapeutic potential of ALCAM blockade in immune-mediated inflammatory disorders has been difficult due to the lack of antibodies with blocking activity toward murine ALCAM. In this study, we identified and characterized a monoclonal antibody with high affinity and specificity for murine ALCAM. This antibody reduced in vitro T cell activation induced by antigen-presenting dendritic cells (DCs) as well as (trans)migration of murine DCs across lymphatic endothelial monolayers. Moreover, it reduced emigration of DCs from in vitro-cultured human skin biopsies. Similarly, antibody-based blockade of ALCAM reduced (lymph)angiogenic processes in vitro and decreased developmental lymphangiogenesis in vivo to levels observed in ALCAM-deficient mice. Since corneal allograft rejection is an important medical condition that also involves (lymph)angiogenesis, DC migration and T cell activation, we investigated the therapeutic potential of ALCAM blockade in murine corneal disease. Blocking ALCAM lead to DC retention in corneas and effectively prevented corneal allograft rejection. Considering that we also detected ALCAM expression in human corneal DCs and lymphatics, our findings identify ALCAM as a potential novel therapeutic target in human corneal allograft rejection.


Assuntos
Antígenos CD/genética , Moléculas de Adesão Celular Neuronais/genética , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Proteínas Fetais/genética , Imunidade , Vasos Linfáticos , Aloenxertos , Animais , Antígenos CD/metabolismo , Biópsia , Moléculas de Adesão Celular Neuronais/antagonistas & inibidores , Moléculas de Adesão Celular Neuronais/metabolismo , Movimento Celular/genética , Movimento Celular/imunologia , Transplante de Córnea , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/metabolismo , Engenharia Genética , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Linfangiogênese , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Pele/imunologia , Pele/metabolismo , Pele/patologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
9.
Mol Cancer Ther ; 17(3): 603-613, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29237806

RESUMO

Chordomas are rare bone tumors with no approved therapy. These tumors express several activated tyrosine kinase receptors, which prompted attempts to treat patients with tyrosine kinase inhibitors. Although clinical benefit was observed in phase II clinical trials with imatinib and sorafenib, and sporadically also with EGFR inhibitors, therapies evaluated to date have shown modest activity. With the goal of identifying new drugs with immediate therapeutic potential for chordoma patients, we collected clinically approved drugs and other advanced inhibitors of MET, PDGFRß, and EGFR tyrosine kinases, and assessed their antiproliferative activity against a panel of chordoma cell lines. Chordoma cell lines were not responsive to MET and PDGFRß inhibitors. U-CH1 and UM-Chor1 were sensitive to all EGFR inhibitors, whereas the remaining cell lines were generally insensitive to these drugs. Afatinib was the only EGFR inhibitor with activity across the chordoma panel. We then investigated the molecular mechanisms behind the responses observed and found that the antiproliferative IC50s correlate with the unique ability of afatinib to promote degradation of EGFR and brachyury, an embryonic transcription factor considered a key driver of chordoma. Afatinib displayed potent antitumor efficacy in U-CH1, SF8894, CF322, and CF365 chordoma tumor models in vivo In the panel analyzed, high EGFR phosphorylation and low AXL and STK33 expression correlated with higher sensitivity to afatinib and deserve further investigation as potential biomarkers of response. These data support the use of afatinib in clinical trials and provide the rationale for the upcoming European phase II study on afatinib in advanced chordoma. Mol Cancer Ther; 17(3); 603-13. ©2017 AACR.


Assuntos
Afatinib/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Cordoma/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Proteínas Fetais/antagonistas & inibidores , Proteínas com Domínio T/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cordoma/genética , Cordoma/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética
10.
Curr Biol ; 5(1): 62-7, 1995 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-7535172

RESUMO

BACKGROUND: The Brachyury (T) gene is required for the formation of posterior mesoderm and for axial development in both mouse and zebrafish embryos. In these species, and in Xenopus, the gene is expressed transiently throughout the presumptive mesoderm, and transcripts then persiste in notochord and posterior tissues. In Xenopus embryos, expression of the Xenopus homologue of Brachyury, Xbra, can be induced in presumptive ectoderm by basic fibroblast growth factor (FGF) and activin; in the absence of functional FGF or activin signalling pathways, expression of the gene is severely reduced. Ectopic expression of Xbra in presumptive ectoderm causes mesoderm to be formed. As Brachyury and its homologues encode sequence-specific DNA-binding proteins, it is likely that each functions by directly activating downstream mesoderm-specific genes. RESULTS: We show that expression in Xenopus embryos of RNA encoding a dominant-negative FGF receptor inhibits the mesoderm-inducing activity of Xbra. We demonstrate that ectopic expression of Xbra activates transcription of the embryonic FGF gene, and that embryonic FGF can induce expression of Xbra. This suggests that the two genes are components of a regulatory loop. Consistent with this idea, dissociation of Xbra-expressing cells causes a dramatic and rapid reduction in levels of Xbra, but the reduction can be inhibited by addition of FGF. CONCLUSION: Formation of mesoderm tissue requires an intact FGF signalling pathway downstream of Brachyury. This requirement is due to a regulatory loop, in which Brachyury activates expression of a member of the FGF family, and FGF maintains expression of Brachyury.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas Fetais/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Mesoderma/fisiologia , Proteínas com Domínio T , Ativinas , Animais , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/genética , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Inibinas/fisiologia , RNA/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Transcrição Gênica/fisiologia , Xenopus
11.
Curr Biol ; 9(17): 967-70, 1999 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-10508592

RESUMO

Programmed cell death in animals is usually associated with apoptotic morphology and requires caspase activation. Necrosis and caspase-independent cell death have been reported, but mostly in experimental conditions that lead some to question their existence it in vivo. Loss of interdigital cells in the mouse embryo, a paradigm of cell death during development [1], is known to include an apoptotic [2] and caspase-dependent [3] [4] mechanism. Here, we report that, when caspase activity was inhibited using drugs or when apoptosis was prevented genetically (using Hammertoe mutant mice, or mice homozygous for a mutation in the gene encoding APAF-1, a caspase-activating adaptor protein), interdigital cell death still occurred. This cell death was negative for the terminal-deoxynucleotidyl-mediated dUTP nick end-labelling (TUNEL) assay and there was no overall cell condensation. At the electron microscopy level, peculiar 'mottled' chromatin alterations and marked mitochondrial and membrane lesions, suggestive of classical necrotic cell death, were observed with no detectable phagocytosis and no local inflammatory response. Thus, in this developmental context, although caspase activity confers cell death with an apoptotic morphotype, in the absence of caspase activity an underlying mechanism independent of known caspases can also confer cell death, but with a necrotic morphotype. This cell death can go undetected when using apoptosis-specific methodology, and cannot be blocked by agents that act on caspases.


Assuntos
Caspases/fisiologia , Proteínas Fetais/fisiologia , Membro Posterior/embriologia , Necrose , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Fator Apoptótico 1 Ativador de Proteases , Proteínas Morfogenéticas Ósseas/agonistas , Proteínas Morfogenéticas Ósseas/fisiologia , Inibidores de Caspase , Cromatina/ultraestrutura , Inibidores de Cisteína Proteinase/farmacologia , Desenvolvimento Embrionário e Fetal , Proteínas Fetais/antagonistas & inibidores , Membro Posterior/anormalidades , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Camundongos Mutantes , Morfogênese/fisiologia , Organelas/ultraestrutura , Proteínas/genética , Receptores de Fatores de Crescimento/agonistas , Transdução de Sinais/efeitos dos fármacos , Tacrolimo/análogos & derivados , Tacrolimo/farmacologia
12.
Science ; 349(6247): 535-9, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26228148

RESUMO

The actin cross-linking domain (ACD) is an actin-specific toxin produced by several pathogens, including life-threatening spp. of Vibrio cholerae, Vibrio vulnificus, and Aeromonas hydrophila. Actin cross-linking by ACD is thought to lead to slow cytoskeleton failure owing to a gradual sequestration of actin in the form of nonfunctional oligomers. Here, we found that ACD converted cytoplasmic actin into highly toxic oligomers that potently "poisoned" the ability of major actin assembly proteins, formins, to sustain actin polymerization. Thus, ACD can target the most abundant cellular protein by using actin oligomers as secondary toxins to efficiently subvert cellular functions of actin while functioning at very low doses.


Assuntos
Actinas/metabolismo , Antígenos de Bactérias/química , Antígenos de Bactérias/toxicidade , Toxinas Bacterianas/química , Toxinas Bacterianas/toxicidade , Proteínas Fetais/antagonistas & inibidores , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas Nucleares/antagonistas & inibidores , Animais , Antígenos de Bactérias/genética , Toxinas Bacterianas/genética , Linhagem Celular , Forminas , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Polimerização/efeitos dos fármacos , Estrutura Terciária de Proteína , Ratos
13.
J Cell Biol ; 210(3): 401-17, 2015 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-26216902

RESUMO

Although actin at neuronal growth cones is well-studied, much less is known about actin organization and dynamics along axon shafts and presynaptic boutons. Using probes that selectively label filamentous-actin (F-actin), we found focal "actin hotspots" along axons-spaced ∼3-4 µm apart-where actin undergoes continuous assembly/disassembly. These foci are a nidus for vigorous actin polymerization, generating long filaments spurting bidirectionally along axons-a phenomenon we call "actin trails." Super-resolution microscopy reveals intra-axonal deep actin filaments in addition to the subplasmalemmal "actin rings" described recently. F-actin hotspots colocalize with stationary axonal endosomes, and blocking vesicle transport diminishes the actin trails, suggesting mechanistic links between vesicles and F-actin kinetics. Actin trails are formin-but not Arp2/3-dependent and help enrich actin at presynaptic boutons. Finally, formin inhibition dramatically disrupts synaptic recycling. Collectively, available data suggest a two-tier F-actin organization in axons, with stable "actin rings" providing mechanical support to the plasma membrane and dynamic "actin trails" generating a flexible cytoskeletal network with putative physiological roles.


Assuntos
Actinas/metabolismo , Axônios/metabolismo , Proteínas Fetais/metabolismo , Cones de Crescimento/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Nucleares/metabolismo , Vesículas Sinápticas/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Actinas/biossíntese , Animais , Membrana Celular/fisiologia , Proteínas Fetais/antagonistas & inibidores , Forminas , Proteínas de Fluorescência Verde , Camundongos , Proteínas dos Microfilamentos/antagonistas & inibidores , Microtúbulos/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Terminações Pré-Sinápticas/metabolismo
14.
Mol Med Rep ; 12(1): 995-1001, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25683840

RESUMO

The objective of the current study was to investigate the impact of Brachyury on epithelial-mesenchymal transitions and chemosensitivity in non-small cell lung cancer (NSCLC). In 115 archived NSCLC tissue samples, the expression of Brachyury was observed to be significantly higher than that in adjacent normal lung tissues. In addition, the current study demonstrated that the expression of Brachyury is closely associated with TNM staging, lymph node metastasis and the prognosis of NSCLC, although not with patient age, gender or tumor differentiation. Brachyury expression is also accompanied by the downregulation of E-cadherin and the upregulation of N-cadherin. Brachyury may promote lung cancer through induction of epithelial-mesenchymal transition, which leads to metastasis and consequent poor prognosis in patients with lung cancer. Furthermore, the present study observed that interfering with Brachyury increases the sensitivity of cells to chemotherapeutic treatment with cisplatin. These results, in combination with those of additional studies, suggest that Brachyury may be used as a novel target for the prevention and treatment of lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Transição Epitelial-Mesenquimal/genética , Proteínas Fetais/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Proteínas com Domínio T/genética , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/genética , Antígenos CD/metabolismo , Antineoplásicos/farmacologia , Caderinas/genética , Caderinas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Análise de Sobrevida , Proteínas com Domínio T/antagonistas & inibidores , Proteínas com Domínio T/metabolismo
15.
Am J Med Genet ; 87(3): 207-16, 1999 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-10564872

RESUMO

The Smith-Lemli-Opitz syndrome (SLOS) is an autosomal recessive condition involving craniofacial and central nervous system malformations with occasional holoprosencephaly (HPE). It is caused by a defect in the 7-dehydrocholesterol (7-DHC) reductase, the enzyme catalyzing the last step of cholesterol biosynthesis. Treatment of pregnant rats with inhibitors of 7-DHC reductase, either AY9944 or BM15.766, has provided a valuable model to study the pathogenesis in SLOS. Recently, cholesterol has been shown to be involved in the post-translational activation of the signaling protein Sonic Hedgehog. To identify the early defects associated with HPE in a rat model of SLOS, and to compare the phenotype of the treated embryos with that of the Shh(-/-) mutants, we examined brain morphology and expression of three developmental genes (Shh, Otx2, and Pax6 ) in 23-somite stage embryos from AY9944-treated dams. We report clearly abnormal morphology of the developing brain, concerning primarily the ventral aspect of the neural tube. We observed a reduced or absent expression of Shh and Otx2 in their ventral domain associated with extended ventral expression of Pax6. The results suggest an absence of the midline ventral cell type at all levels of the cranial neural tube. They provide further evidence that cholesterol-deficiency-induced HPE originates from impaired Shh signaling activity in the ventral neural tube.


Assuntos
Encéfalo/patologia , Colesterol/fisiologia , Modelos Animais de Doenças , Inibidores Enzimáticos/toxicidade , Proteínas Fetais/deficiência , Proteínas de Homeodomínio , Defeitos do Tubo Neural/embriologia , Neurônios/patologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/deficiência , Síndrome de Smith-Lemli-Opitz , Dicloridrato de trans-1,4-Bis(2-clorobenzaminometil)ciclo-hexano/toxicidade , Animais , Encéfalo/embriologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Desenvolvimento Embrionário e Fetal/genética , Proteínas do Olho , Feminino , Doenças Fetais/induzido quimicamente , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/genética , Idade Gestacional , Proteínas Hedgehog , Hibridização In Situ , Masculino , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/genética , Fatores de Transcrição Otx , Oxirredutases/antagonistas & inibidores , Oxirredutases/genética , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados , Gravidez , Prosencéfalo/embriologia , Prosencéfalo/patologia , Biossíntese de Proteínas , Proteínas/genética , Proteínas/fisiologia , Ratos , Ratos Wistar , Proteínas Repressoras , Rombencéfalo/embriologia , Rombencéfalo/patologia , Transdução de Sinais/fisiologia , Síndrome de Smith-Lemli-Opitz/genética , Transativadores/biossíntese , Transativadores/genética
16.
Brain Res Mol Brain Res ; 98(1-2): 58-66, 2002 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-11834296

RESUMO

Dehydroepiandrosterone (DHEA) can function to protect neural precursors and their progeny targeted with toxic insults; however, the molecular mechanisms underlying the neuroprotective effects of DHEA are not understood. We cultured neural precursors from the embryonic forebrain of rats and examined the effects of DHEA and its sulfated derivative (DHEAS) on the activation of the serine-threonine protein kinase Akt, which is widely implicated in cell survival signaling. We found that DHEA activated Akt in neural precursor culture, in association with a decrease in apoptosis. In contrast, DHEAS decreased activated Akt levels and increased apoptosis. The effects of DHEA on neural cell survival and activation of Akt were not blocked by the steroid hormone antagonists flutamide and tamoxifen, but both were blocked by a PI3-K inhibitor, LY294002. These findings suggest that during neurogenesis in the developing cortex, DHEA and DHEAS regulate the survival of neural precursors and progeny through the Akt signaling pathway.


Assuntos
Apoptose/efeitos dos fármacos , Sulfato de Desidroepiandrosterona/farmacologia , Desidroepiandrosterona/farmacologia , Proteínas Fetais/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/efeitos dos fármacos , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais/efeitos dos fármacos , Telencéfalo/embriologia , Antagonistas de Androgênios/farmacologia , Animais , Cromonas/farmacologia , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Proteínas Fetais/antagonistas & inibidores , Flutamida/farmacologia , Morfolinas/farmacologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neurônios/citologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Sprague-Dawley , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Telencéfalo/citologia
17.
J Immunol Methods ; 415: 57-62, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25260423

RESUMO

The complement system is an important aspect of immune defense against microbial invasion. Eukaryotic cells express various complement regulatory proteins to protect them from uncontrolled complement activation. However, some eukaryotic cells possess constitutive complement system activation that does not require specific triggering factors, which is known to have unexpected effects on cell proliferation and survival. This area of research is still preliminary and a standard method to measure complement system activation in eukaryotic cells has yet to be identified. Here, we present a quantitative in vitro method to measure complement system activation in eukaryotic cells by detecting C5b-9, the membrane attack complex, on cell surfaces. The results obtained using this assay correlated with C3b deposition measured using flow cytometry and C5b-9 deposition detected using an immunofluorescence assay. Furthermore, we showed that various cancer cell lines displayed different levels of complement system activation by using this assay.


Assuntos
Membrana Celular/química , Ativação do Complemento , Complexo de Ataque à Membrana do Sistema Complemento/análise , Ensaio de Imunoadsorção Enzimática/métodos , Anticorpos/farmacologia , Antígenos CD/genética , Antígenos CD/imunologia , Moléculas de Adesão Celular Neuronais/antagonistas & inibidores , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/imunologia , Linhagem Celular Tumoral , Membrana Celular/imunologia , Complemento C3b/farmacologia , Endoglina , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/genética , Proteínas Fetais/imunologia , Citometria de Fluxo , Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/imunologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Especificidade de Órgãos , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia
18.
J Cell Biol ; 202(7): 1075-90, 2013 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-24062339

RESUMO

Vaccinia virus dissemination relies on the N-WASP-ARP2/3 pathway, which mediates actin tail formation underneath cell-associated extracellular viruses (CEVs). Here, we uncover a previously unappreciated role for the formin FHOD1 and the small GTPase Rac1 in vaccinia actin tail formation. FHOD1 depletion decreased the number of CEVs forming actin tails and impaired the elongation rate of the formed actin tails. Recruitment of FHOD1 to actin tails relied on its GTPase binding domain in addition to its FH2 domain. In agreement with previous studies showing that FHOD1 is activated by the small GTPase Rac1, Rac1 was enriched and activated at the membrane surrounding actin tails. Rac1 depletion or expression of dominant-negative Rac1 phenocopied the effects of FHOD1 depletion and impaired the recruitment of FHOD1 to actin tails. FHOD1 overexpression rescued the actin tail formation defects observed in cells overexpressing dominant-negative Rac1. Altogether, our results indicate that, to display robust actin-based motility, vaccinia virus integrates the activity of the N-WASP-ARP2/3 and Rac1-FHOD1 pathways.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Actinas/metabolismo , Movimento Celular/fisiologia , Proteínas Fetais/metabolismo , Proteínas Nucleares/metabolismo , Vaccinia virus/fisiologia , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/genética , Imunofluorescência , Forminas , Células HeLa , Humanos , Processamento de Imagem Assistida por Computador , Immunoblotting , Camundongos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Ligação Proteica , RNA Interferente Pequeno/genética , Vacínia/metabolismo , Vacínia/virologia
19.
Cell Cycle ; 12(21): 3421-32, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24047696

RESUMO

Interleukin 6 (IL-6) signaling plays a role in inflammation, cancer, and senescence. Here, we identified soluble IL-6 receptor (sIL-6R) as a member of the senescence-associated secretory phenotype (SASP). Senescence-associated sIL-6R upregulation was mediated by mammalian target of rapamycin (mTOR). sIL-6R was mainly generated by a disintegrin and metalloprotease 10 (ADAM10)-dependent ectodomain shedding to enable IL-6 trans-signaling. In vivo, heterozygous PTEN-knockout mice exhibited higher mTOR activity and increased sIL-6R levels. Moreover, aberrant EGF receptor (EGFR) activation triggered IL-6 synthesis. In analogy to senescence, EGFR-induced activation of mTOR also induced IL-6R expression and sIL-6R generation. Hence, mTOR activation reprograms IL-6 non-responder cells into IL-6 responder cells. Our data suggest that mTOR serves as a central molecular switch to facilitate cellular IL-6 classic and trans-signaling via IL-6R upregulation with direct implications for cellular senescence and tumor development.


Assuntos
Transformação Celular Neoplásica/genética , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Interleucina-6/genética , Receptores de Interleucina-6/genética , Serina-Treonina Quinases TOR/genética , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Senescência Celular/genética , Receptores ErbB/metabolismo , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Humanos , Interleucina-6/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Interleucina-6/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
20.
Cancer Res ; 71(15): 5296-306, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21653678

RESUMO

The switch of tumor cells from an epithelial to a mesenchymal-like phenotype [designated as epithelial-to-mesenchymal transition (EMT)] is known to induce tumor cell motility and invasiveness, therefore promoting metastasis of solid carcinomas. Although multiple studies have focused on elucidating the signaling events that initiate this phenotypic switch, there has been so far no characterization of the pattern of soluble mediators released by tumor cells undergoing EMT, and the potential impact that this phenotypic switch could have on the remodeling of the tumor microenvironment. Here we show that induction of EMT in human carcinoma cells via overexpression of the transcription factor Brachyury is associated with enhanced secretion of multiple cytokines, chemokines, and angiogenic factors and, in particular, with the induction of the IL-8/IL-8R axis. Our results also indicate the essential role of interleukin 8 (IL-8) signaling for the acquisition and/or maintenance of the mesenchymal and invasive features of Brachyury-overexpressing tumor cells and show that IL-8 secreted by tumor cells undergoing EMT could potentiate tumor progression by inducing adjacent epithelial tumor cells into EMT. Altogether, our results emphasize the potential role of EMT in the modulation of the tumor microenvironment via secretion of multiple soluble mediators and suggest that IL-8 signaling blockade may provide a means of targeting mesenchymal-like, invasive tumor cells.


Assuntos
Carcinoma/patologia , Transição Epitelial-Mesenquimal/fisiologia , Interleucina-8/fisiologia , Proteínas de Neoplasias/fisiologia , Microambiente Tumoral/fisiologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Efeito Espectador , Carcinoma/metabolismo , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/patologia , Movimento Celular , Quimiocinas/metabolismo , Meios de Cultivo Condicionados/farmacologia , Meios de Cultura Livres de Soro , Citocinas/metabolismo , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/biossíntese , Proteínas Fetais/genética , Proteínas Fetais/fisiologia , Fibronectinas/biossíntese , Fibronectinas/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Invasividade Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Regiões Promotoras Genéticas , RNA Interferente Pequeno/farmacologia , Receptores de Interleucina-8/biossíntese , Receptores de Interleucina-8/genética , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas com Domínio T/antagonistas & inibidores , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/genética , Proteínas com Domínio T/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA