Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 21(3): 241, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32047316
2.
Semin Immunol ; 43: 101300, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31771760

RESUMEN

Outbreaks of severe virus infections with the potential to cause global pandemics are increasing. In many instances these outbreaks have been newly emerging (SARS coronavirus), re-emerging (Ebola virus, Zika virus) or zoonotic (avian influenza H5N1) virus infections. In the absence of a targeted vaccine or a pathogen-specific antiviral, broad-spectrum antivirals would function to limit virus spread. Given the direct antiviral effects of type I interferons (IFNs) in inhibiting the replication of both DNA and RNA viruses at different stages of their replicative cycles, and the effects of type I IFNs on activating immune cell populations to clear virus infections, IFNs-α/ß present as ideal candidate broad-spectrum antivirals.


Asunto(s)
Coronavirus/fisiología , Brotes de Enfermedades , Ebolavirus/fisiología , Subtipo H5N1 del Virus de la Influenza A/fisiología , Interferones/metabolismo , Virosis/inmunología , Animales , Antivirales/uso terapéutico , Control de Enfermedades Transmisibles , Interacciones Huésped-Patógeno , Humanos , Replicación Viral
3.
Blood ; 133(11): 1171-1185, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30587525

RESUMEN

Aberrant activation of mTOR signaling in acute myeloid leukemia (AML) results in a survival advantage that promotes the malignant phenotype. To improve our understanding of factors that contribute to mammalian target of rapamycin (mTOR) signaling activation and identify novel therapeutic targets, we searched for unique interactors of mTOR complexes through proteomics analyses. We identify cyclin dependent kinase 9 (CDK9) as a novel binding partner of the mTOR complex scaffold protein, mLST8. Our studies demonstrate that CDK9 is present in distinct mTOR-like (CTOR) complexes in the cytoplasm and nucleus. In the nucleus, CDK9 binds to RAPTOR and mLST8, forming CTORC1, to promote transcription of genes important for leukemogenesis. In the cytoplasm, CDK9 binds to RICTOR, SIN1, and mLST8, forming CTORC2, and controls messenger RNA (mRNA) translation through phosphorylation of LARP1 and rpS6. Pharmacological targeting of CTORC complexes results in suppression of growth of primitive human AML progenitors in vitro and elicits strong antileukemic responses in AML xenografts in vivo.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , ARN Mensajero/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Antimetabolitos Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Proliferación Celular , Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/metabolismo , Citarabina/farmacología , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Desnudos , Fosforilación , Biosíntesis de Proteínas , Proteoma/análisis , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
BMC Infect Dis ; 21(1): 814, 2021 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-34388972

RESUMEN

BACKGROUND: SARS-CoV-2 infection rapidly spreads in populations due to the high rates of community transmission. Interrupting the shedding of SARS-CoV-2 may reduce the incidence of Coronavirus Disease 19 (COVID-19). Herein we provide a protocol for a cluster randomized trial that will examine the effectiveness of treatment with interferon (IFN) ß-1a compared to standard of care in limiting the transmission of SARS-CoV-2. Co-primary objectives are to determine whether IFN therapy reduces (a) the proportion of infected cases shedding SARS-CoV-2 at day 11 post randomization and (b) the incidence of transmission of SARS-CoV-2 infection from index cases to treatment-eligible household post-exposure contacts at day 11 after randomization. Secondary objectives include assessing the impact of IFN treatment on duration of viral clearance, hospitalizations and fatalities, and evaluating the safety of IFN treatment. METHODS: Three hundred and ten households, each including an index case with a recent COVID-19 diagnosis and at least one asymptomatic treatment-eligible household contact, will be randomized to receive 3 doses of 125 µg IFN ß-1a by subcutaneous administration (days 1, 6, and 11), or standard of care. All participants will be followed until day 29. DISCUSSION: The results from this trial will identify whether IFN ß treatment of mild or moderate COVID-19 cases accelerates viral clearance and prevents disease progression and whether IFN ß treatment of post-exposure contacts of COVID-19 cases reduces transmission of infection. TRIAL REGISTRATION: This trial is registered at ClinicalTrials.gov NCT04552379; date of registration September 17, 2020.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Interferón beta/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , COVID-19/diagnóstico , COVID-19/transmisión , Prueba de COVID-19 , Humanos , SARS-CoV-2 , Resultado del Tratamiento
5.
J Biol Chem ; 294(3): 827-837, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30487288

RESUMEN

Type I interferons (IFNs) induce expression of multiple genes that control innate immune responses to invoke both antiviral and antineoplastic activities. Transcription of these interferon-stimulated genes (ISGs) occurs upon activation of the canonical Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathways. Phosphorylation and acetylation are both events crucial to tightly regulate expression of ISGs. Here, using mouse embryonic fibroblasts and an array of biochemical methods including immunoblotting and kinase assays, we show that sirtuin 2 (SIRT2), a member of the NAD-dependent protein deacetylase family, is involved in type I IFN signaling. We found that SIRT2 deacetylates cyclin-dependent kinase 9 (CDK9) in a type I IFN-dependent manner and that the CDK9 deacetylation is essential for STAT1 phosphorylation at Ser-727. We also found that SIRT2 is subsequently required for the transcription of ISGs and for IFN-driven antiproliferative responses in both normal and malignant cells. These findings establish the existence of a previously unreported signaling pathway whose function is essential for the control of JAK-STAT signaling and the regulation of IFN responses. Our findings suggest that targeting sirtuin activities may offer an avenue in the development of therapies for managing immune-related diseases and cancer.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/metabolismo , Interferón Tipo I/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Sirtuina 2/metabolismo , Acetilación , Animales , Quinasa 9 Dependiente de la Ciclina/genética , Humanos , Interferón Tipo I/genética , Ratones , Ratones Noqueados , Fosforilación , Factor de Transcripción STAT1/genética , Sirtuina 2/genética , Transcripción Genética , Células U937
6.
J Immunol ; 199(8): 2618-2623, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28904124

RESUMEN

The transitional stage of B cell development is a formative stage in the spleen where autoreactive specificities are censored as B cells gain immune competence, but the intrinsic and extrinsic factors regulating survival of transitional stage 1 (T1) B cells are unknown. We report that B cell expression of IFN-ß is required for optimal survival and TLR7 responses of transitional B cells in the spleen and was overexpressed in T1 B cells from BXD2 lupus-prone mice. Single-cell gene expression analysis of B6 Ifnb+/+ versus B6 Ifnb-/- T1 B cells revealed heterogeneous expression of Ifnb in wild-type B cells and distinct gene expression patterns associated with endogenous IFN-ß. Single-cell analysis of BXD2 T1 B cells revealed that Ifnb is expressed in early T1 B cell development with subsequent upregulation of Tlr7 and Ifna1 Together, these data suggest that T1 B cell expression of IFN-ß plays a key role in regulating responsiveness to external factors.


Asunto(s)
Linfocitos B/inmunología , Interferón beta/metabolismo , Nefritis Lúpica/inmunología , Células Precursoras de Linfocitos B/inmunología , Bazo/inmunología , Animales , Subgrupos de Linfocitos B/inmunología , Diferenciación Celular , Supervivencia Celular , Susceptibilidad a Enfermedades , Interferón beta-1a/genética , Interferón beta-1a/metabolismo , Interferón-alfa , Activación de Linfocitos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos , Análisis de la Célula Individual , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
7.
Nat Rev Immunol ; 8(9): 737-44, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18728636

RESUMEN

Despite accumulating evidence in support of sex-based differences in innate and adaptive immune responses, in the susceptibility to infectious diseases and in the prevalence of autoimmune diseases, health research and clinical practice do not address these distinctions, and most research studies of immune responses do not stratify by sex. X-linked genes, hormones and societal context are among the many factors that contribute to disparate immune responses in males and females. It is crucial to address sex-based differences in disease pathogenesis and in the pharmacokinetics and pharmacodynamics of therapeutic medications to provide optimal disease management for both sexes.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Inmunidad/inmunología , Caracteres Sexuales , Cromosoma X/genética , Cromosoma X/inmunología , Animales , Enfermedad , Hormonas/inmunología , Humanos
8.
J Biol Chem ; 292(11): 4743-4752, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28174303

RESUMEN

The precise signaling mechanisms by which type II IFN receptors control expression of unique genes to induce biological responses remain to be established. We provide evidence that Sin1, a known element of the mammalian target of rapamycin complex 2 (mTORC2), is required for IFNγ-induced phosphorylation and activation of AKT and that such activation mediates downstream regulation of mTORC1 and its effectors. These events play important roles in the assembly of the eukaryotic translation initiation factor 4F (eIF4F) and mRNA translation of IFN-stimulated genes. Interestingly, IFNγ-induced tyrosine phosphorylation of STAT1 is reduced in cells with targeted disruption of Sin1, leading to decreased transcription of several IFNγ-inducible genes in an mTORC2-independent manner. Additionally, our studies establish that Sin1 is essential for generation of type II IFN-dependent antiviral effects and antiproliferative responses in normal and malignant hematopoiesis. Together, our findings establish an important role for Sin1 in both transcription and translation of IFN-stimulated genes and type II IFN-mediated biological responses, involving both mTORC2-dependent and -independent functions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Portadoras/inmunología , Interferón gamma/inmunología , Animales , Línea Celular , Humanos , Inmunidad Innata , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/inmunología , Factor de Transcripción STAT1/química , Factor de Transcripción STAT1/inmunología , Transducción de Señal
9.
Cytokine ; 109: 57-64, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29903574

RESUMEN

Accumulating evidence indicates that chemokine-chemokine receptor interactions invoke biological responses beyond their originally described function of orchestrating leukocyte trafficking. In this review we will extend the findings that chemokines participate actively in the neoplastic process, and consider the contribution of CCL5 activation of CCR5 on breast cancer cells to upregulation of anabolic metabolic events that would support the energy demands of cell replication and proliferation.


Asunto(s)
Neoplasias de la Mama/patología , Carcinogénesis/patología , Quimiocina CCL5/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Receptores CCR5/metabolismo , Linfocitos T Citotóxicos/inmunología , Proliferación Celular/fisiología , Femenino , Humanos , Leucocitos/inmunología , Macrófagos/inmunología , Metástasis de la Neoplasia/patología , Neovascularización Patológica/patología
11.
J Biol Chem ; 291(5): 2389-96, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26645692

RESUMEN

We provide evidence for a unique pathway engaged by the type II IFN receptor, involving mTORC2/AKT-mediated downstream regulation of mTORC1 and effectors. These events are required for formation of the eukaryotic translation initiation factor 4F complex (eIF4F) and initiation of mRNA translation of type II interferon-stimulated genes. Our studies establish that Rictor is essential for the generation of type II IFN-dependent antiviral and antiproliferative responses and that it controls the generation of type II IFN-suppressive effects on normal and malignant hematopoiesis. Together, our findings establish a central role for mTORC2 in IFNγ signaling and type II IFN responses.


Asunto(s)
Proteínas Portadoras/metabolismo , Factor 4F Eucariótico de Iniciación/metabolismo , Interferón gamma/metabolismo , Complejos Multiproteicos/metabolismo , Receptores de Interferón/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Antivirales/química , Antivirales/metabolismo , Quimiocina CXCL10/metabolismo , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Hematopoyesis , Células Madre Hematopoyéticas/citología , Humanos , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Noqueados , Fosforilación , Polirribosomas/metabolismo , Biosíntesis de Proteínas , Proteína Asociada al mTOR Insensible a la Rapamicina , Células U937
12.
Immunology ; 152(3): 439-450, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28646573

RESUMEN

CD11c+ dendritic cells (DCs) exert a critical role as antigen-presenting cells in regulating pathogenic T cells in multiple sclerosis (MS). To determine whether the therapeutic benefit of interferon-ß (IFN-ß) treatment for MS is in part influenced by IFN regulation of DC function, we examined the immunophenotype of DCs derived from IFN-ß+/+ and IFN-ß-/- mice using a myelin oligodendrocyte glycoprotein (MOG) peptide-induced mouse model of MS, experimental autoimmune encephalomyelitis (EAE). Our earlier work identified that IFN-ß-/- mice exhibit earlier onset and more rapid progression of neurological impairment compared with IFN-ß+/+ mice. In this study we show that lipopolysaccharide-/MOG peptide-stimulated IFN-ß-/- DCs secrete cytokines associated with pathological T helper type 17 rather than regulatory T-cell polarization and exhibit increased CD80 and MHCII expression when compared with stimulated IFN-ß+/+ DCs. IFN-ß-/- DCs from mice immunized to develop EAE induce greater proliferation of MOG-transgenic CD4+ T cells and promote interleukin-17 production by these T cells. Adoptive transfer of MOG peptide-primed IFN-ß-/- DCs into IFN-ß+/+ and IFN-ß-/- mice immunized to develop EAE resulted in their rapid migration into the central nervous system of recipient mice, before onset of disease, which we attribute to failed signal transducer and activator of transcription 1-mediated inhibition of CCR7. Taken together, our data support immunoregulatory roles for IFN-ß in the activation and migration of DCs during EAE.


Asunto(s)
Quimiotaxis , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Interferón beta/metabolismo , Traslado Adoptivo , Animales , Antígeno CD11c/inmunología , Antígeno CD11c/metabolismo , Linaje de la Célula , Plasticidad de la Célula , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Predisposición Genética a la Enfermedad , Interferón beta/deficiencia , Interferón beta/genética , Interferón beta/inmunología , Lipopolisacáridos/inmunología , Activación de Linfocitos , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/inmunología , Fenotipo , Receptores CCR7/inmunología , Receptores CCR7/metabolismo , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Células Th17/inmunología , Células Th17/metabolismo
13.
BMC Cancer ; 17(1): 834, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29216863

RESUMEN

BACKGROUND: In earlier studies we have shown that CCL5 activation of CCR5 induces the proliferation and survival of breast cancer cells in a mechanistic target of rapamycin (mTOR)-dependent manner and that this is in part due to CCR5-mediated increases in glycolytic metabolism. METHODS: Using the MDA-MB-231 triple negative human breast cancer cell line and mouse mammary tumor virus - polyomavirus middle T-antigen (MMTV-PyMT) mouse primary breast cancer cells, we conducted in vivo tumor transplant experiments to examine the effects of CCL5-CCR5 interactions in the context of regulating tumor metabolism. Additionally, we employed Matrix-Assisted Laser Desorption/Ionization Fourier Transform Ion Cyclotron Resonance Mass Spectrometry imaging (MALDI-FTICR-MSI) to evaluate tumor utilization of cellular metabolites. RESULTS: We provide evidence that, in the absence of CCR5, the early events associated with rapid tumor growth in the MMTV-PyMT mouse model of spontaneous breast cancer development, are diminished, as demonstrated by a delay in tumor onset. In tumor transplant studies into immunocompromised mice we identify a direct correlation between reduced tumor proliferation and decreased metabolic activity, specifically associated with tumor expression of CCR5. The reduction in tumorigenesis is accompanied by decreases in glucose uptake, glucose transporter-1 (GLUT-1) cell surface expression, intracellular ATP and lactate levels, as well as reduced CCL5 production. Using MALDI-FTICR-MS, we show that the rapid early tumor growth of CCR5+/+ triple negative breast cancer cells in vivo is attributable to increased levels of glycolytic intermediates required for anabolic processes, in contrast to the slower growth rate of their corresponding CCR5-/- cells, that exhibit reduced glycolytic metabolism. CONCLUSIONS: These findings suggest that CCL5-CCR5 interactions in the tumor microenvironment modulate metabolic events during tumor onset to promote tumorigenesis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinogénesis/metabolismo , Quimiocina CCL5/metabolismo , Receptores CCR5/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Glucólisis , Humanos , Ratones , Ratones Endogámicos NOD , Células Tumorales Cultivadas
14.
Proc Natl Acad Sci U S A ; 111(31): 11377-82, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25049393

RESUMEN

We provide evidence that S6 kinase 1 (S6K1) Aly/REF-like target (SKAR) is engaged in IFN-α signaling and plays a key role in the generation of IFN responses. Our data demonstrate that IFN-α induces phosphorylation of SKAR, which is mediated by either the p90 ribosomal protein S6 kinase (RSK) or p70 S6 kinase (S6K1), in a cell type-specific manner. This type I IFN-inducible phosphorylation of SKAR results in enhanced interaction with the eukaryotic initiation factor (eIF)4G and recruitment of activated RSK1 to 5' cap mRNA. Our studies also establish that SKAR is present in cap-binding CBP80 immune complexes and that this interaction is mediated by eIF4G. We demonstrate that inducible protein expression of key IFN-α-regulated protein products such as ISG15 and p21(WAF1/CIP1) requires SKAR activity. Importantly, our studies define a requirement for SKAR in the generation of IFN-α-dependent inhibitory effects on malignant hematopoietic progenitors from patients with chronic myeloid leukemia or myeloproliferative neoplasms. Taken altogether, these findings establish critical and essential roles for SKAR in the regulation of mRNA translation of IFN-sensitive genes and induction of IFN-α biological responses.


Asunto(s)
Interferón-alfa/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citocinas/metabolismo , Guanosina/análogos & derivados , Guanosina/metabolismo , Humanos , Ratones , Complejo Proteico Nuclear de Unión a la Caperuza/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitinas/metabolismo
15.
J Autoimmun ; 69: 38-50, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26948996

RESUMEN

Rheumatoid arthritis (RA) is a systemic autoimmune disease. We previously identified a circulating cell population, fibrocytes, which is activated early in disease. As RA is characterized by the formation of autoantibodies and autoreactive T cells, which often precede symptom onset, the objective of these studies was to characterize fibrocyte activation in the context of T cell activation. Multidimensional flow cytometry was used to characterize the activation status of peripheral blood (PB) fibrocytes and T cells derived from RA patients with different levels of disease activity. Compared to healthy controls, fibrocytes from RA patients exhibited increased activation, denoted as elevated levels of phosphorylation of STAT3 and NF-κB. RA patients had higher numbers of circulating activated Th17 cells and Tregs compared with healthy controls, Th17 cell numbers being higher in patients with moderate to high disease activity. Additionally, increased numbers of FOXP3+ RORγt+ double positive CD4+ T cells were observed in RA patients with more severe disease. Our data confirm that circulating fibrocytes are expanded in RA and that there is a direct correlation between the increase in number of activated fibrocytes and increased number of CD4+ T cells. Moreover, our data suggest that interactions between circulating fibrocytes and activated T cells may promote disease activity. Specifically, we provide in vitro evidence that mouse-derived CD4+ T cells produce GM-CSF which induces fibrocyte proliferation. In turn, activated fibrocytes produce IL-6, promoting Th17 polarization.


Asunto(s)
Artritis Reumatoide/etiología , Artritis Reumatoide/metabolismo , Comunicación Celular , Células del Tejido Conectivo/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Adulto , Animales , Artritis Reumatoide/diagnóstico , Biomarcadores , Estudios de Casos y Controles , Citocinas/metabolismo , Femenino , Humanos , Inmunofenotipificación , Activación de Linfocitos , Recuento de Linfocitos , Masculino , Ratones , Persona de Mediana Edad , Fenotipo
16.
J Biol Chem ; 289(10): 6581-6591, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24469448

RESUMEN

We provide evidence that type I IFN-induced STAT activation is diminished in cells with targeted disruption of the Rictor gene, whose protein product is a key element of mTOR complex 2. Our studies show that transient or stable knockdown of Rictor or Sin1 results in defects in activation of elements of the STAT pathway and reduced STAT-DNA binding complexes. This leads to decreased expression of several IFN-inducible genes that mediate important biological functions. Our studies also demonstrate that Rictor and Sin1 play essential roles in the generation of the suppressive effects of IFNα on malignant erythroid precursors from patients with myeloproliferative neoplasms. Altogether, these findings provide evidence for critical functions for Rictor/Sin1 complexes in type I IFN signaling and the generation of type I IFN antineoplastic responses.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Interferón Tipo I/farmacología , Transcripción Genética/efectos de los fármacos , Animales , Proteínas Portadoras/genética , Células Cultivadas , Fibroblastos/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Fosforilación , Policitemia Vera/metabolismo , Policitemia Vera/patología , Proteína Asociada al mTOR Insensible a la Rapamicina , Transducción de Señal
17.
Trends Immunol ; 33(4): 190-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22321608

RESUMEN

Interferons (IFNs)-α/ß are critical effectors of the innate immune response to virus infections. Through activation of the IFN-α/ß receptor (IFNAR), they induce expression of IFN-stimulated genes (ISGs) that encode antiviral proteins capable of suppressing viral replication and promoting viral clearance. Many highly pathogenic viruses have evolved mechanisms to evade an IFN response and the balance between the robustness of the host immune response and viral antagonistic mechanisms determines whether or not the virus is cleared. Here, we discuss IFNs as broad-spectrum antivirals for treatment of acute virus infections. In particular, they are useful for treatment of re-emerging virus infections, where direct-acting antivirals (DAAs) have limited utility due to DAA-resistant mutations, and for newly emerging virus strains in which the time to vaccine availability precludes vaccination at the onset of an outbreak.


Asunto(s)
Interferón-alfa/inmunología , Interferón beta/inmunología , Virosis/inmunología , Animales , Brotes de Enfermedades , Humanos , Inmunidad Innata , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Transducción de Señal , Virosis/metabolismo
18.
Proc Natl Acad Sci U S A ; 109(20): 7723-8, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22550181

RESUMEN

IFNs transduce signals by binding to cell surface receptors and activating cellular pathways and regulatory networks that control transcription of IFN-stimulated genes (ISGs) and mRNA translation, leading to generation of protein products that mediate biological responses. Previous studies have shown that type I IFN receptor-engaged pathways downstream of AKT and mammalian target of rapamycin complex (mTORC) 1 play important roles in mRNA translation of ISGs and the generation of IFN responses, but the roles of mTORC2 complexes in IFN signaling are unknown. We provide evidence that mTORC2 complexes control IFN-induced phosphorylation of AKT on serine 473 and their function is ultimately required for IFN-dependent gene transcription via interferon-stimulated response elements. We also demonstrate that such complexes exhibit regulatory effects on other IFN-dependent mammalian target of rapamycin-mediated signaling events, likely via engagement of the AKT/mTORC1 axis, including IFN-induced phosphorylation of S6 kinase and its effector rpS6, as well as phosphorylation of the translational repressor 4E-binding protein 1. We also show that induction of ISG protein expression and the generation of antiviral responses are defective in Rictor and mLST8-KO cells. Together, our data provide evidence for unique functions of mTORC2 complexes in the induction of type I IFN responses and suggest a critical role for mTORC2-mediated signals in IFN signaling.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Interferones/metabolismo , Complejos Multiproteicos/metabolismo , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Transactivadores/metabolismo , Animales , Proteínas Portadoras/genética , Células HeLa , Humanos , Immunoblotting , Interferones/inmunología , Luciferasas , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína S6 Ribosómica/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo
19.
J Biol Chem ; 288(33): 23814-22, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23814052

RESUMEN

The mechanisms of generation of the antineoplastic effects of interferons (IFNs) in malignant hematopoietic cells remain to be precisely defined. We examined the activation of type I IFN-dependent signaling pathways in malignant cells transformed by Jak2V617F, a critical pathogenic mutation in myeloproliferative neoplasms (MPNs). Our studies demonstrate that during engagement of the type I IFN receptor (IFNAR), there is activation of Jak-Stat pathways and also engagement of Mnk kinases. Activation of Mnk kinases is regulated by the Mek/Erk pathway and is required for the generation of IFN-induced growth inhibitory responses, but Mnk kinase activation does not modulate IFN-regulated Jak-Stat signals. We demonstrate that for type I IFNs to exert suppressive effects in malignant hematopoietic progenitors from patients with polycythemia vera, induction of Mnk kinase activity is required, as evidenced by studies involving pharmacological inhibition of Mnk or siRNA-mediated Mnk knockdown. Altogether, these findings provide evidence for key and essential roles of the Mnk kinase pathway in the generation of the antineoplastic effects of type I IFNs in Jak2V617F-dependent MPNs.


Asunto(s)
Neoplasias de la Médula Ósea/metabolismo , Interferón-alfa/farmacología , Interferón beta/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Trastornos Mieloproliferativos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Neoplasias de la Médula Ósea/patología , Diferenciación Celular , Línea Celular Transformada , Células Eritroides/efectos de los fármacos , Células Eritroides/metabolismo , Células Eritroides/patología , Factor 4E Eucariótico de Iniciación/metabolismo , Humanos , Quinasas Janus/genética , Quinasas Janus/metabolismo , Ratones , Mutación/genética , Trastornos Mieloproliferativos/patología
20.
Bioorg Med Chem ; 22(3): 978-85, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24433965

RESUMEN

Small molecules that mimic IFN-α epitopes that interact with the cell surface receptor, IFNAR, would be useful therapeutics. One such 8-amino acid region in IFN-α2, designated IRRP-1, was used to derive 11 chemical compounds that belong to 5 distinct chemotypes, containing the molecular features represented by the key residues Leu30, Arg33, and Asp35 in IRRP-1. Three of these compounds exhibited potential mimicry to IRRP-1 and, in cell based assays, as predicted, effectively inhibited IFNAR activation by IFN-α. Of these, compound 3 did not display cell toxicity and reduced IFN-α-inducible STAT1 phosphorylation and STAT-DNA binding. Based on physicochemical properties' analyses, our data suggest that moieties with acidic pKa on the small molecule may be a necessary element for mimicking the carboxyl group of Asp35 in IRRP-1. Our data confirm the relevance of this strategy of molecular mimicry of ligand-receptor interaction domains of protein partners for small molecule drug discovery.


Asunto(s)
Epítopos/química , Receptor de Interferón alfa y beta/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Ácido Aspártico/química , Línea Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Epítopos/metabolismo , Humanos , Interferón-alfa/metabolismo , Modelos Moleculares , Imitación Molecular , Péptidos/química , Fosforilación/efectos de los fármacos , Conformación Proteica , Estructura Terciaria de Proteína , Receptor de Interferón alfa y beta/química , Factor de Transcripción STAT1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA