Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 21(3)2020 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-32019116

RESUMEN

Recent approval of chimeric antigen receptor (CAR) T cell therapy by the European Medicines Agency (EMA)/Federal and Drug Administration (FDA) and the remarkable results of CAR T clinical trials illustrate the curative potential of this therapy. While CARs against a multitude of different antigens are being developed and tested (pre)clinically, there is still a need for optimization. The use of single-chain variable fragments (scFvs) as targeting moieties hampers the quick generation of functional CARs and could potentially limit the efficacy. Instead, nanobodies may largely circumvent these difficulties. We used an available nanobody library generated after immunization of llamas against Cluster of Differentiation (CD) 20 through DNA vaccination or against the ectodomain of CD33 using soluble protein. The nanobody specific sequences were amplified by PCR and cloned by Gibson Assembly into a retroviral vector containing two different second-generation CAR constructs. After transduction in T cells, we observed high cell membrane nanoCAR expression in all cases. Following stimulation of nanoCAR-expressing T cells with antigen-positive cell lines, robust T cell activation, cytokine production and tumor cell lysis both in vitro and in vivo was observed. The use of nanobody technology in combination with PCR and Gibson Assembly allows for the rapid and effective generation of compact CARs.


Asunto(s)
Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T/inmunología , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Dominio Único/inmunología , Línea Celular , Vectores Genéticos , Humanos , Activación de Linfocitos , Reacción en Cadena de la Polimerasa , Receptores de Antígenos de Linfocitos T/genética , Anticuerpos de Cadena Única/genética , Linfocitos T/inmunología
2.
Oncoimmunology ; 7(3): e1398876, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29399401

RESUMEN

Despite approval for the treatment of various malignancies, clinical application of cytokines such as type I interferon (IFN) is severely impeded by their systemic toxicity. AcTakines (Activity-on-Target cytokines) are optimized immunocytokines that, when injected in mice, only reveal their activity upon cell-specific impact. We here show that type I IFN-derived AcTaferon targeted to the tumor displays strong antitumor activity without any associated toxicity, in contrast with wild type IFN. Treatment with CD20-targeted AcTaferon of CD20+ lymphoma tumors or melanoma tumors engineered to be CD20+, drastically reduced tumor growth. This antitumor effect was completely lost in IFNAR- or Batf3-deficient mice, and depended on IFN signaling in conventional dendritic cells. Also the presence of, but not the IFN signaling in, CD8+ T lymphocytes was critical for proficient antitumor effects. When combined with immunogenic chemotherapy, low-dose TNF, or immune checkpoint blockade strategies such as anti-PDL1, anti-CTLA4 or anti-LAG3, complete tumor regressions and subsequent immunity (memory) were observed, still without any concomitant morbidity, again in sharp contrast with wild type IFN. Interestingly, the combination therapy of tumor-targeted AcTaferon with checkpoint inhibiting antibodies indicated its ability to convert nonresponding tumors into responders. Collectively, our findings demonstrate that AcTaferon targeted to tumor-specific surface markers may provide a safe and generic addition to cancer (immuno)therapies.

3.
Cancer Res ; 78(2): 463-474, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29187401

RESUMEN

An ideal generic cancer immunotherapy should mobilize the immune system to destroy tumor cells without harming healthy cells and remain active in case of recurrence. Furthermore, it should preferably not rely on tumor-specific surface markers, as these are only available in a limited set of malignancies. Despite approval for treatment of various cancers, clinical application of cytokines is still impeded by their multiple toxic side effects. Type I IFN has a long history in the treatment of cancer, but its multifaceted activity pattern and complex side effects prevent its clinical use. Here we develop AcTakines (Activity-on-Target cytokines), optimized (mutated) immunocytokines that are up to 1,000-fold more potent on target cells, allowing specific signaling in selected cell types only. Type I IFN-derived AcTaferon (AFN)-targeting Clec9A+ dendritic cells (DC) displayed strong antitumor activity in murine melanoma, breast carcinoma, and lymphoma models and against human lymphoma in humanized mice without any detectable toxic side effects. Combined with immune checkpoint blockade, chemotherapy, or low-dose TNF, complete tumor regression and long-lasting tumor immunity were observed, still without adverse effects. Our findings indicate that DC-targeted AFNs provide a novel class of highly efficient, safe, and broad-spectrum off-the-shelf cancer immunotherapeutics with no need for a tumor marker.Significance: Targeted type I interferon elicits powerful antitumor efficacy, similar to wild-type IFN, but without any toxic side effects. Cancer Res; 78(2); 463-74. ©2017 AACR.


Asunto(s)
Citocinas/química , Células Dendríticas/inmunología , Inmunoterapia , Interferón Tipo I/farmacología , Neoplasias Mamarias Experimentales/terapia , Melanoma Experimental/terapia , Animales , Apoptosis , Proliferación Celular , Terapia Combinada , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/patología , Femenino , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Tumorales Cultivadas
4.
Sci Rep ; 7(1): 15919, 2017 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-29162862

RESUMEN

STAT3 is a pleiotropic transcription factor involved in homeostatic and host defense processes in the human body. It is activated by numerous cytokines and growth factors and generates a series of cellular effects. Of the STAT-mediated signal transduction pathways, STAT3 transcriptional control is best understood. Jak kinase dependent activation of STAT3 relies on Y705 phosphorylation triggering a conformational switch that is stabilized by intermolecular interactions between SH2 domains and the pY705 motif. We here show that a second tyrosine phosphorylation within the SH2 domain at position Y640, induced by Tyk2, negatively controls STAT3 activity. The Y640F mutation leads to stabilization of activated STAT3 homodimers, accelerated nuclear translocation and superior transcriptional activity following IL-6 and LIF stimulation. Moreover, it unlocks type I IFN-dependent STAT3 signalling in cells that are normally refractory to STAT3 transcriptional activation.


Asunto(s)
Fosfotirosina/metabolismo , Factor de Transcripción STAT3/metabolismo , TYK2 Quinasa/metabolismo , Transcripción Genética , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células HEK293 , Humanos , Interferón-alfa/farmacología , Ratones , Proteínas Mutantes/metabolismo , Mutación/genética , Células 3T3 NIH , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Multimerización de Proteína/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Factor de Transcripción STAT3/genética , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , TYK2 Quinasa/química , Transcripción Genética/efectos de los fármacos
5.
Mol Cancer Ther ; 16(12): 2828-2839, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29054987

RESUMEN

Anti-CD20 radioimmunotherapy is an effective approach for therapy of relapsed or refractory CD20pos lymphomas, but faces limitations due to poor tumor penetration and undesirable pharmacokinetics of full antibodies. Camelid single-domain Ab fragments (sdAb) might circumvent some of the limitations of radiolabeled full antibodies. In this study, a set of hCD20-targeting sdAbs was generated, and their capacity to bind hCD20 was evaluated in vitro and in vivo A lead sdAb, sdAb 9079, was selected on the basis of its specific tumor targeting and significant lower kidney accumulation compared with other sdAbs. SdAb 9079 was then radiolabeled with 68Ga and 177Lu for PET imaging and targeted therapy. The therapeutic potential of 177Lu-DTPA-sdAb was compared with that of 177Lu-DTPA-rituximab and unlabeled rituximab in mice bearing hCD20pos tumors. Radiolabeled with 68Ga, sdAb 9079 showed specific tumor uptake, with very low accumulation in nontarget organs, except kidneys. The tumor uptake of 177Lu-DTPA-sdAb 9079 after 1.5 h was 3.4 ± 1.3% ID/g, with T/B and T/M ratios of 13.3 ± 4.6 and 32.9 ± 15.6. Peak tumor accumulation of 177Lu-DTPA-rituximab was about 9 times higher, but concomitantly with high accumulation in nontarget organs and very low T/B and T/M ratios (0.8 ± 0.1 and 7.1 ± 2.4). Treatment of mice with 177Lu-DTPA-sdAb 9079 significantly prolonged median survival compared with control groups and was as effective as treatment with rituximab or its 177Lu-labeled variant. Taken together, sdAb 9079 displays promising features as a theranostic drug to treat CD20pos lymphomas. Mol Cancer Ther; 16(12); 2828-39. ©2017 AACR.


Asunto(s)
Antígenos CD20/metabolismo , Linfoma no Hodgkin/radioterapia , Radioinmunoterapia/métodos , Nanomedicina Teranóstica/métodos , Animales , Modelos Animales de Enfermedad , Humanos , Linfoma no Hodgkin/tratamiento farmacológico , Ratones
6.
Bioinformatics ; 33(9): 1424-1425, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28453684

RESUMEN

Summary: Protein-protein interaction (PPI) studies have dramatically expanded our knowledge about cellular behaviour and development in different conditions. A multitude of high-throughput PPI techniques have been developed to achieve proteome-scale coverage for PPI studies, including the microarray based Mammalian Protein-Protein Interaction Trap (MAPPIT) system. Because such high-throughput techniques typically report thousands of interactions, managing and analysing the large amounts of acquired data is a challenge. We have therefore built the MAPPIT cell microArray Protein Protein Interaction-Data management & Analysis Tool (MAPPI-DAT) as an automated data management and analysis tool for MAPPIT cell microarray experiments. MAPPI-DAT stores the experimental data and metadata in a systematic and structured way, automates data analysis and interpretation, and enables the meta-analysis of MAPPIT cell microarray data across all stored experiments. Availability and Implementation: MAPPI-DAT is developed in Python, using R for data analysis and MySQL as data management system. MAPPI-DAT is cross-platform and can be ran on Microsoft Windows, Linux and OS X/macOS. The source code and a Microsoft Windows executable are freely available under the permissive Apache2 open source license at https://github.com/compomics/MAPPI-DAT. Contact: jan.tavernier@vib-ugent.be or lennart.martens@vib-ugent.be. Supplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Análisis por Matrices de Proteínas/métodos , Mapeo de Interacción de Proteínas/métodos , Programas Informáticos , Animales , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Mamíferos/metabolismo
7.
Mol Cell Proteomics ; 15(12): 3624-3639, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27803151

RESUMEN

Because proteins are the main mediators of most cellular processes they are also prime therapeutic targets. Identifying physical links among proteins and between drugs and their protein targets is essential in order to understand the mechanisms through which both proteins themselves and the molecules they are targeted with act. Thus, there is a strong need for sensitive methods that enable mapping out these biomolecular interactions. Here we present a robust and sensitive approach to screen proteome-scale collections of proteins for binding to proteins or small molecules using the well validated MAPPIT (Mammalian Protein-Protein Interaction Trap) and MASPIT (Mammalian Small Molecule-Protein Interaction Trap) assays. Using high-density reverse transfected cell microarrays, a close to proteome-wide collection of human ORF clones can be screened for interactors at high throughput. The versatility of the platform is demonstrated through several examples. With MAPPIT, we screened a 15k ORF library for binding partners of RNF41, an E3 ubiquitin protein ligase implicated in receptor sorting, identifying known and novel interacting proteins. The potential related to the fact that MAPPIT operates in living human cells is illustrated in a screen where the protein collection is scanned for interactions with the glucocorticoid receptor (GR) in its unliganded versus dexamethasone-induced activated state. Several proteins were identified the interaction of which is modulated upon ligand binding to the GR, including a number of previously reported GR interactors. Finally, the screening technology also enables detecting small molecule target proteins, which in many drug discovery programs represents an important hurdle. We show the efficiency of MASPIT-based target profiling through screening with tamoxifen, a first-line breast cancer drug, and reversine, an investigational drug with interesting dedifferentiation and antitumor activity. In both cases, cell microarray screens yielded known and new potential drug targets highlighting the utility of the technology beyond fundamental biology.


Asunto(s)
Mapeo de Interacción de Proteínas/métodos , Proteoma/metabolismo , Análisis de Matrices Tisulares/métodos , Células HEK293 , Humanos , Bibliotecas de Moléculas Pequeñas/metabolismo , Tamoxifeno/metabolismo
8.
Cell Mol Life Sci ; 72(3): 629-644, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25098352

RESUMEN

The adipocyte-derived cytokine leptin acts as a metabolic switch, connecting the body's metabolism to high-energy consuming processes such as reproduction and immune responses. We here provide genetic and biochemical evidence that the metabolic and immune functions of leptin can be uncoupled at the receptor level. First, homozygous mutant fatt/fatt mice carry a spontaneous splice mutation causing deletion of the leptin receptor (LR) immunoglobulin-like domain (IGD) in all LR isoforms. These mice are hyperphagic and morbidly obese, but display only minimal changes in size and cellularity of the thymus, and cellular immune responses are unaffected. These animals also displayed liver damage in response to concavalin A comparable to wild-type and heterozygous littermates. Second, treatment of healthy mice with a neutralizing nanobody targeting IGD induced weight gain and hyperinsulinaemia, but completely failed to block development of experimentally induced autoimmune diseases. These data indicate that leptin receptor deficiency or antagonism profoundly affects metabolism, with little concomitant effects on immune functions.


Asunto(s)
Leptina/inmunología , Leptina/metabolismo , Receptores de Leptina/metabolismo , Análisis de Varianza , Animales , Artritis Experimental/patología , Secuencia de Bases , Western Blotting , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Cartilla de ADN/genética , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/patología , Citometría de Flujo , Células HEK293 , Humanos , Células MCF-7 , Masculino , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Glicoproteína Mielina-Oligodendrócito/toxicidad , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Receptores de Leptina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Eliminación de Secuencia/genética
9.
Nat Commun ; 5: 3016, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24398568

RESUMEN

Systemic toxicity currently prevents exploiting the huge potential of many cytokines for medical applications. Here we present a novel strategy to engineer immunocytokines with very high targeting efficacies. The method lies in the use of mutants of toxic cytokines that markedly reduce their receptor-binding affinities, and that are thus rendered essentially inactive. Upon fusion to nanobodies specifically binding to marker proteins, activity of these cytokines is selectively restored for cell populations expressing this marker. This 'activity-by-targeting' concept was validated for type I interferons and leptin. In the case of interferon, activity can be directed to target cells in vitro and to selected cell populations in mice, with up to 1,000-fold increased specific activity. This targeting strategy holds promise to revitalize the clinical potential of many cytokines.


Asunto(s)
Citocinas/metabolismo , Sistemas de Liberación de Medicamentos , Leptina/metabolismo , Receptores de Citocinas/metabolismo , Anticuerpos de Dominio Único/metabolismo , Animales , Humanos , Interferón Tipo I/metabolismo , Interferón-alfa/metabolismo , Interleucina-15/metabolismo , Interleucina-2/metabolismo , Ratones , Unión Proteica , Receptor de Interferón alfa y beta/metabolismo , Receptores de Leptina , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo
10.
Methods Mol Biol ; 812: 283-94, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22218867

RESUMEN

Mammalian protein-protein interaction trap (MAPPIT) is a two-hybrid technology to identify and characterize interactions of proteins with other proteins or organic molecules in living mammalian (human) cells. The method relies on complementation of a modified cytokine receptor complex. Protein interaction restores the signalling competence of the complex, which is monitored through the activation of a reporter gene. Here, we describe a protocol that has been recently developed to increase the utility of MAPPIT as a tool to identify novel interactions. In the ArrayMAPPIT assay, a collection of prey proteins which is arrayed in high-density microtiter plates is efficiently screened for interaction partners using reverse transfection into a bait-expressing cell pool.


Asunto(s)
Mapeo de Interacción de Proteínas/instrumentación , Proteínas/análisis , Proteínas/metabolismo , Animales , Humanos , Plásmidos/genética , Mapeo de Interacción de Proteínas/métodos , Factores de Tiempo , Transfección
11.
Methods Mol Biol ; 812: 295-307, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22218868

RESUMEN

The discovery of novel antivirals for HIV and HCV has been a focus of intensive research for many years. Where the inhibition of critical viral enzymes by small molecules has proven effective for many viruses, there is considerable merit in pursuing protein-protein interactions (PPIs) as targets for therapeutic intervention. The mammalian protein-protein interaction trap (MAPPIT) is a two-hybrid system used for the study of PPIs. The bait and prey proteins are linked to deficient cytokine receptor chimeras, where the bait and prey interaction and subsequent ligand stimulation restores JAK-STAT signaling, resulting in reporter gene expression controlled by a STAT3-responsive promoter. We report the use of MAPPIT as a high-throughput screening assay for the discovery of inhibitors or stimulators of the Vif-APOBEC3G interaction and the reverse transcriptase heterodimerization (RTp66-RTp51) for HIV and the NS4A-NS3 interaction for HCV.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , VIH/metabolismo , Hepacivirus/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Mapas de Interacción de Proteínas/efectos de los fármacos , Células HEK293 , Humanos , Unión Proteica/efectos de los fármacos , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Reproducibilidad de los Resultados , Proteínas Virales/química , Proteínas Virales/metabolismo
12.
Methods Mol Biol ; 820: 231-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22131035

RESUMEN

We have designed a sensitive and versatile bioassay for quantification of series of cytokines. The assay makes use of chimeric receptors composed of the extracellular, ligand-binding part of the cognate cytokine receptor and the transmembrane and cytosolic part of the type I interferon receptor. Receptors can be homo- (e.g. erythropoietin), di- (e.g. interleukin-5), or even trimeric (e.g. interleukin-2). Stable expression of these chimeras in the 2fTGH cell line allows an interferon-type signaling, which makes a positive selection in conditioned medium possible or a negative selection using a toxic guanine analog. The cytokine of interest is quantified by the extent of cell survival or cell toxicity respectively, which can be measured by easy and cheap crystal violet staining. This bioassay is sensitive in the lower picogram per milliliter range and, in contrast to ELISA methods, only measures the concentration of biologically active cytokines. Using this approach, hypersensitive 2fTGH cell lines have been developed for type I and II interferons, erythropoietin, interleukin-2, and interleukin-5.


Asunto(s)
Bioensayo/métodos , Interferón Tipo I/aislamiento & purificación , Transducción de Señal , Secuencia de Bases , Línea Celular , Eritropoyetina/análisis , Eritropoyetina/metabolismo , Humanos , Interleucina-2/análisis , Interleucina-2/metabolismo , Interleucina-5/análisis , Interleucina-5/metabolismo , Datos de Secuencia Molecular , Receptor de Interferón alfa y beta/metabolismo , Sensibilidad y Especificidad
13.
Nucleic Acids Res ; 38(6): 1902-12, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20015971

RESUMEN

The host restriction factor Apobec3G is a cytidine deaminase that incorporates into HIV-1 virions and interferes with viral replication. The HIV-1 accessory protein Vif subverts Apobec3G by targeting it for proteasomal degradation. We propose a model in which Apobec3G N-terminal domains symmetrically interact via a head-to-head interface containing residues 122 RLYYFW 127. To validate this model and to characterize the Apobec3G-Apobec3G and the Apobec3G-Vif interactions, the mammalian protein-protein interaction trap two-hybrid technique was used. Mutations in the head-to-head interface abrogate the Apobec3G-Apobec3G interaction. All mutations that inhibit Apobec3G-Apobec3G binding also inhibit the Apobec3G-Vif interaction, indicating that the head-to head interface plays an important role in the interaction with Vif. Only the D128K, P129A and T32Q mutations specifically affect the Apobec3G-Vif association. In our model, D128, P129 and T32 cluster at the edge of the head-to-head interface, possibly forming a Vif binding site composed of two Apobec3G molecules. We propose that Vif either binds at the Apobec3G head-to-head interface or associates with an RNA-stabilized Apobec3G oligomer.


Asunto(s)
Citidina Desaminasa/química , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/química , Desaminasa APOBEC-3G , Sitios de Unión , Línea Celular , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Citosina Desaminasa/química , Dimerización , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Dominios y Motivos de Interacción de Proteínas , Homología Estructural de Proteína , Técnicas del Sistema de Dos Híbridos , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo
14.
J Proteome Res ; 8(2): 877-86, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19159283

RESUMEN

Physical interactions between proteins play a key role in probably every cellular process. Efforts to chart the protein interaction networks are ongoing in a number of model organisms using a diversity of approaches. The resulting genome-wide interaction maps will provide a scaffold for further detailed functional analysis. We developed MAPPIT, a mammalian two-hybrid approach that allows identification and analysis of mammalian protein-protein interactions in their native environment. Here, we introduce an efficient MAPPIT assay that permits high-throughput screening of arrayed collections of proteins and complements a previously published cDNA library screening approach. We validated both methods in screens for interaction partners of the Cullin-based E3 ubiquitin ligase subunits SKP1 and Elongin C. In addition to a number of known interactors, novel SKP1 and Elongin C binding proteins were identified. The array assay is an important addition to the MAPPIT suite of technologies that is expected to significantly increase its utility as a toolbox to screen for novel interactors of proteins or small molecules.


Asunto(s)
Análisis por Micromatrices/métodos , Mapeo de Interacción de Proteínas/métodos , Proteínas/análisis , Técnicas del Sistema de Dos Híbridos , Animales , Línea Celular , Biblioteca de Genes , Genes Reporteros , Humanos , Mapeo de Interacción de Proteínas/instrumentación , Reproducibilidad de los Resultados
15.
J Virol Methods ; 153(1): 7-15, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18640157

RESUMEN

The high mutation rate of Human Immunodeficiency Virus (HIV) leads to the rapid derivation of compound-resistant virus strains and thus necessitates the identification and development of compounds with alternative mode of actions. MAPPIT (MAmmalian Protein-Protein Interaction Trap) is a highly efficient tool to study protein-protein interactions in intact human cells and is applied to study the dimerization process of the HIV reverse transcriptase complex. Highly specific signals for the p66/p51 and p66/p66 interactions could readily be detected. Specificity was established further by introducing mutations in either subunit. Treatment with efavirenz resulted in an increased MAPPIT signal, with an EC50 value of 64nM for the p66/p51 interaction, and allowed detection of the p51/p51 homodimerization, confirming the context-dependent asymmetric contribution of both subunits. These results show that MAPPIT can be used as a novel screening tool for anti-HIV compounds in intact human cells.


Asunto(s)
Transcriptasa Inversa del VIH/metabolismo , VIH/fisiología , Mapeo de Interacción de Proteínas/métodos , Alquinos , Benzoxazinas/farmacología , Línea Celular , Ciclopropanos , Dimerización , Transcriptasa Inversa del VIH/genética , Humanos , Concentración 50 Inhibidora , Unión Proteica , Subunidades de Proteína/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología
16.
Biochem J ; 407(1): 141-51, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17627610

RESUMEN

Type I IFNs (interferons) (IFNalpha/beta) form a family of related cytokines that control a variety of cellular functions through binding to a receptor composed of IFNAR (IFNalpha receptor subunit) 1 and 2. Among type I IFNs, the alpha2 and beta subtypes exhibit a large difference in their binding affinities to IFNAR1, and it was suggested that high concentrations of IFNAR1 may compensate for its low intrinsic binding affinity for IFNalpha2. We tested whether receptor-proximal signalling events are sensitive to IFNAR1 surface concentration by investigating the relationship between relative IFNAR1/IFNAR2 surface levels and IFNalpha2 and IFNbeta signalling potencies in several cell lines. For this, we monitored the activation profile of JAK (Janus kinase)/STAT (signal transducer and activator of transcription) proteins, measured basal and ligand-induced surface decay of each receptor subunit and tested the effect of variable IFNAR1 levels on IFNalpha2 signalling potency. Our data show that the cell-surface IFNAR1 level is indeed a limiting factor for assembly of the functional complex, but an increased concentration of it does not translate into an IFNalpha/beta differential JAK/STAT signalling nor does it change the dynamics of the engaged receptor. Importantly, however, our data highlight a differential effect upon routing of IFNAR2. Following binding of IFNalpha2, IFNAR2 is internalized, but, instead of being routed towards degradation as it is when complexed to IFNbeta, it recycles back to the cell surface. These observations suggest strongly that the stability and the intracellular lifetime of the ternary complex account for the differential control of IFNAR2. Moreover, the present study opens up the attractive possibility that endosomal-initiated signalling may contribute to IFNalpha/beta differential bioactivities.


Asunto(s)
Regulación hacia Abajo , Interferón-alfa/farmacología , Interferón beta/farmacología , Quinasas Janus/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Factores de Transcripción STAT/metabolismo , Células Cultivadas , Células HeLa , Humanos , Células Jurkat , Ligandos , Receptor de Interferón alfa y beta/genética , Transducción de Señal , Factores de Tiempo , Transfección
17.
Mol Cell Biol ; 26(5): 1888-97, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16479007

RESUMEN

Alpha and beta interferons (IFN-alpha and IFN-beta) are multifunctional cytokines that exhibit differential activities through a common receptor composed of the subunits IFNAR1 and IFNAR2. Here we combined biophysical and functional studies to explore the mechanism that allows the alpha and beta IFNs to act differentially. For this purpose, we have engineered an IFN-alpha2 triple mutant termed the HEQ mutant that mimics the biological properties of IFN-beta. Compared to wild-type (wt) IFN-alpha2, the HEQ mutant confers a 30-fold higher binding affinity towards IFNAR1, comparable to that measured for IFN-beta, resulting in a much higher stability of the ternary complex as measured on model membranes. The HEQ mutant, like IFN-beta, promotes a differentially higher antiproliferative effect than antiviral activity. Both bring on a down-regulation of the IFNAR2 receptor upon induction, confirming an increased ternary complex stability of the plasma membrane. Oligonucleotide microarray experiments showed similar gene transcription profiles induced by the HEQ mutant and IFN-beta and higher levels of gene induction or repression than those for wt IFN-alpha2. Thus, we show that the differential activities of IFN-beta are directly related to the binding affinity for IFNAR1. Conservation of the residues mutated in the HEQ mutant within IFN-alpha subtypes suggests that IFN-alpha has evolved to bind IFNAR1 weakly, apparently to sustain differential levels of biological activities compared to those induced by IFN-beta.


Asunto(s)
Interferón-alfa/genética , Interferón-alfa/farmacología , Interferón beta/metabolismo , Interferón beta/farmacología , Proteínas de la Membrana/metabolismo , Receptores de Interferón/metabolismo , Antivirales/farmacología , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo , Regulación de la Expresión Génica , Humanos , Interferón-alfa/metabolismo , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Complejos Multiproteicos , Mutación , Regiones Promotoras Genéticas , Receptor de Interferón alfa y beta , Receptores de Interferón/efectos de los fármacos , Receptores de Interferón/genética , Activación Transcripcional
18.
J Mol Biol ; 350(3): 476-88, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15946680

RESUMEN

Ligand-induced cross-linking of the type I interferon (IFN) receptor subunits ifnar1 and ifnar2 induces a pleiotrophic cellular response. Several studies have suggested differential signal activation by flexible recruitment of the accessory receptor subunit ifnar1. We have characterized the roles of the four Ig-like sub-domains (SDs) of the extracellular domain of ifnar1 (ifnar1-EC) for ligand recognition and receptor assembling. Various sub-fragments of ifnar1-EC were expressed in insect cells and purified to homogeneity. Solid phase binding assays with the ligands IFN(alpha)2 and IFN(beta) revealed that all three N-terminal SDs were required and sufficient for ligand binding, and that IFN(alpha)2 and IFN(beta) compete for this binding site. Cellular binding assays with different fragments, however, highlighted the key role of the membrane-proximal SD for the formation of an in situ IFN-receptor complex. Even substitution with the corresponding SD from homologous cytokine receptors did not restore high-affinity ligand binding. Receptor assembling analysis on supported lipid bilayers in vitro revealed that the membrane-proximal SD controls appropriate orientation of the receptor on the membrane, which is required for efficient association of ifnar1 into the ternary complex.


Asunto(s)
Membrana Dobles de Lípidos/química , Proteínas de la Membrana/química , Receptores de Interferón/química , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Unión Competitiva , Línea Celular , Membrana Celular/metabolismo , Separación Celular , Cromatografía , Electroforesis en Gel de Poliacrilamida , Escherichia coli/metabolismo , Citometría de Flujo , Glicosilación , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Cinética , Ligandos , Membrana Dobles de Lípidos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Receptor de Interferón alfa y beta , Homología de Secuencia de Aminoácido , Transducción de Señal , Transfección
19.
Methods Mol Biol ; 263: 293-310, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14976373

RESUMEN

The mammalian protein-protein interaction trap (MAPPIT) is a two-hybrid assay based on insights in type I cytokine signal transduction. Bait and prey polypeptides are tethered to mutant cytokine receptor chimeras which are impaired in signaling. On bait-prey interaction and after ligand stimulation, the JAK-STAT signaling cascade is initiated leading to transcription of a reporter or marker gene under the control of the STAT3-responsive rPAP1 promoter. In addition to a physiologically relevant context for mammalian protein-protein interactions this method provides separation of interactor and effector zones, and can be applied for both analytical and screening purposes. In the protocol described here, a cytokine receptor derived surface tag is used as a selectable marker. After an initial presort step using magnetic-activated cell sorting (MACS), "positive" cells are selected by fluorescence-activated cell sorting (FACS).


Asunto(s)
Separación Celular/métodos , Citometría de Flujo/métodos , Animales , Línea Celular , ADN Complementario/metabolismo , Genes Reporteros , Vectores Genéticos , Humanos , Ligandos , Magnetismo , Modelos Biológicos , Péptidos/química , Plásmidos/metabolismo , Unión Proteica , Retroviridae/genética , Transducción de Señal , Técnicas del Sistema de Dos Híbridos
20.
Mol Endocrinol ; 18(1): 150-61, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14525952

RESUMEN

The leptin receptor (LR), a member of the class I cytokine receptor family, is composed of a single subunit. Its extracellular domain consists of two so-called cytokine receptor homology domains, separated by an Ig-like domain, and two additional fibronectin type III modules. Requirements for LR activation were examined using a complementation strategy. Two LR mutants, LR-FFY-Deltabox 1 and LR-F3, deficient in Janus kinase or signal transducer and activator of transcription (STAT) activation, respectively, were only able to generate a STAT3-dependent signal when coexpressed. Based on the requirements for Janus kinase/STAT signaling, and on the lack of complementation with similar receptor constructs, but containing the extracellular domain of the homodimeric erythropoietin receptor, this observation can be explained only by higher order LR clustering. Using a panel of deletion mutants we were able to define a role for the cytokine receptor homology 1 and Ig-like domains in leptin signaling. Moreover, we demonstrate a nonredundant function for the individual receptor chains within the homomeric LR complex. Based on these data, we propose a possible model for LR clustering.


Asunto(s)
Regulación de la Expresión Génica/genética , Receptores de Superficie Celular/genética , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Eritropoyetina/farmacología , Humanos , Leptina/farmacología , Ratones , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos , Receptores de Superficie Celular/metabolismo , Receptores de Leptina , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...