Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nat Commun ; 15(1): 3909, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724493

RESUMEN

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.


Asunto(s)
Neoplasias del Colon , Resistencia a Antineoplásicos , Fosfoproteínas , Proteómica , Transducción de Señal , Humanos , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Proteómica/métodos , Fosfoproteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/genética , Línea Celular Tumoral , Fosforilación , Algoritmos , Proteoma/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
2.
bioRxiv ; 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37873470

RESUMEN

The Mechanism of Action (MoA) of a drug is generally represented as a small, non-tissue-specific repertoire of high-affinity binding targets. Yet, drug activity and polypharmacology are increasingly associated with a broad range of off-target and tissue-specific effector proteins. To address this challenge, we have implemented an efficient integrative experimental and computational framework leveraging the systematic generation and analysis of drug perturbational profiles representing >700 FDA-approved and experimental oncology drugs, in cell lines selected as high-fidelity models of 23 aggressive tumor subtypes. Protein activity-based analyses revealed highly reproducible, drug-mediated modulation of tissue-specific targets, leading to generation of a proteome-wide polypharmacology map, characterization of MoA-related drug clusters and off-target effects, and identification and experimental validation of novel, tissue-specific inhibitors of undruggable oncoproteins. The proposed framework, which is easily extended to elucidating the MoA of novel small-molecule libraries, could help support more systematic and quantitative approaches to precision oncology.

3.
bioRxiv ; 2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36824919

RESUMEN

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.

4.
Cell ; 184(2): 334-351.e20, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33434495

RESUMEN

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.


Asunto(s)
Neoplasias/genética , Transcripción Genética , Adenocarcinoma/genética , Animales , Línea Celular Tumoral , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Genoma Humano , Células HEK293 , Humanos , Ratones Desnudos , Mutación/genética , Reproducibilidad de los Resultados
5.
Cell Rep ; 33(10): 108474, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33296649

RESUMEN

Bi-species, fusion-mediated, somatic cell reprogramming allows precise, organism-specific tracking of unknown lineage drivers. The fusion of Tcf7l1-/- murine embryonic stem cells with EBV-transformed human B cell lymphocytes, leads to the generation of bi-species heterokaryons. Human mRNA transcript profiling at multiple time points permits the tracking of the reprogramming of B cell nuclei to a multipotent state. Interrogation of a human B cell regulatory network with gene expression signatures identifies 8 candidate master regulator proteins. Of these 8 candidates, ectopic expression of BAZ2B, from the bromodomain family, efficiently reprograms hematopoietic committed progenitors into a multipotent state and significantly enhances their long-term clonogenicity, stemness, and engraftment in immunocompromised mice. Unbiased systems biology approaches let us identify the early driving events of human B cell reprogramming.


Asunto(s)
Reprogramación Celular/genética , Células Madre Hematopoyéticas/metabolismo , Factores Generales de Transcripción/metabolismo , Animales , Linfocitos B/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Reprogramación Celular/fisiología , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Femenino , Sangre Fetal/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Células Madre Multipotentes/metabolismo , Factores de Transcripción/metabolismo , Factores Generales de Transcripción/genética , Factores Generales de Transcripción/fisiología
6.
Nat Genet ; 50(7): 979-989, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29915428

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Tumores Neuroendocrinos/tratamiento farmacológico , Benzamidas/farmacología , Línea Celular Tumoral , Estudios de Cohortes , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Humanos , Neoplasias Intestinales/tratamiento farmacológico , Neoplasias Intestinales/genética , Tumores Neuroendocrinos/genética , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Medicina de Precisión/métodos , Piridinas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética
7.
Cell Stem Cell ; 18(4): 441-55, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27058937

RESUMEN

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.


Asunto(s)
Carcinogénesis/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pancreatitis/metabolismo , Pancreatitis/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Administración Oral , Animales , Carcinogénesis/patología , Carcinoma Ductal Pancreático/inducido químicamente , Carcinoma Ductal Pancreático/etiología , Carcinoma Ductal Pancreático/patología , Proliferación Celular , Quinasas Similares a Doblecortina , Ratones , Organoides/citología , Organoides/crecimiento & desarrollo , Neoplasias Pancreáticas/inducido químicamente , Neoplasias Pancreáticas/etiología , Pancreatitis/inducido químicamente , Pancreatitis/complicaciones , Proteínas Serina-Treonina Quinasas/genética , Tamoxifeno/administración & dosificación
8.
Cell ; 162(2): 441-451, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26186195

RESUMEN

Genome-wide identification of the mechanism of action (MoA) of small-molecule compounds characterizing their targets, effectors, and activity modulators represents a highly relevant yet elusive goal, with critical implications for assessment of compound efficacy and toxicity. Current approaches are labor intensive and mostly limited to elucidating high-affinity binding target proteins. We introduce a regulatory network-based approach that elucidates genome-wide MoA proteins based on the assessment of the global dysregulation of their molecular interactions following compound perturbation. Analysis of cellular perturbation profiles identified established MoA proteins for 70% of the tested compounds and elucidated novel proteins that were experimentally validated. Finally, unknown-MoA compound analysis revealed altretamine, an anticancer drug, as an inhibitor of glutathione peroxidase 4 lipid repair activity, which was experimentally confirmed, thus revealing unexpected similarity to the activity of sulfasalazine. This suggests that regulatory network analysis can provide valuable mechanistic insight into the elucidation of small-molecule MoA and compound similarity.


Asunto(s)
Algoritmos , Antineoplásicos/farmacología , Terapia Molecular Dirigida , Antineoplásicos/química , Epistasis Genética , Estudio de Asociación del Genoma Completo , Neoplasias/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas
9.
Cancer Cell ; 21(4): 459-72, 2012 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-22516257

RESUMEN

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.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Isoformas de Proteínas , Purinas/uso terapéutico , Quinazolinonas/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Fosfatidilinositol 3-Quinasa Clase I , Fosfatidilinositol 3-Quinasa Clase Ib/química , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Diseño de Fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Ratones , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Purinas/química , Purinas/farmacología , Quinazolinonas/química , Quinazolinonas/farmacología
10.
Bioorg Med Chem ; 17(14): 5027-37, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19553129

RESUMEN

VIM-2 is an Ambler class B metallo-beta-lactamase (MBL) capable of hydrolyzing a broad-spectrum of beta-lactam antibiotics. Although the discovery and development of MBL inhibitors continue to be an area of active research, an array of potent, small molecule inhibitors is yet to be fully characterized for VIM-2. In the presented research, a compound library screening approach was used to identify and characterize VIM-2 inhibitors from a library of pharmacologically active compounds as well as a focused 'click' chemistry library. The four most potent VIM-2 inhibitors resulting from a VIM-2 screen were characterized by kinetic studies in order to determine K(i) and mechanism of enzyme inhibition. As a result, two previously described pharmacologic agents, mitoxantrone (1,4-dihydroxy-5,8-bis([2-([2-hydroxyethyl]amino)ethyl]amino)-9,10-anthracenedione) and 4-chloromercuribenzoic acid (pCMB) were found to be active, the former as a non-competitive inhibitor (K(i)=K(i)(')=1.5+/-0.2microM) and the latter as a slowly reversible or irreversible inhibitor. Additionally, two novel sulfonyl-triazole analogs from the click library were identified as potent, competitive VIM-2 inhibitors: N-((4-((but-3-ynyloxy)methyl)-1H-1,2,3-triazol-5-yl)methyl)-4-iodobenzenesulfonamide (1, K(i)=0.41+/-0.03microM) and 4-iodo-N-((4-(methoxymethyl)-1H-1,2,3-triazol-5-yl)methyl)benzenesulfonamide (2, K(i)=1.4+/-0.10microM). Mitoxantrone and pCMB were also found to potentiate imipenem efficacy in MIC and synergy assays employing Escherichia coli. Taken together, all four compounds represent useful chemical probes to further investigate mechanisms of VIM-2 inhibition in biochemical and microbiology-based assays.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Escherichia coli/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Inhibidores de beta-Lactamasas , beta-Lactamasas/metabolismo , Analgésicos/farmacología , Antibacterianos/farmacología , Dominio Catalítico , Sinergismo Farmacológico , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Escherichia coli/enzimología , Imipenem/farmacología , Pruebas de Sensibilidad Microbiana , Mitoxantrona/farmacología , Modelos Moleculares , Unión Proteica , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , beta-Lactamasas/química , Ácido p-Cloromercuribenzoico/farmacología
11.
FEBS Lett ; 582(11): 1569-74, 2008 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-18405666

RESUMEN

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.


Asunto(s)
Núcleo Celular/metabolismo , Interferón gamma/metabolismo , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo , Secuencia de Aminoácidos , Animales , Materiales Biomiméticos/metabolismo , Células COS , Chlorocebus aethiops , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interferón gamma/genética , Datos de Secuencia Molecular , Señales de Localización Nuclear/metabolismo , Péptidos , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Activación Transcripcional
13.
J Virol ; 79(9): 5632-9, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15827178

RESUMEN

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.


Asunto(s)
Biomimética , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/metabolismo , Interferón gamma/farmacología , Virus Vaccinia/efectos de los fármacos , Virus Vaccinia/metabolismo , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Virus de la Estomatitis Vesicular Indiana/metabolismo , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Adenoviridae/genética , Antivirales/farmacología , Línea Celular , Vectores Genéticos , Humanos , Interferón gamma/síntesis química , Interferón gamma/genética , Replicación Viral/efectos de los fármacos
14.
Oncogene ; 24(12): 2114-20, 2005 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-15688010

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Interleucina-6/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Secuencia de Aminoácidos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Janus Quinasa 2 , Masculino , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Neoplasias de la Próstata , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Factor de Transcripción STAT3 , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas
15.
FEBS Lett ; 578(3): 207-10, 2004 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-15589821

RESUMEN

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.


Asunto(s)
Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Señales de Localización Nuclear , Receptores de Interferón/química , Receptores de Interferón/metabolismo , Transporte Activo de Núcleo Celular , Adenosina Trifosfato/metabolismo , Antígenos Transformadores de Poliomavirus/química , Antígenos Transformadores de Poliomavirus/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Células Clonales , Citosol/metabolismo , Metabolismo Energético , Técnica del Anticuerpo Fluorescente , Guanosina Trifosfato/metabolismo , Humanos , Interferón Tipo I/inmunología , Interferón beta/genética , Interferón beta/farmacología , Interferón gamma/química , Interferón gamma/genética , Cinética , Ligandos , Proteínas de la Membrana , Ficocianina/metabolismo , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Receptor de Interferón alfa y beta , Receptores de Interferón/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Temperatura
16.
Bioessays ; 26(9): 993-1004, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15351969

RESUMEN

Interaction of ligands such as epidermal growth factor and interferon-gamma with the extracellular domains of their plasma membrane receptors results in internalization followed by translocation into the nucleus of the ligand and/or receptor. There has been reluctance, however, to ascribe signaling importance to this, the focus instead being on second messenger pathways, including mobilization of kinases and inducible transcription factors (TFs). The latter, however, fails to explain the fact that so many ligands stimulate the same second messenger cascades/TFs, and yet show distinct gene activation profiles. This is particularly apt in the case of the seven STAT TFs that are held to be the mediators of the distinct cellular functions of over 60 ligands. The current review focuses on five representative nuclear localizing ligands for which there is documentation of translocation into the cytosol and nucleus through well-characterized pathways, in addition to a role in gene activation by ligand/receptor in the nucleus.


Asunto(s)
Núcleo Celular/metabolismo , Transporte Activo de Núcleo Celular , Animales , Dimerización , Factor de Crecimiento Epidérmico/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Hormona del Crecimiento/metabolismo , Humanos , Interferón gamma/metabolismo , Ligandos , Prolactina/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Interferón/metabolismo , Transducción de Señal , Activación Transcripcional , Receptor de Interferón gamma
17.
J Immunol ; 172(12): 7510-8, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15187130

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/fisiología , Péptidos y Proteínas de Señalización Intracelular , Fragmentos de Péptidos/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas , Proteínas Represoras/fisiología , Animales , Aorta , Proteínas Portadoras/química , Bovinos , Línea Celular , Quinasas Ciclina-Dependientes , Endotelio Vascular/citología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Fibroblastos , Regulación de la Expresión Génica/efectos de los fármacos , Antígenos de Histocompatibilidad Clase I/biosíntesis , Humanos , Interferón gamma/antagonistas & inhibidores , Janus Quinasa 2 , Ratones , Imitación Molecular , Fragmentos de Péptidos/química , Fosforilación/efectos de los fármacos , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Represoras/química , Proteína 1 Supresora de la Señalización de Citocinas , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Factores de Transcripción/química , Factores de Transcripción/fisiología
18.
Biochemistry ; 43(18): 5445-54, 2004 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-15122910

RESUMEN

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.


Asunto(s)
Interferón gamma/fisiología , Fragmentos de Péptidos/fisiología , Transducción de Señal , Transporte Activo de Núcleo Celular/genética , Alanina/genética , Secuencia de Aminoácidos , Animales , Unión Competitiva/genética , Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Humanos , Factor 3 de Genes Estimulados por el Interferón , Interferón gamma/síntesis química , Interferón gamma/genética , Interferón gamma/metabolismo , Ratones , Chaperonas Moleculares/genética , Chaperonas Moleculares/fisiología , Imitación Molecular/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Señales de Localización Nuclear/química , Señales de Localización Nuclear/metabolismo , Señales de Localización Nuclear/fisiología , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosforilación , Unión Proteica/genética , Estructura Secundaria de Proteína/genética , Receptores de Interferón/metabolismo , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Receptor de Interferón gamma
19.
Biol Reprod ; 71(1): 170-6, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-14985247

RESUMEN

A series of experiments were undertaken to examine the effects of interferon (IFN)-tau on regulation of prostaglandin H synthase (PGHS)-2 mRNA in bovine endometrial (BEND) cells as a means to elucidate the actions of IFN-tau to maintain pregnancy. The objective was to determine if IFN-tau mediates posttranscriptional regulation of PGHS-2 mRNA. Cells were treated with phorbol 12,13-dibutyrate (PdBu) for 3 h to induce PGHS-2 mRNA expression. Actinomycin D (0 or 1 microg/ml) or the p38 mitogen-activated protein kinase (MAPK) inhibitor, SB203580 (1 microM), were added at 3 h, followed by addition of IFN-tau (0 or 50 ng/ml) at 3.5 h and extraction of RNA at 4.5 h. The concentrations of PGHS-2 mRNA were stable between 3 and 4.5 h regardless of actinomycin D. Simultaneous treatment of PdBu-treated cells with actinomycin D and SB203580 (1 microM) decreased PGHS-2 mRNA. Addition of IFN-tau (50 ng/ml) reduced PGHS-2 mRNA, which was not observed when actinomycin D was present. Concurrent treatments of cells with SB203580 and IFN-tau (5 ng/ml) decreased concentrations of PGHS-2 mRNA in an additive manner. Although IFN-tau reduced PGHS-2 mRNA concentrations, phosphorylation of p38 MAPK was induced by IFN-tau, PdBu, and PdBu combined with IFN-tau after 10 min of treatment. Both the p38 MAPK inhibitor and IFN-tau decreased prostaglandin F(2alpha) secretion, and decreases were additive when the two were given together. In summary, activation of p38 MAPK by PdBu is required for continued presence of PGHS-2 mRNA and secretion of prostaglandin F(2alpha) in BEND cells. Interferon-tau mediates a transcription-dependent mechanism, which induces degradation of PGHS-2 mRNA. However, the consequences of an IFN-tau-induced activation of p38 MAPK warrant further investigation, because inhibition of p38 MAPK caused a degradation of PGHS-2 mRNA.


Asunto(s)
Endometrio/enzimología , Interferón Tipo I/farmacología , Isoenzimas/genética , Proteínas Gestacionales/farmacología , Prostaglandina-Endoperóxido Sintasas/genética , ARN Mensajero/metabolismo , Transcripción Genética/fisiología , Animales , Bovinos , Ciclooxigenasa 2 , Dinoprost/metabolismo , Sinergismo Farmacológico , Endometrio/citología , Endometrio/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Imidazoles/farmacología , Forbol 12,13-Dibutirato/farmacología , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Piridinas/farmacología , ARN Mensajero/antagonistas & inhibidores , Proteínas Recombinantes/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
20.
J Cell Sci ; 116(Pt 15): 3089-98, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12799413

RESUMEN

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.


Asunto(s)
Interferón gamma/metabolismo , Señales de Localización Nuclear/metabolismo , Isoformas de Proteínas/metabolismo , Transducción de Señal/fisiología , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Regulación de la Expresión Génica/fisiología , Genes MHC Clase I/fisiología , Vectores Genéticos , Humanos , Factor 3 de Genes Estimulados por el Interferón , Interferón gamma/genética , Ratones , Mutación , Fosforilación , Unión Proteica , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína , Receptores de Interferón/metabolismo , Factores de Transcripción/metabolismo , alfa Carioferinas/metabolismo , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA