Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
ACS Med Chem Lett ; 15(6): 864-872, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38894924

RESUMEN

We were attracted to the therapeutic potential of inhibiting Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b), a RING E3 ligase that plays a critical role in regulating the activation of T cells. However, given that only protein-protein interactions were involved, it was unclear whether inhibition by a small molecule would be a viable approach. After screening an ∼6 billion member DNA-encoded library (DEL) using activated Cbl-b, we identified compound 1 as a hit for which the cis-isomer (2) was confirmed by biochemical and surface plasmon resonance (SPR) assays. Our hit optimization effort was greatly accelerated when we obtained a cocrystal structure of 2 with Cbl-b, which demonstrated induced binding at the substrate binding site, namely, the Src homology-2 (SH2) domain. This was quite noteworthy given that there are few reports of small molecule inhibitors that bind to SH2 domains and block protein-protein interactions. Structure- and property-guided optimization led to compound 27, which demonstrated measurable cell activity, albeit only at high concentrations.

3.
Br J Pharmacol ; 181(13): 2033-2052, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38486310

RESUMEN

BACKGROUND AND PURPOSE: RO7502175 is an afucosylated antibody designed to eliminate C-C motif chemokine receptor 8 (CCR8)+ Treg cells in the tumour microenvironment through enhanced antibody-dependent cellular cytotoxicity (ADCC). EXPERIMENTAL APPROACH: We report findings from preclinical studies characterizing pharmacology, pharmacokinetics (PK)/pharmacodynamics (PD) and safety profile of RO7502175 and discuss the translational PK/PD approach used to inform first-in-human (FiH) dosing strategy and clinical development in solid tumour indications. KEY RESULTS: RO7502175 demonstrated selective ADCC against human CCR8+ Treg cells from dissociated tumours in vitro. In cynomolgus monkeys, RO7502175 exhibited a biphasic concentration-time profile consistent with immunoglobulin G1 (IgG1) antibodies, reduced CCR8+ Treg cells in the blood, induced minimal and transient cytokine secretion, and was well tolerated with a no-observed-adverse-effect level (NOAEL) of 100 mg·kg-1. Moreover, RO7502175 caused minimal cytokine release from peripheral blood mononuclear cells (PBMCs) in vitro. A quantitative model was developed to capture surrogate anti-murine CCR8 antibody PK/PD and tumour dynamics in mice and RO7502175 PK/PD in cynomolgus monkeys. Subsequently, the model was used to project RO7502175 human PK and receptor occupancy (RO) in patients. Because traditional approaches resulted in a low FiH dose for this molecule, even with its superior preclinical safety profile, an integrated approach based on the totality of preclinical data and modelling insights was used for starting dose selection. CONCLUSION AND IMPLICATIONS: This work demonstrates a translational research strategy for collecting and utilizing relevant nonclinical data, developing a mechanistic PK/PD model and using a comprehensive approach to inform clinical study design for RO7502175.


Asunto(s)
Macaca fascicularis , Receptores CCR8 , Linfocitos T Reguladores , Animales , Humanos , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Receptores CCR8/antagonistas & inhibidores , Receptores CCR8/inmunología , Ratones , Femenino , Masculino , Investigación Biomédica Traslacional , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/administración & dosificación , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Relación Dosis-Respuesta a Droga , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos
4.
Nat Commun ; 14(1): 7940, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38040762

RESUMEN

The C-C motif chemokine receptor 8 (CCR8) is a class A G-protein coupled receptor that has emerged as a promising therapeutic target in cancer. Targeting CCR8 with an antibody has appeared to be an attractive therapeutic approach, but the molecular basis for chemokine-mediated activation and antibody-mediated inhibition of CCR8 are not fully elucidated. Here, we obtain an antagonist antibody against human CCR8 and determine structures of CCR8 in complex with either the antibody or the endogenous agonist ligand CCL1. Our studies reveal characteristic antibody features allowing recognition of the CCR8 extracellular loops and CCL1-CCR8 interaction modes that are distinct from other chemokine receptor - ligand pairs. Informed by these structural insights, we demonstrate that CCL1 follows a two-step, two-site binding sequence to CCR8 and that antibody-mediated inhibition of CCL1 signaling can occur by preventing the second binding event. Together, our results provide a detailed structural and mechanistic framework of CCR8 activation and inhibition that expands our molecular understanding of chemokine - receptor interactions and offers insight into the development of therapeutic antibodies targeting chemokine GPCRs.


Asunto(s)
Quimiocinas CC , Receptores de Quimiocina , Humanos , Quimiocinas CC/metabolismo , Quimiocinas CC/farmacología , Receptores CCR8/genética , Ligandos , Quimiocina CCL1/metabolismo , Receptores de Quimiocina/genética , Anticuerpos
5.
Cancer Immunol Res ; 7(6): 963-976, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31064777

RESUMEN

Exhausted T cells have been described in cancer patients and murine tumor models largely based on their expression of various inhibitory receptors. Understanding of the functional attributes of these cells is limited. Here, we report that among CD8+ T cells in commonly used syngeneic tumor models, the coexpression of inhibitory receptors PD-1, LAG3, and TIM3 defined a group of highly activated and functional effector cells. Coexpression of these receptors further enriched for antigen-specific cells with increased T-cell receptor clonality. Anti-PD-L1 treatment increased the number and activation of these triple-positive CD8+ T cells without affecting the density of PD-1- cells. The intratumoral density of CD8+ T cells coexpressing inhibitory receptors negatively correlated with tumor burden. The density ratio and pretreatment phenotype of CD8+ T cells coexpressing inhibitory receptors was positively correlated with response across a variety of tumor models. Our results demonstrate that coexpression of inhibitory receptors is not a signifier of exhausted T cells, but rather can define a group of activated and functional effector cells in syngeneic tumor models. In the cancer setting, these cells could represent a heterogeneous population of not only exhausted but also highly activated cells responsive to treatment.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Receptores Coestimuladores e Inhibidores de Linfocitos T/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Neoplasias/etiología , Neoplasias/metabolismo , Animales , Antígeno B7-H1/antagonistas & inhibidores , Biomarcadores de Tumor , Línea Celular Tumoral , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Femenino , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Isoinjertos , Ratones , Neoplasias/patología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
6.
Nat Immunol ; 20(4): 471-481, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30778241

RESUMEN

Foxp3+ regulatory T cells (Treg cells) are crucial for the maintenance of immune homeostasis both in lymphoid tissues and in non-lymphoid tissues. Here we demonstrate that the ability of intestinal Treg cells to constrain microbiota-dependent interleukin (IL)-17-producing helper T cell (TH17 cell) and immunoglobulin A responses critically required expression of the transcription factor c-Maf. The terminal differentiation and function of several intestinal Treg cell populations, including RORγt+ Treg cells and follicular regulatory T cells, were c-Maf dependent. c-Maf controlled Treg cell-derived IL-10 production and prevented excessive signaling via the kinases PI(3)K (phosphatidylinositol-3-OH kinase) and Akt and the metabolic checkpoint kinase complex mTORC1 (mammalian target of rapamycin) and expression of inflammatory cytokines in intestinal Treg cells. c-Maf deficiency in Treg cells led to profound dysbiosis of the intestinal microbiota, which when transferred to germ-free mice was sufficient to induce exacerbated intestinal TH17 responses, even in a c-Maf-competent environment. Thus, c-Maf acts to preserve the identity and function of intestinal Treg cells, which is essential for the establishment of host-microbe symbiosis.


Asunto(s)
Inmunoglobulina A/biosíntesis , Intestinos/inmunología , Microbiota , Proteínas Proto-Oncogénicas c-maf/fisiología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Células Cultivadas , Colitis/inmunología , Citocinas/metabolismo , Disbiosis , Regulación de la Expresión Génica , Homeostasis , Interleucina-10/biosíntesis , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-maf/genética , Proteínas Proto-Oncogénicas c-maf/metabolismo , Linfocitos T Reguladores/enzimología
7.
Front Immunol ; 9: 1482, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30022979

RESUMEN

Peripheral T lymphocytes share many functional properties with hematopoietic stem cells (HSCs), including long-term maintenance, quiescence, and latent proliferative potential. In addition, peripheral T cells retain the capacity for further differentiation into a variety of subsets, much like HSCs. While the similarities between T cells and HSC have long been hypothesized, the potential common genetic regulation of HSCs and T cells has not been widely explored. We have studied the T cell-intrinsic role of Zfx, a transcription factor specifically required for HSC maintenance. We report that T cell-specific deletion of Zfx caused age-dependent depletion of naïve peripheral T cells. Zfx-deficient T cells also failed to undergo homeostatic proliferation in a lymphopenic environment, and showed impaired antigen-specific expansion and memory response. In addition, the invariant natural killer T cell compartment was severely reduced. RNA-Seq analysis revealed that the most dysregulated genes in Zfx-deficient T cells were similar to those observed in Zfx-deficient HSC and B cells. These studies identify Zfx as an important regulator of peripheral T cell maintenance and expansion and highlight the common molecular basis of HSC and lymphocyte homeostasis.

8.
Sci Immunol ; 3(22)2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29678836

RESUMEN

Loss of function of the nuclear deubiquitinating enzyme BRCA1-associated protein-1 (BAP1) is associated with a wide spectrum of cancers. We report that tamoxifen-induced BAP1 deletion in adult mice resulted in severe thymic atrophy. BAP1 was critical for T cell development at several stages. In the thymus, BAP1 was required for progression through the pre-T cell receptor checkpoint. Peripheral T cells lacking BAP1 demonstrated a defect in homeostatic and antigen-driven expansion. Deletion of BAP1 resulted in suppression of E2F target genes and defects in cell cycle progression, which was dependent on the catalytic activity of BAP1, but did not require its interaction with host cell factor-1 (HCF-1). Loss of BAP1 led to increased monoubiquitination of histone H2A at Lys119 (H2AK119ub) throughout the T cell lineage, in particular in immature thymocytes, but did not alter trimethylation of histone H3 at Lys27 (H3K27me3). Deletion of BAP1 also abrogated B cell development in the bone marrow. Our findings uncover a nonredundant function for BAP1 in maintaining the lymphoid lineage.


Asunto(s)
Linfocitos T/metabolismo , Timocitos/metabolismo , Timo/metabolismo , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Animales , Atrofia , Ciclo Celular/genética , Perfilación de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Lisina/genética , Lisina/metabolismo , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Timo/patología , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación
9.
Genes Dev ; 29(17): 1801-16, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26314708

RESUMEN

The DNA-binding protein Ikaros is a potent tumor suppressor and hematopoietic regulator. However, the mechanisms by which Ikaros functions remain poorly understood, due in part to its atypical DNA-binding properties and partnership with the poorly understood Mi-2/NuRD complex. In this study, we analyzed five sequential stages of thymocyte development in a mouse strain containing a targeted deletion of Ikaros zinc finger 4, which exhibits a select subset of abnormalities observed in Ikaros-null mice. By examining thymopoiesis in vivo and in vitro, diverse abnormalities were observed at each developmental stage. RNA sequencing revealed that each stage is characterized by the misregulation of a limited number of genes, with a strong preference for stage-specific rather than lineage-specific genes. Strikingly, individual genes rarely exhibited Ikaros dependence at all stages. Instead, a consistent feature of the aberrantly expressed genes was a reduced magnitude of expression level change during developmental transitions. These results, combined with analyses of the interplay between Ikaros loss of function and Notch signaling, suggest that Ikaros may not be a conventional activator or repressor of defined sets of genes. Instead, a primary function may be to sharpen the dynamic range of gene expression changes during developmental transitions via atypical molecular mechanisms that remain undefined.


Asunto(s)
Proteínas Portadoras/metabolismo , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas del Tejido Nervioso/metabolismo , Timocitos/citología , Animales , Proteínas Portadoras/genética , Puntos de Control del Ciclo Celular , Células Cultivadas , Proteínas de Unión al ADN , Femenino , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Masculino , Ratones , Proteínas del Tejido Nervioso/genética , Receptor Notch1/genética , Receptor Notch1/metabolismo , Análisis de Secuencia de ARN
10.
Mol Cell Biol ; 34(11): 2017-28, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24662052

RESUMEN

Chronic inflammation is known to be associated with prostate cancer development, but how epithelium-associated cancer-initiating events cross talk to inflammatory cells during prostate cancer initiation and progression is largely unknown. Using the Pten null murine prostate cancer model, we show an expansion of Gr-1(+) CD11b(+) myeloid-derived suppressor cells (MDSCs) occurring intraprostatically immediately following epithelium-specific Pten deletion without expansion in hematopoietic tissues. This MDSC expansion is accompanied by sustained immune suppression. Prostatic Gr-1(+) CD11b(+) cells, but not those isolated from the spleen of the same tumor-bearing mice, suppress T cell proliferation and express high levels of Arginase 1 and iNOS. Mechanistically, the loss of PTEN in the epithelium leads to a significant upregulation of genes within the inflammatory response and cytokine-cytokine receptor interaction pathways, including Csf1 and Il1b, two genes known to induce MDSC expansion and immunosuppressive activities. Treatment of Pten null mice with the selective CSF-1 receptor inhibitor GW2580 decreases MDSC infiltration and relieves the associated immunosuppressive phenotype. Our study indicates that epithelium-associated tumor-initiating events trigger the secretion of inflammatory cytokines and promote localized MDSC expansion and immune suppression, thereby promoting tumor progression.


Asunto(s)
Tolerancia Inmunológica , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/inmunología , Animales , Anisoles/farmacología , Arginasa/biosíntesis , Antígeno CD11b/metabolismo , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Epitelio/inmunología , Epitelio/patología , Inflamación/inmunología , Inflamación/patología , Interleucina-1beta/biosíntesis , Activación de Linfocitos/inmunología , Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Factor Estimulante de Colonias de Macrófagos/biosíntesis , Masculino , Ratones , Ratones Transgénicos , Células Mieloides/inmunología , Células Mieloides/patología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Próstata/patología , Neoplasias de la Próstata/genética , Pirimidinas/farmacología , Receptores de Quimiocina/metabolismo , Transducción de Señal , Bazo/citología , Bazo/inmunología , Linfocitos T/inmunología , Regulación hacia Arriba
11.
Cell Rep ; 6(3): 528-40, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24485662

RESUMEN

Tumor-propagating cells in acute leukemia maintain a stem/progenitor-like immature phenotype and proliferative capacity. Acute myeloid leukemia (AML) and acute T-lymphoblastic leukemia (T-ALL) originate from different lineages through distinct oncogenic events such as MLL fusions and Notch signaling, respectively. We found that Zfx, a transcription factor that controls hematopoietic stem cell self-renewal, controls the initiation and maintenance of AML caused by MLL-AF9 fusion and of T-ALL caused by Notch1 activation. In both leukemia types, Zfx prevents differentiation and activates gene sets characteristic of immature cells of the respective lineages. In addition, endogenous Zfx contributes to gene induction and transformation by Myc overexpression in myeloid progenitors. Key Zfx target genes include the mitochondrial enzymes Ptpmt1 and Idh2, whose overexpression partially rescues the propagation of Zfx-deficient AML. These results show that distinct leukemia types maintain their undifferentiated phenotype and self-renewal by exploiting a common stem-cell-related genetic regulator.


Asunto(s)
Diferenciación Celular , Factores de Transcripción de Tipo Kruppel/metabolismo , Leucemia Mieloide Aguda/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/patología , Células Clonales , Regulación Leucémica de la Expresión Génica , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Leucemia Mieloide Aguda/genética , Ratones , Mitocondrias/enzimología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fenotipo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores Notch/metabolismo
12.
Nat Immunol ; 14(10): 1073-83, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24013668

RESUMEN

C2H2 zinc fingers are found in several key transcriptional regulators in the immune system. However, these proteins usually contain more fingers than are needed for sequence-specific DNA binding, which suggests that different fingers regulate different genes and functions. Here we found that mice lacking finger 1 or finger 4 of Ikaros exhibited distinct subsets of the hematological defects of Ikaros-null mice. Most notably, the two fingers controlled different stages of lymphopoiesis, and finger 4 was selectively required for tumor suppression. The distinct defects support the hypothesis that only a small number of genes that are targets of Ikaros are critical for each of its biological functions. The subcategorization of functions and target genes by mutagenesis of individual zinc fingers will facilitate efforts to understand how zinc-finger transcription factors regulate development, immunity and disease.


Asunto(s)
Transformación Celular Neoplásica/genética , Regulación de la Expresión Génica , Factor de Transcripción Ikaros/genética , Leucemia/genética , Linfopoyesis/genética , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Secuencia de Bases , Sitios de Unión , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Inmunoprecipitación de Cromatina , Análisis por Conglomerados , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Perfilación de la Expresión Génica , Mutación de Línea Germinal , Secuenciación de Nucleótidos de Alto Rendimiento , Factor de Transcripción Ikaros/metabolismo , Inmunofenotipificación , Leucemia/metabolismo , Leucemia/mortalidad , Linfoma/genética , Linfoma/metabolismo , Linfoma/mortalidad , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Motivos de Nucleótidos , Fenotipo , Posición Específica de Matrices de Puntuación , Unión Proteica , Timocitos/metabolismo
13.
Blood ; 113(23): 5857-67, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19329779

RESUMEN

The development, homeostasis, and function of B lymphocytes involve multiple rounds of B-cell receptor (BCR)-controlled proliferation and prolonged maintenance. We analyzed the role of transcription factor Zfx, a recently identified regulator of hematopoietic stem cell maintenance, in B-cell development and homeostasis. Panhematopoietic or B cell-specific deletion of Zfx in the bone marrow blocked B-cell development at the pre-BCR selection checkpoint. Zfx deficiency in peripheral B cells caused accelerated B-cell turnover, depletion of mature recirculating B cells, and delayed T-dependent antibody responses. In addition, the numbers and function of B-1 cell lineage were reduced. Zfx-deficient B cells showed normal proximal BCR signaling, but impaired BCR-induced proliferation and survival in vitro. This was accompanied by aberrantly enhanced and prolonged integrated stress response and by delayed induction of cyclin D2 and Bcl-xL proteins. Thus, Zfx restrains the stress response and couples antigen receptor signaling to cell expansion and maintenance during B-cell development and peripheral homeostasis. These results identify a novel transcriptional regulator of the B-cell lineage and highlight the common genetic control of stem cell maintenance and lymphocyte homeostasis.


Asunto(s)
Linfocitos B/citología , Linfocitos B/inmunología , Factores de Transcripción de Tipo Kruppel/metabolismo , Receptores de Antígenos de Linfocitos B/inmunología , Animales , Linfocitos B/metabolismo , Diferenciación Celular/inmunología , Proliferación Celular , Supervivencia Celular/inmunología , Homeostasis/inmunología , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Noqueados , Linfocitos T/inmunología
14.
Cell ; 129(2): 345-57, 2007 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-17448993

RESUMEN

Stem cells (SC) exhibit a unique capacity for self-renewal in an undifferentiated state. It is unclear whether the self-renewal of pluripotent embryonic SC (ESC) and of tissue-specific adult SC such as hematopoietic SC (HSC) is controlled by common mechanisms. The deletion of transcription factor Zfx impaired the self-renewal but not the differentiation capacity of murine ESC; conversely, Zfx overexpression facilitated ESC self-renewal by opposing differentiation. Furthermore, Zfx deletion abolished the maintenance of adult HSC but did not affect erythromyeloid progenitors or fetal HSC. Zfx-deficient ESC and HSC showed increased apoptosis and SC-specific upregulation of stress-inducible genes. Zfx directly activated common target genes in ESC and HSC, as well as ESC-specific target genes including ESC self-renewal regulators Tbx3 and Tcl1. These studies identify Zfx as a shared transcriptional regulator of ESC and HSC, suggesting a common genetic basis of self-renewal in embryonic and adult SC.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Factores de Transcripción de Tipo Kruppel/metabolismo , Animales , Apoptosis , Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/metabolismo , Femenino , Expresión Génica , Marcación de Gen , Células Madre Hematopoyéticas/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Proteínas Proto-Oncogénicas/genética , Proteínas de Dominio T Box/genética
15.
Mol Cell Biol ; 24(20): 9092-101, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15456882

RESUMEN

The regulation of cytokine signaling is critical for controlling cellular proliferation and activation during an immune response. SOCS-1 is a potent inhibitor of Jak kinase activity and of signaling initiated by several cytokines. SOCS-1 protein levels are tightly regulated, and recent data suggest that SOCS-1 may regulate the protein levels of some signaling proteins by the ubiquitin proteasome pathway; however, the cellular mechanism by which SOCS-1 directs proteins for degradation is unknown. In this report, SOCS-1 is found to colocalize and biochemically copurify with the microtubule organizing complex (MTOC) and its associated 20S proteasome. The SOCS-1 SH2 domain is required for the localization of SOCS-1 to the MTOC. Overexpression of SOCS-1 targets Jak1 in an SH2-dependent manner to a perinuclear distribution resembling the MTOC-associated 20S proteasome. Analysis of MTOCs fractionated from SOCS-1-deficient cells demonstrates that SOCS-1 may function redundantly to regulate the localization of Jak1 to the MTOC. Nocodazole inhibits the protein turnover of SOCS-1, demonstrating that the minus-end transport of SOCS-1 to the MTOC-associated 20S proteasome is required to regulate SOCS-1 protein levels. These data link SOCS-1 directly with the proteasome pathway and suggest another function for the SH2 domain of SOCS-1 in the regulation of Jak/STAT signaling.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Centro Organizador de los Microtúbulos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Represoras/metabolismo , Animales , Fraccionamiento Celular , Línea Celular , Centrosoma/química , Centrosoma/metabolismo , Chlorocebus aethiops , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Janus Quinasa 1 , Microtúbulos/metabolismo , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Fracciones Subcelulares/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA