Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Commun ; 15(1): 3909, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38724493

RESUMO

Aberrant signaling pathway activity is a hallmark of tumorigenesis and progression, which has guided targeted inhibitor design for over 30 years. Yet, adaptive resistance mechanisms, induced by rapid, context-specific signaling network rewiring, continue to challenge therapeutic efficacy. Leveraging progress in proteomic technologies and network-based methodologies, we introduce Virtual Enrichment-based Signaling Protein-activity Analysis (VESPA)-an algorithm designed to elucidate mechanisms of cell response and adaptation to drug perturbations-and use it to analyze 7-point phosphoproteomic time series from colorectal cancer cells treated with clinically-relevant inhibitors and control media. Interrogating tumor-specific enzyme/substrate interactions accurately infers kinase and phosphatase activity, based on their substrate phosphorylation state, effectively accounting for signal crosstalk and sparse phosphoproteome coverage. The analysis elucidates time-dependent signaling pathway response to each drug perturbation and, more importantly, cell adaptive response and rewiring, experimentally confirmed by CRISPR knock-out assays, suggesting broad applicability to cancer and other diseases.


Assuntos
Neoplasias do Colo , Resistencia a Medicamentos Antineoplásicos , Fosfoproteínas , Proteômica , Transdução de Sinais , Humanos , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Proteômica/métodos , Fosfoproteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Neoplasias do Colo/genética , Linhagem Celular Tumoral , Fosforilação , Algoritmos , Proteoma/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico
2.
bioRxiv ; 2023 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-36824919

RESUMO

Aberrant signaling pathway activity is a hallmark of tumorigenesis and progression, which has guided targeted inhibitor design for over 30 years. Yet, adaptive resistance mechanisms, induced by rapid, context-specific signaling network rewiring, continue to challenge therapeutic efficacy. By leveraging progress in proteomic technologies and network-based methodologies, over the past decade, we developed VESPA-an algorithm designed to elucidate mechanisms of cell response and adaptation to drug perturbations-and used it to analyze 7-point phosphoproteomic time series from colorectal cancer cells treated with clinically-relevant inhibitors and control media. Interrogation of tumor-specific enzyme/substrate interactions accurately inferred kinase and phosphatase activity, based on their inferred substrate phosphorylation state, effectively accounting for signal cross-talk and sparse phosphoproteome coverage. The analysis elucidated time-dependent signaling pathway response to each drug perturbation and, more importantly, cell adaptive response and rewiring that was experimentally confirmed by CRISPRko assays, suggesting broad applicability to cancer and other diseases.

3.
Cell ; 184(2): 334-351.e20, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33434495

RESUMO

Despite considerable efforts, the mechanisms linking genomic alterations to the transcriptional identity of cancer cells remain elusive. Integrative genomic analysis, using a network-based approach, identified 407 master regulator (MR) proteins responsible for canalizing the genetics of individual samples from 20 cohorts in The Cancer Genome Atlas (TCGA) into 112 transcriptionally distinct tumor subtypes. MR proteins could be further organized into 24 pan-cancer, master regulator block modules (MRBs), each regulating key cancer hallmarks and predictive of patient outcome in multiple cohorts. Of all somatic alterations detected in each individual sample, >50% were predicted to induce aberrant MR activity, yielding insight into mechanisms linking tumor genetics and transcriptional identity and establishing non-oncogene dependencies. Genetic and pharmacological validation assays confirmed the predicted effect of upstream mutations and MR activity on downstream cellular identity and phenotype. Thus, co-analysis of mutational and gene expression profiles identified elusive subtypes and provided testable hypothesis for mechanisms mediating the effect of genetic alterations.


Assuntos
Neoplasias/genética , Transcrição Gênica , Adenocarcinoma/genética , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Genoma Humano , Células HEK293 , Humanos , Camundongos Nus , Mutação/genética , Reprodutibilidade dos Testes
4.
Nat Genet ; 50(7): 979-989, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29915428

RESUMO

We introduce and validate a new precision oncology framework for the systematic prioritization of drugs targeting mechanistic tumor dependencies in individual patients. Compounds are prioritized on the basis of their ability to invert the concerted activity of master regulator proteins that mechanistically regulate tumor cell state, as assessed from systematic drug perturbation assays. We validated the approach on a cohort of 212 gastroenteropancreatic neuroendocrine tumors (GEP-NETs), a rare malignancy originating in the pancreas and gastrointestinal tract. The analysis identified several master regulator proteins, including key regulators of neuroendocrine lineage progenitor state and immunoevasion, whose role as critical tumor dependencies was experimentally confirmed. Transcriptome analysis of GEP-NET-derived cells, perturbed with a library of 107 compounds, identified the HDAC class I inhibitor entinostat as a potent inhibitor of master regulator activity for 42% of metastatic GEP-NET patients, abrogating tumor growth in vivo. This approach may thus complement current efforts in precision oncology.


Assuntos
Antineoplásicos/farmacologia , Tumores Neuroendócrinos/tratamento farmacológico , Benzamidas/farmacologia , Linhagem Celular Tumoral , Estudos de Coortes , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Humanos , Neoplasias Intestinais/tratamento farmacológico , Neoplasias Intestinais/genética , Tumores Neuroendócrinos/genética , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Medicina de Precisão/métodos , Piridinas/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética
5.
Cell Stem Cell ; 18(4): 441-55, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27058937

RESUMO

The existence of adult pancreatic progenitor cells has been debated. While some favor the concept of facultative progenitors involved in homeostasis and repair, neither a location nor markers for such cells have been defined. Using genetic lineage tracing, we show that Doublecortin-like kinase-1 (Dclk1) labels a rare population of long-lived, quiescent pancreatic cells. In vitro, Dclk1+ cells proliferate readily and sustain pancreatic organoid growth. In vivo, Dclk1+ cells are necessary for pancreatic regeneration following injury and chronic inflammation. Accordingly, their loss has detrimental effects after cerulein-induced pancreatitis. Expression of mutant Kras in Dclk1+ cells does not affect their quiescence or longevity. However, experimental pancreatitis converts Kras mutant Dclk1+ cells into potent cancer-initiating cells. As a potential effector of Kras, Dclk1 contributes functionally to the pathogenesis of pancreatic cancer. Taken together, these observations indicate that Dclk1 marks quiescent pancreatic progenitors that are candidates for the origin of pancreatic cancer.


Assuntos
Carcinogênese/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Pancreatite/metabolismo , Pancreatite/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Administração Oral , Animais , Carcinogênese/patologia , Carcinoma Ductal Pancreático/induzido quimicamente , Carcinoma Ductal Pancreático/etiologia , Carcinoma Ductal Pancreático/patologia , Proliferação de Células , Quinases Semelhantes a Duplacortina , Camundongos , Organoides/citologia , Organoides/crescimento & desenvolvimento , Neoplasias Pancreáticas/induzido quimicamente , Neoplasias Pancreáticas/etiologia , Pancreatite/induzido quimicamente , Pancreatite/complicações , Proteínas Serina-Treonina Quinases/genética , Tamoxifeno/administração & dosagem
6.
Cancer Cell ; 21(4): 459-72, 2012 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-22516257

RESUMO

Constitutive phosphoinositide 3-kinase (PI3K)/Akt activation is common in T cell acute lymphoblastic leukemia (T-ALL). Although four distinct class I PI3K isoforms (α, ß, γ, δ) could participate in T-ALL pathogenesis, none has been implicated in this process. We report that in the absence of PTEN phosphatase tumor suppressor function, PI3Kγ or PI3Kδ alone can support leukemogenesis, whereas inactivation of both isoforms suppressed tumor formation. The reliance of PTEN null T-ALL on the combined activities of PI3Kγ/δ was further demonstrated by the ability of a dual inhibitor to reduce disease burden and prolong survival in mice as well as prevent proliferation and promote activation of proapoptotic pathways in human tumors. These results support combined inhibition of PI3Kγ/δ as therapy for T-ALL.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores de Fosfoinositídeo-3 Quinase , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Isoformas de Proteínas , Purinas/uso terapêutico , Quinazolinonas/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Classe I de Fosfatidilinositol 3-Quinases , Classe Ib de Fosfatidilinositol 3-Quinase/química , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Desenho de Fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Camundongos , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Purinas/química , Purinas/farmacologia , Quinazolinonas/química , Quinazolinonas/farmacologia
7.
FEBS Lett ; 582(11): 1569-74, 2008 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-18405666

RESUMO

Interferon (IFN) gamma's ability to localise in the nucleus and function in gene activation has been known for some time, although the role of the conventional nuclear transporting importin molecules is unclear. Here, we demonstrate for the first time the direct recognition of IFNgamma and an IFNgamma mimetic peptide by IMPalpha and the IMPalpha/beta heterodimer, where the IFNgamma mimetic shows higher affinity. Significantly, this correlates well both with in vivo ability to target green fluorescent protein to the nucleus in transfected cells as determined by quantitative confocal laser scanning microscopy, as well as GAS promoter activity of a luciferase reporter. This has important implications for IFNgamma's anti-viral action, and the potential use of the IFNgamma mimetic in antiviral therapies.


Assuntos
Núcleo Celular/metabolismo , Interferon gama/metabolismo , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo , Sequência de Aminoácidos , Animais , Materiais Biomiméticos/metabolismo , Células COS , Chlorocebus aethiops , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interferon gama/genética , Dados de Sequência Molecular , Sinais de Localização Nuclear/metabolismo , Peptídeos , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ativação Transcricional
8.
J Virol ; 79(9): 5632-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15827178

RESUMO

We have developed peptide mimetics of gamma interferon (IFN-gamma) that play a direct role in the activation and nuclear translocation of STAT1alpha transcription factor. These mimetics do not act through recognition by the extracellular domain of IFN-gamma receptor but rather bind to the cytoplasmic domain of the receptor chain 1, IFNGR-1, and thereby initiate the cellular signaling. Thus, we hypothesized that these mimetics would bypass the poxvirus virulence factor B8R protein that binds to intact IFN-gamma and prevents its interaction with the receptor. Human and murine IFN-gamma mimetic peptides were introduced into an adenoviral vector for intracellular expression. Murine IFN-gamma mimetic peptide was also expressed via chemical synthesis with an attached lipophilic group for penetration of cell plasma membrane. In contrast to intact human IFN-gamma, the mimetics did not bind poxvirus B8R protein, a homolog of the IFN-gamma receptor extracellular domain. Expression of B8R protein in WISH cells did not block the antiviral effect of the mimetics against encephalomyocarditis or vesicular stomatitis virus, while the antiviral activity of human IFN-gamma was neutralized. Consistent with the antiviral activity, the upregulation of MHC class I molecules on WISH cells by the IFN-gamma mimetics was not affected by B8R protein, while IFN-gamma-induced upregulation was blocked. Finally, the mimetics, but not IFN-gamma, inhibited vaccinia virus replication in African green monkey kidney BSC-40 cells. The data presented demonstrate that small peptide mimetics of IFN-gamma can avoid the B8R virulence factor for poxviruses and, thus, are potential candidates for antivirals against smallpox virus.


Assuntos
Biomimética , Vírus da Encefalomiocardite/efeitos dos fármacos , Vírus da Encefalomiocardite/metabolismo , Interferon gama/farmacologia , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/metabolismo , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/metabolismo , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Adenoviridae/genética , Antivirais/farmacologia , Linhagem Celular , Vetores Genéticos , Humanos , Interferon gama/síntese química , Interferon gama/genética , Replicação Viral/efeitos dos fármacos
9.
Oncogene ; 24(12): 2114-20, 2005 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-15688010

RESUMO

Prostate cancer is the second highest cause of cancer-related deaths of men in the US. Signal transducers and activators of transcription (STATs) proteins are a small family of latent cytoplasmic transcription factors that act downstream of Janus kinase (JAK) activation and mediate intracellular signaling from a wide variety of cytokines, growth factors, and hormones. Aberrant activation of STAT3 has been implicated in the progression of many human carcinomas, including prostate cancer. Previously, we have characterized a novel tyrosine kinase inhibitor peptide, Tkip, that is a mimetic of suppressor of cytokine signaling 1 (SOCS-1). Similar to SOCS-1, Tkip binds to the autophosphorylation site of JAK2 and inhibits phosphorylation of STAT1alpha. In this study, we determined the inhibitory effects of Tkip on the human prostate cancer cell lines DU145 and LNCaP. Tkip inhibited cellular proliferation of both DU145 and LNCaP cells, with a slightly greater antiproliferative effect on DU145 cells. Cell cycle analysis using flow cytometry showed Tkip blockage of progression into the S phase of the cell cycle. Tkip also inhibited constitutive (DU145) and IL-6-induced (LNCaP) activation of STAT3, consistent with the fact that STAT3 activation is mediated by JAK2. Tkip also slightly reduced the levels of cyclin D1, an important regulator of cell cycle progression into S phase, in DU145 and LNCaP cancer cell lines. These data describe a potentially important therapeutic that targets both constitutive and IL-6-induced STAT3 activation in human prostate cancer cell lines.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Interleucina-6/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fragmentos de Peptídeos/farmacologia , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Sequência de Aminoácidos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Janus Quinase 2 , Masculino , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Neoplasias da Próstata , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Fator de Transcrição STAT3 , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina
10.
FEBS Lett ; 578(3): 207-10, 2004 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-15589821

RESUMO

A nuclear localization sequence (NLS) in the type II interferon (IFN) IFN gamma, which is responsible for the nuclear translocation of both the ligand and the alpha-subunit (IFNGR1) of the receptor complex, has previously been characterized and its role in signaling examined in detail. We have now identified an NLS in the type I IFN receptor (IFNAR) common subunit IFNAR1 from humans and show that the human IFNAR1 subunit can translocate to the nucleus following human IFN beta stimulation. An NLS in human IFNAR1 is located in the extracellular domain of IFNAR1 within the sequence (382)RKIIEKKT (numbered for the precursor form). Nuclear import by the NLS functions in a conventional fashion requiring cytosolic import factors, is energy-dependent and inhibited by the prototypical NLS of the SV40 large T-antigen. These studies provide a mechanism for nuclear import of IFNAR1, as well as for type I IFN ligands, and a starting point for studying an alternate role for IFNAR1 in nuclear signaling within the type I IFN system.


Assuntos
Interferon Tipo I/metabolismo , Interferon gama/metabolismo , Sinais de Localização Nuclear , Receptores de Interferon/química , Receptores de Interferon/metabolismo , Transporte Ativo do Núcleo Celular , Trifosfato de Adenosina/metabolismo , Antígenos Transformantes de Poliomavirus/química , Antígenos Transformantes de Poliomavirus/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Células Clonais , Citosol/metabolismo , Metabolismo Energético , Imunofluorescência , Guanosina Trifosfato/metabolismo , Humanos , Interferon Tipo I/imunologia , Interferon beta/genética , Interferon beta/farmacologia , Interferon gama/química , Interferon gama/genética , Cinética , Ligantes , Proteínas de Membrana , Ficocianina/metabolismo , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Receptor de Interferon alfa e beta , Receptores de Interferon/imunologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Temperatura
11.
J Immunol ; 172(12): 7510-8, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15187130

RESUMO

Positive and negative regulation of cytokines such as IFN-gamma are key to normal homeostatic function. Negative regulation of IFN-gamma in cells occurs via proteins called suppressors of cytokine signaling (SOCS)1 and -3. SOCS-1 inhibits IFN-gamma function by binding to the autophosphorylation site of the tyrosine kinase Janus kinase (JAK)2. We have developed a short 12-mer peptide, WLVFFVIFYFFR, that binds to the autophosphorylation site of JAK2, resulting in inhibition of its autophosphorylation as well as its phosphorylation of IFN-gamma receptor subunit IFNGR-1. The JAK2 tyrosine kinase inhibitor peptide (Tkip) did not bind to or inhibit tyrosine autophosphorylation of vascular endothelial growth factor receptor or phosphorylation of a substrate peptide by the protooncogene tyrosine kinase c-src. Tkip also inhibited epidermal growth factor receptor autophosphorylation, consistent with the fact that epidermal growth factor receptor is regulated by SOCS-1 and SOCS-3, similar to JAK2. Although Tkip binds to unphosphorylated JAK2 autophosphorylation site peptide, it binds significantly better to tyrosine-1007 phosphorylated JAK2 autophosphorylation site peptide. SOCS-1 only recognizes the JAK2 site in its phosphorylated state. Thus, Tkip recognizes the JAK2 autophosphorylation site similar to SOCS-1, but not precisely the same way. Consistent with inhibition of JAK2, Tkip inhibited the ability of IFN-gamma to induce an antiviral state as well as up-regulate MHC class I molecules on cells at a concentration of approximately 10 microM. This is similar to the K(d) of SOCS-3 for the erythropoietin receptor. These data represent a proof-of-concept demonstration of a peptide mimetic of SOCS-1 that regulates JAK2 tyrosine kinase function.


Assuntos
Proteínas de Transporte/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Fragmentos de Peptídeos/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas , Proteínas Repressoras/fisiologia , Animais , Aorta , Proteínas de Transporte/química , Bovinos , Linhagem Celular , Quinases Ciclina-Dependentes , Endotélio Vascular/citologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Fibroblastos , Regulação da Expressão Gênica/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/biossíntese , Humanos , Interferon gama/antagonistas & inibidores , Janus Quinase 2 , Camundongos , Mimetismo Molecular , Fragmentos de Peptídeos/química , Fosforilação/efeitos dos fármacos , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Repressoras/química , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina , Fatores de Transcrição/química , Fatores de Transcrição/fisiologia
12.
Biochemistry ; 43(18): 5445-54, 2004 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-15122910

RESUMO

The C-terminus of interferon-gamma (IFNgamma) contains a nuclear localization sequence (NLS) required for the activation and nuclear translocation of the transcription factor STAT1alpha and induction of IFNgamma-activated genes. On the basis of this and other studies, we developed a peptide mimetic of IFNgamma that possesses the IFNgamma functions of antiviral activity and upregulation of MHC class II molecules. The mimetic also shares with IFNgamma the ability to induce the activation and nuclear translocation of STAT1alpha and the IFNgamma receptor (IFNGR)-1 subunit. The mimetic, IFNgamma(95-132), is a peptide that consists of the C-terminal residues 95-132 of murine IFNgamma and contains a required alpha-helical domain and the NLS of IFNgamma. In this study, we determined the mechanism of the intracellular action of the mimetic at the level of signal transduction. We show that the mimetic mediates the nuclear transport of IFNGR-1 through its interaction with IFNGR-1 cytoplasmic region 253-287 via both the helical region and the NLS of IFNgamma(95-132). Alanine substitutions of the NLS of the mimetic showed that the NLS was required for nuclear translocation and that the nuclear transport properties of the mimetic correlated with its ability to bind IFNGR-1. These data also show that the NLS of IFNgamma(95-132) can interact simultaneously with IFNGR-1 and the nuclear import machinery. We found that in in vitro nuclear transport assays tyrosine-phosphorylated STAT1alpha failed to undergo nuclear translocation in the presence of nuclear import factors, but was transported to nucleus in the presence of IFNgamma(95-132) and JAK2-phosphorylated IFNGR-1, to which STAT1alpha binds, as a complex of IFNgamma(95-132)/IFNGR-1/STAT1alpha. Thus, the mimetic, which possesses IFNgamma function, is directly involved as a chaperone in the nuclear transport of STAT1alpha and shares this mechanism of action with that previously described for IFNgamma. The mimetic, like IFNgamma, is able to upregulate the tumor suppressor p21WAF1/CIP1, a direct target of STAT1alpha, and this ability requires the NLS of the mimetic. However, unlike IFNgamma, the mimetic is unable to downregulate c-myc and hence does not inhibit the cycling of cells. This suggests that IFNgamma has additional functions that are not tied directly to the nuclear translocation of STAT1alpha.


Assuntos
Interferon gama/fisiologia , Fragmentos de Peptídeos/fisiologia , Transdução de Sinais , Transporte Ativo do Núcleo Celular/genética , Alanina/genética , Sequência de Aminoácidos , Animais , Ligação Competitiva/genética , Ciclo Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Fator Gênico 3 Estimulado por Interferon , Interferon gama/síntese química , Interferon gama/genética , Interferon gama/metabolismo , Camundongos , Chaperonas Moleculares/genética , Chaperonas Moleculares/fisiologia , Mimetismo Molecular/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/metabolismo , Sinais de Localização Nuclear/fisiologia , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosforilação , Ligação Proteica/genética , Estrutura Secundária de Proteína/genética , Receptores de Interferon/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo , Receptor de Interferon gama
13.
J Cell Sci ; 116(Pt 15): 3089-98, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12799413

RESUMO

Intracellularly expressed interferon gamma (IFNgamma) has been reported to possess biological activity similar to that of IFNgamma added to cells. This study addresses the mechanisms for such similar biological effects. Adenoviral vectors were used to express a non-secreted form of human IFNgamma or a non-secreted mutant form in which a previously demonstrated nuclear localization sequence (NLS), 128KTGKRKR134, was replaced with alanines at K and R positions. With the vector expressing non-secreted wild-type IFNgamma, biological responses normally associated with extracellular IFNgamma, such as antiviral activity and MHC class I upregulation, were observed, although the mutant IFNgamma did not possess biological activity. Intracellular human IFNgamma possessed biological activity in mouse L cells, which do not recognize extracellularly added human IFNgamma. Thus, the biological activity was not due to leakage of IFNgamma to the surroundings and subsequent interaction with the receptor on the cell surface. Biological function was associated with activation of STAT1alpha and nuclear translocation of IFNgamma, IFNGR1 and STAT1alpha. Immunoprecipitation of cellular extracts with antibody to the nuclear transporter NPI-1 showed the formation of a complex with IFNgamma-IFNGR1-STAT1alpha. To provide the physiological basis for these effects we show that extracellularly added IFNgamma possesses intracellular signaling activity that is NLS dependent, as suggested by our previous studies, and that this activity occurs via the receptor-mediated endocytosis of IFNgamma. The data are consistent with previous observations that the NLS of extracellularly added IFNgamma plays a role in IFNgamma signaling.


Assuntos
Interferon gama/metabolismo , Sinais de Localização Nuclear/metabolismo , Isoformas de Proteínas/metabolismo , Transdução de Sinais/fisiologia , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Regulação da Expressão Gênica/fisiologia , Genes MHC Classe I/fisiologia , Vetores Genéticos , Humanos , Fator Gênico 3 Estimulado por Interferon , Interferon gama/genética , Camundongos , Mutação , Fosforilação , Ligação Proteica , Isoformas de Proteínas/genética , Estrutura Terciária de Proteína , Receptores de Interferon/metabolismo , Fatores de Transcrição/metabolismo , alfa Carioferinas/metabolismo , Receptor de Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA