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.
Mol Cell Proteomics ; 23(3): 100741, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38387774

RESUMEN

Exogenous glucocorticoids are frequently used to treat inflammatory disorders and as adjuncts for the treatment of solid cancers. However, their use is associated with severe side effects and therapy resistance. Novel glucocorticoid receptor (GR) ligands with a patient-validated reduced side effect profile have not yet reached the clinic. GR is a member of the nuclear receptor family of transcription factors and heavily relies on interactions with coregulator proteins for its transcriptional activity. To elucidate the role of the GR interactome in the differential transcriptional activity of GR following treatment with the selective GR agonist and modulator dagrocorat compared to classic (ant)agonists, we generated comprehensive interactome maps by high-confidence proximity proteomics in lung epithelial carcinoma cells. We found that dagrocorat and the antagonist RU486 both reduced GR interaction with CREB-binding protein/p300 and the mediator complex compared to the full GR agonist dexamethasone. Chromatin immunoprecipitation assays revealed that these changes in GR interactome were accompanied by reduced GR chromatin occupancy with dagrocorat and RU486. Our data offer new insights into the role of differential coregulator recruitment in shaping ligand-specific GR-mediated transcriptional responses.


Asunto(s)
Benzamidas , Cromatina , Fenantrenos , Receptores de Glucocorticoides , Humanos , Receptores de Glucocorticoides/genética , Mifepristona/farmacología , Complejo Mediador/metabolismo , Glucocorticoides/farmacología , Glucocorticoides/metabolismo , Dexametasona/farmacología
2.
Mol Cancer ; 22(1): 191, 2023 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-38031106

RESUMEN

Despite major improvements in immunotherapeutic strategies, the immunosuppressive tumor microenvironment remains a major obstacle for the induction of efficient antitumor responses. In this study, we show that local delivery of a bispecific Clec9A-PD-L1 targeted type I interferon (AcTaferon, AFN) overcomes this hurdle by reshaping the tumor immune landscape.Treatment with the bispecific AFN resulted in the presence of pro-immunogenic tumor-associated macrophages and neutrophils, increased motility and maturation profile of cDC1 and presence of inflammatory cDC2. Moreover, we report empowered diversity in the CD8+ T cell repertoire and induction of a shift from naive, dysfunctional CD8+ T cells towards effector, plastic cytotoxic T lymphocytes together with increased presence of NK and NKT cells as well as decreased regulatory T cell levels. These dynamic changes were associated with potent antitumor activity. Tumor clearance and immunological memory, therapeutic immunity on large established tumors and blunted tumor growth at distant sites were obtained upon co-administration of a non-curative dose of chemotherapy.Overall, this study illuminates further application of type I interferon as a safe and efficient way to reshape the suppressive tumor microenvironment and induce potent antitumor immunity; features which are of major importance in overcoming the development of metastases and tumor cell resistance to immune attack. The strategy described here has potential for application across to a broad range of cancer types.


Asunto(s)
Interferón Tipo I , Neoplasias , Humanos , Linfocitos T CD8-positivos , Interferón Tipo I/metabolismo , Microambiente Tumoral , Antígeno B7-H1/metabolismo , Neoplasias/metabolismo , Inmunoterapia , Línea Celular Tumoral
3.
Sci Rep ; 11(1): 21575, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732771

RESUMEN

Type I Interferon (IFN) was the very first drug approved for the treatment of Multiple Sclerosis (MS), and is still frequently used as a first line therapy. However, systemic IFN also causes considerable side effects, affecting therapy adherence and dose escalation. In addition, the mechanism of action of IFN in MS is multifactorial and still not completely understood. Using AcTaferons (Activity-on-Target IFNs, AFNs), optimized IFN-based immunocytokines that allow cell-specific targeting, we have previously demonstrated that specific targeting of IFN activity to dendritic cells (DCs) can protect against experimental autoimmune encephalitis (EAE), inducing in vivo tolerogenic protective effects, evidenced by increased indoleamine-2,3-dioxygenase (IDO) and transforming growth factor ß (TGFß) release by plasmacytoid (p) DCs and improved immunosuppressive capacity of regulatory T and B cells. We here report that targeting type I IFN activity specifically towards B cells also provides strong protection against EAE, and that targeting pDCs using SiglecH-AFN can significantly add to this protective effect. The superior protection achieved by simultaneous targeting of both B lymphocytes and pDCs correlated with improved IL-10 responses in B cells and conventional cDCs, and with a previously unseen very robust IDO response in several cells, including all B and T lymphocytes, cDC1 and cDC2.


Asunto(s)
Linfocitos B/metabolismo , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/terapia , Interferones/metabolismo , Animales , Anticuerpos/química , Biotecnología , Progresión de la Enfermedad , Inmunosupresores , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón Tipo I/metabolismo , Recuento de Linfocitos , Linfocitos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Péptidos/química , Transducción de Señal , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/metabolismo
4.
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
5.
EMBO Mol Med ; 12(2): e11223, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31912630

RESUMEN

Systemic toxicities have severely limited the clinical application of tumor necrosis factor (TNF) as an anticancer agent. Activity-on-Target cytokines (AcTakines) are a novel class of immunocytokines with improved therapeutic index. A TNF-based AcTakine targeted to CD13 enables selective activation of the tumor neovasculature without any detectable toxicity in vivo. Upregulation of adhesion markers supports enhanced T-cell infiltration leading to control or elimination of solid tumors by, respectively, CAR T cells or a combination therapy with CD8-targeted type I interferon AcTakine. Co-treatment with a CD13-targeted type II interferon AcTakine leads to very rapid destruction of the tumor neovasculature and complete regression of large, established tumors. As no tumor markers are needed, safe and efficacious elimination of a broad range of tumor types becomes feasible.


Asunto(s)
Inmunoterapia , Neoplasias , Factor de Necrosis Tumoral alfa , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/terapia
6.
J Autoimmun ; 97: 70-76, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30467068

RESUMEN

Type I Interferon (IFN) is widely used for multiple sclerosis (MS) treatment, but its side effects are limiting and its mechanism of action still unknown. Furthermore, 30-50% of MS patients are unresponsive, and IFN can even induce relapses. Fundamental understanding of the cellular target(s) of IFN will help to optimize treatments by reducing side effects and separating beneficial from detrimental effects. To improve clinical systemic IFN usage, we are developing AcTaferons (Activity-on-Target IFNs = AFNs), optimized IFN-based immunocytokines that allow cell-specific targeting. In experimental autoimmune encephalitis (EAE) in mice, high dose WT mIFNα could delay disease, but caused mortality and severe hematological deficits. In contrast, AFN targeted to dendritic cells (DC, via Clec9A) protected without mortality or hematological consequences. Conversely, CD8-targeted AFN did not protect and exacerbated weight loss, indicating the presence of both protective and unfavorable IFN effects in EAE. Comparing Clec9A-, XCR1-and SiglecH-targeting, we found that targeting AFN to plasmacytoid (p) and conventional (c) DC is superior and non-toxic compared to WT mIFN. DC-targeted AFN increased pDC numbers and their tolerogenic potential, evidenced by increased TGFß and IDO synthesis and regulatory T cell induction. In addition, both regulatory T and B cells produced significantly more immunosuppressive TGFß and IL-10. In conclusion, specific DC-targeting of IFN activity induces a robust in vivo tolerization, efficiently protecting against EAE, without noticeable side effects. Thus, dissecting positive and negative IFN effects via cell-specific targeting may result in better and safer MS therapy and response rates.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/metabolismo , Tolerancia Inmunológica , Interferones/metabolismo , Animales , Antígeno B7-H1/metabolismo , Biomarcadores , Antígeno CTLA-4/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Encefalomielitis Autoinmune Experimental/patología , Masculino , Ratones , Modelos Biológicos
7.
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.

8.
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
9.
Nat Protoc ; 12(5): 881-898, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28358392

RESUMEN

The analysis of protein interaction networks is one of the key challenges in the study of biology. It connects genotypes to phenotypes, and disruption of such networks is associated with many pathologies. Virtually all the approaches to the study of protein complexes require cell lysis, a dramatic step that obliterates cellular integrity and profoundly affects protein interactions. This protocol starts with Virotrap, a novel approach that avoids the need for cell homogenization by fusing the protein of interest to the HIV-1 Gag protein, trapping protein complexes in virus-like particles. By using the straightforward filtering index (SFINX), which is a powerful and intuitive online tool (http://sfinx.ugent.be) that enables contaminant removal from candidate lists resulting from mass-spectrometry-based analysis, we provide a complete workflow for researchers interested in mammalian protein complexes. Given direct access to mass spectrometers, researchers can process up to 24 samples in 7 d.


Asunto(s)
Mapas de Interacción de Proteínas , Proteínas/aislamiento & purificación , Proteínas/metabolismo , Proteómica/métodos , Animales , Humanos , Mamíferos , Espectrometría de Masas/métodos , Unión Proteica , Multimerización de Proteína
10.
Nat Commun ; 7: 11416, 2016 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-27122307

RESUMEN

Cell lysis is an inevitable step in classical mass spectrometry-based strategies to analyse protein complexes. Complementary lysis conditions, in situ cross-linking strategies and proximal labelling techniques are currently used to reduce lysis effects on the protein complex. We have developed Virotrap, a viral particle sorting approach that obviates the need for cell homogenization and preserves the protein complexes during purification. By fusing a bait protein to the HIV-1 GAG protein, we show that interaction partners become trapped within virus-like particles (VLPs) that bud from mammalian cells. Using an efficient VLP enrichment protocol, Virotrap allows the detection of known binary interactions and MS-based identification of novel protein partners as well. In addition, we show the identification of stimulus-dependent interactions and demonstrate trapping of protein partners for small molecules. Virotrap constitutes an elegant complementary approach to the arsenal of methods to study protein complexes.


Asunto(s)
Infecciones por VIH/metabolismo , VIH-1/metabolismo , Mapeo de Interacción de Proteínas/métodos , Proteínas/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/genética , Humanos , Unión Proteica , Proteínas/genética , Virión/genética , Virión/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética
11.
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
12.
Biochem J ; 441(1): 425-34, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21851341

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. Accumulating evidence suggests that leptin plays a role in human pathologies, such as autoimmune diseases and cancer, thus providing a rationale for the development of leptin antagonists. In the present study, we generated and evaluated a panel of neutralizing nanobodies targeting the LR (leptin receptor). A nanobody comprises the variable domain of the naturally occurring single-chain antibodies found in members of the Camelidae family. We identified three classes of neutralizing nanobodies targeting different LR subdomains: i.e. the CRH2 (cytokine receptor homology 2), Ig-like and FNIII (fibronectin type III) domains. Only nanobodies directed against the CRH2 domain inhibited leptin binding. We could show that a nanobody that targets the Ig-like domain potently interfered with leptin-dependent regulation of hypothalamic NPY (neuropeptide Y) expression. As a consequence, daily intraperitoneal injection increased body weight, body fat content, food intake, liver size and serum insulin levels. All of these characteristics resemble the phenotype of leptin and LR-deficient animals. The results of the present study support proposed models of the activated LR complex, and demonstrate that it is possible to block LR signalling without affecting ligand binding. These nanobodies form new tools to study the mechanisms of BBB (blood-brain barrier) leptin transport and the effect of LR inhibition in disease models.


Asunto(s)
Anticuerpos/farmacología , Leptina/antagonistas & inhibidores , Nanoestructuras/química , Receptores de Leptina/antagonistas & inhibidores , Tejido Adiposo , Animales , Anticuerpos/química , Camélidos del Nuevo Mundo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Hiperinsulinismo , Hígado/anatomía & histología , Ratones , Ratones Endogámicos C57BL , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Tamaño de los Órganos , Unión Proteica , Estructura Terciaria de Proteína , Transducción de Señal , Aumento de Peso
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 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
15.
PLoS One ; 3(6): e2427, 2008 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-18560560

RESUMEN

BACKGROUND: ISG15 is an Ubiquitin-like protein, highly induced by Type I Interferons. Upon the cooperative activity of specific Ubiquitinating enzymes, ISG15 can be conjugated to its substrates. Increasing evidence points to a role for protein ISGylation in anti-viral and anti-tumoral defense. PRINCIPAL FINDINGS: We identified ISG15 from Old World Monkeys (OWm) as a hyper-efficient protein modifier. Western blot analysis visualized more efficient conjugation of OWmISG15 relative to HuISG15 in human (Hu), monkey and mouse (Mo) cell-lines. Moreover, the substrates of OWmISG15 identified upon Tandem Affinity Purification followed by LC-MS/MS identification largely outnumbered these of HuISG15 itself. Several Ubiquitin-Conjugating enzymes were identified as novel ISGylated substrates. Introduction of a N89D mutation in HuISG15 improved its ISGylation capacity, and additional Q31K/T33A/D133N mutations yielded a HuISG15 variant with an ISGylation efficiency comparable to OWmISG15. Homology modeling and structural superposition situate N89 in the interaction interface with the Activating enzyme. Analysis of the UbE1L residues in this interface revealed a striking homology between OWmUbE1L and HuUbE1, the Activating enzyme of Ubiquitin. In line with this observation, we found efficient activation of AgmISG15, but not HuISG15 or MoISG15, by HuUbE1, thus providing a likely explanation for OWm hyperISGylation. CONCLUSIONS: This study discloses the poor conjugation competence of HuISG15 compared to OWmISG15 and maps the critical determinants for efficient conjugation. HyperISGylation may greatly assist ISGylation studies and may enhance its function as positive regulator of Interferon-related immune responses or as anti-tumoral modulator.


Asunto(s)
Citocinas/metabolismo , Ubiquitinas/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Línea Celular , Cercopithecidae , Citocinas/química , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Ubiquitinas/química
16.
Mol Endocrinol ; 21(11): 2821-31, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17666591

RESUMEN

Binding of GH to its receptor induces rapid phosphorylation of conserved tyrosine motifs that function as recruitment sites for downstream signaling molecules. Using mammalian protein-protein interaction trap (MAPPIT), a mammalian two-hybrid method, we mapped the binding sites in the GH receptor for signal transducer and activator of transcription 5 (STAT5) a and b and for the negative regulators of cytokine signaling cytokine-inducible Src-homology 2 (SH2)-containing protein (CIS) and suppressor of cytokine signaling 2 (SOCS2). Y534, Y566, and Y627 are the major recruitment sites for STAT5. A non-overlapping recruitment pattern is observed for SOCS2 and CIS with positions Y487 and Y595 as major binding sites, ruling out SOCS-mediated inhibition of STAT5 activation by competition for shared binding sites. More detailed analysis revealed that CIS binding to the Y595, but not to the Y487 motif, depends on both its SH2 domain and the C-terminal part of its SOCS box, with a critical role for the CIS Y253 residue. This functional divergence of the two CIS/SOCS2 recruitment sites is also observed upon substitution of the Y+1 residue by leucine, turning the Y487, but not the Y595 motif into a functional STAT5 recruitment site.


Asunto(s)
Receptores de Somatotropina/metabolismo , Factor de Transcripción STAT5/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Secuencias de Aminoácidos , Línea Celular , Citocinas/metabolismo , Cartilla de ADN/química , Humanos , Modelos Biológicos , Conformación Molecular , Mutación , Péptidos/química , Unión Proteica , Mapeo de Interacción de Proteínas , Técnicas del Sistema de Dos Híbridos
17.
Nat Methods ; 2(6): 427-33, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15908921

RESUMEN

Interactions between proteins are at the heart of the cellular machinery. It is therefore not surprising that altered interaction profiles caused by aberrant protein expression patterns or by the presence of mutations can trigger cellular dysfunction, eventually leading to disease. Moreover, many viral and bacterial pathogens rely on protein-protein interactions to exert their damaging effects. Interfering with such interactions is an obvious pharmaceutical goal, but detailed insights into the protein binding properties as well as efficient screening platforms are needed. In this report, we describe a cytokine receptor-based assay with a positive readout to screen for disrupters of designated protein-protein interactions in intact mammalian cells and evaluate this concept using polypeptides as well as small organic molecules. These reverse mammalian protein-protein interaction trap (MAPPIT) screens were developed to monitor interactions between the erythropoietin receptor (EpoR) and suppressors of cytokine signaling (SOCS) proteins, between FKBP12 and ALK4, and between MDM2 and p53.


Asunto(s)
Mapeo de Interacción de Proteínas/métodos , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Humanos , Janus Quinasa 3 , Técnicas del Sistema de Dos Híbridos
18.
J Biol Chem ; 279(39): 41038-46, 2004 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-15213225

RESUMEN

The leptin/leptin receptor system shows strong similarities to the long-chain cytokine interleukin-6 (IL-6) and granulocyte colony-stimulating factor cytokine/receptor systems. The IL-6 family cytokines interact with their receptors through three different binding sites I-III. The leptin structure was superposed on the crystal structures of several long-chain cytokines, and a series of leptin mutants was generated focusing on binding sites I-III. The effect of the mutations on leptin receptor (LR) signaling and on binding to the membrane proximal cytokine receptor homology domain (CRH2) of the LR was determined. Mutations in binding site I at the C terminus of helix D show a modest effect on signaling and do not affect binding to CRH2. Binding site II is composed of residues at the surface of helices A and C. Mutations in this site impair binding to CRH2 but have only limited effect on signaling. Site III mutations around the N terminus of helix D impair receptor activation without affecting binding to CRH2. We identified an S120A/T121A mutant in binding site III, which lacks any signaling capacity, but which still binds to CRH2 with wild type affinity. This leptin mutant behaves as a potent leptin antagonist both in vitro and in vivo.


Asunto(s)
Diseño de Fármacos , Leptina/antagonistas & inhibidores , Leptina/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Unión Competitiva , Células COS , Línea Celular , Relación Dosis-Respuesta a Droga , Genes Reporteros , Humanos , Luciferasas/metabolismo , Ratones , Ratones Endogámicos DBA , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Homología de Secuencia de Aminoácido , Transducción de Señal , Factores de Tiempo , Transfección
19.
J Biol Chem ; 279(36): 37779-88, 2004 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-15218032

RESUMEN

In a previous characterization of the ABCA subfamily of the ATP-binding cassette (ABC) transporters, we identified potential protein kinase 2 (CK2) phosphorylation sites, which are conserved in eukaryotic and prokaryotic members of the ABCA transporters. These phosphorylation residues are located in the conserved cytoplamic R1 and R2 domains, downstream of the nucleotide binding domains NBD1 and NBD2. To study the possible regulation of the ABCA1 transporter by CK2, we expressed the recombinant cytoplasmic domains of ABCA1, NBD1+R1 and NBD2+R2. We demonstrated that in vitro ABCA1 NBD1+R1, and not NBD2+R2, is phosphorylated by CK2, and we identified Thr-1242, Thr-1243, and Ser-1255 as the phosphorylated residues in the R1 domain by mass spectrometry. We further investigated the functional significance of the threonine and serine phosphorylation sites in NBD1 by site-directed mutagenesis of the entire ABCA1 followed by transfection into Hek-293 Tet-Off cells. The ABCA1 flippase activity, apolipoprotein AI and AII binding, and cellular phospholipid and cholesterol efflux were enhanced by mutations preventing CK2 phosphorylation of the threonine and serine residues. This was confirmed by the effect of specific protein kinase CK2 inhibitors upon the activity of wild type and mutant ABCA1 in transfected Hek-293 Tet-Off cells. The activities of the mutants mimicking threonine phosphorylation were close to that of wild type ABCA1. Our data, therefore, suggest that besides protein kinase A and C, protein kinase CK2 might play an important role in vivo in regulating the function and transport activity of ABCA1 and possibly of other members of the ABCA subfamily.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Quinasa de la Caseína II/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Colesterol/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , Fosfolípidos/metabolismo , Fosforilación , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
20.
Sci STKE ; 2002(162): pl18, 2002 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-12476003

RESUMEN

Identifying the interaction partners of a protein is a straightforward way to gain insight into the protein's function and to position it in an interaction network such as a signal transduction pathway. Various techniques have been developed to serve this purpose, and some are specifically designed to study posttranslational modifications in mammalian proteins and to clarify their normal physiological context. However, several intrinsic constraints limit the use of these technologies, and most are not suitable for screening for new interacting partners. In the Mammalian Protein-Protein Interaction Trap (MAPPIT) Protocol described here, knowledge of cytokine receptor signaling has been used to design a versatile genetic tool that can be used analytically and for detection of new protein-protein interactions in mammalian cells.


Asunto(s)
Mapeo de Interacción de Proteínas/métodos , Proyectos de Investigación , Animales , Western Blotting , Línea Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Genes Reporteros/genética , Vectores Genéticos/genética , Humanos , Riñón/citología , Riñón/embriología , Riñón/metabolismo , Fosfotirosina/inmunología , Procesamiento Proteico-Postraduccional/fisiología , Proteínas/fisiología , Agregación de Receptores/fisiología , Receptores de Citocinas/química , Receptores de Citocinas/inmunología , Receptores de Citocinas/fisiología , Proteínas Recombinantes de Fusión/genética , Transducción de Señal/fisiología , Transfección , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA