Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Chem Biol ; 31(8): 1473-1489, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39111317

RESUMEN

The growing clinical success of bispecific antibodies (bsAbs) has led to rapid interest in leveraging dual targeting in order to generate novel modes of therapeutic action beyond mono-targeting approaches. While bsAbs that bind targets on two different cells (trans-targeting) are showing promise in the clinic, the co-targeting of two proteins on the same cell surface through cis-targeting bsAbs (cis-bsAbs) is an emerging strategy to elicit new functionalities. This includes the ability to induce proximity, enhance binding to a target, increase target/cell selectivity, and/or co-modulate function on the cell surface with the goal of altering, reversing, or eradicating abnormal cellular activity that contributes to disease. In this review, we focus on the impact of cis-bsAbs in the clinic, their emerging applications, and untangle the intricacies of improving bsAb discovery and development.


Asunto(s)
Anticuerpos Biespecíficos , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/inmunología , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Animales
2.
mBio ; 15(3): e0008324, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38376210

RESUMEN

Mice that lack the genes for IL-27, or the IL-27 receptor, and infected with Toxoplasma gondii develop T cell-mediated pathology. Here, studies were performed to determine the impact of endogenous IL-27 on the immune response to T. gondii in wild-type (WT) mice. Analysis of infected mice revealed the early production of IL-27p28 by a subset of Ly6Chi, inflammatory monocytes, and sustained IL-27p28 production at sites of acute and chronic infection. Administration of anti-IL-27p28 prior to infection resulted in an early (day 5) increase in levels of macrophage and granulocyte activation, as well as enhanced effector T cell responses, as measured by both cellularity, cytokine production, and transcriptional profiling. This enhanced acute response led to immune pathology, while blockade during the chronic phase of infection resulted in enhanced T cell responses but no systemic pathology. In the absence of IL-27, the enhanced monocyte responses observed at day 10 were a secondary consequence of activated CD4+ T cells. Thus, in WT mice, IL-27 has distinct suppressive effects that impact innate and adaptive immunity during different phases of this infection. IMPORTANCE: The molecule IL-27 is critical in limiting the immune response to the parasite Toxoplasma gondii. In the absence of IL-27, a lethal, overactive immune response develops during infection. However, when exactly in the course of infection this molecule is needed was unclear. By selectively inhibiting IL-27 during this parasitic infection, we discovered that IL-27 was only needed during, but not prior to, infection. Additionally, IL-27 is only needed in the active areas in which the parasite is replicating. Finally, our work found that a previously unstudied cell type, monocytes, was regulated by IL-27, which contributes further to our understanding of the regulatory networks established by this molecule.


Asunto(s)
Interleucina-27 , Toxoplasma , Toxoplasmosis , Animales , Ratones , Interleucina-27/metabolismo , Ratones Endogámicos C57BL , Monocitos , Linfocitos T , Toxoplasmosis/parasitología
3.
Arthritis Rheumatol ; 75(8): 1344-1356, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36862144

RESUMEN

OBJECTIVE: CD28 and inducible T cell costimulator (ICOS) appear to have nonredundant roles in T cell activation and adaptive immunity. We undertook this study to characterize in vitro and in vivo the therapeutic potential of acazicolcept (ALPN-101), an Fc fusion protein of a human variant ICOS ligand (ICOSL) domain designed to inhibit both CD28 and ICOS costimulation, in inflammatory arthritis. METHODS: Acazicolcept was compared in vitro with inhibitors of either the CD28 or ICOS pathways (abatacept and belatacept [CTLA-4Ig], prezalumab [anti-ICOSL monoclonal antibody]) in receptor binding and signaling assays, and in a collagen-induced arthritis (CIA) model. Acazicolcept was also compared in cytokine and gene expression assays of peripheral blood mononuclear cells (PBMCs) from healthy donors or rheumatoid arthritis (RA) or psoriatic arthritis (PsA) patients stimulated with artificial antigen-presenting cells (APCs) expressing CD28 and ICOS ligands*. RESULTS: Acazicolcept bound CD28 and ICOS, prevented ligand binding, and inhibited human T cell functional interactions, matching or exceeding the activity of CD28 or ICOS costimulatory single-pathway inhibitors tested individually or in combination. Acazicolcept administration significantly reduced disease in the CIA model and more potently than abatacept. Acazicolcept also inhibited proinflammatory cytokine production from stimulated PBMCs in cocultures with artificial APCs and demonstrated unique effects on gene expression distinct from those induced by abatacept, prezalumab, or a combination of both. CONCLUSION: Both CD28 and ICOS signaling play critical roles in inflammatory arthritis. Therapeutic agents such as acazicolcept that coinhibit both ICOS and CD28 signaling may mitigate inflammation and/or disease progression in RA and PsA more effectively than inhibitors of either pathway alone.


Asunto(s)
Artritis Psoriásica , Artritis Reumatoide , Humanos , Antígenos CD28/metabolismo , Abatacept/farmacología , Abatacept/uso terapéutico , Leucocitos Mononucleares/metabolismo , Ligandos , Proteína Coestimuladora de Linfocitos T Inducibles , Linfocitos T , Factores Inmunológicos , Artritis Reumatoide/tratamiento farmacológico , Anticuerpos Monoclonales/farmacología , Citocinas
4.
Arthritis Rheumatol ; 75(7): 1187-1202, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36705554

RESUMEN

OBJECTIVE: Dysregulated APRIL/BAFF signaling is implicated in the pathogenesis of multiple autoimmune diseases, including systemic lupus erythematosus and lupus nephritis. We undertook this study to develop and evaluate a high-affinity APRIL/BAFF antagonist to overcome the clinical limitations of existing B cell inhibitors. METHODS: A variant of TACI-Fc generated by directed evolution showed enhanced binding for both APRIL and BAFF and was designated povetacicept (ALPN-303). Povetacicept was compared to wild-type (WT) TACI-Fc and related molecules in vitro and in vivo. RESULTS: Povetacicept inhibited APRIL and BAFF more effectively than all evaluated forms of WT TACI-Fc and selective APRIL and BAFF inhibitors in cell-based reporter assays and primary human B cell assays, mediating potent suppression of B cell proliferation, differentiation, and immunoglobulin (Ig) secretion. In mouse immunization models, povetacicept significantly reduced serum immunoglobulin titers and antibody-secreting cells more effectively than anti-CD20 monoclonal antibodies, WT TACI-Fc, or APRIL and BAFF inhibitors. In the NZB × NZW mouse lupus nephritis model, povetacicept significantly enhanced survival and suppressed proteinuria, anti-double-stranded DNA antibody titers, blood urea nitrogen, glomerulonephritis, and renal immunoglobulin deposition. In the bm12 mouse lupus model, povetacicept significantly reduced splenic plasmablasts, follicular helper T cells, and germinal center B cells. In non-human primates, povetacicept was well tolerated, exhibited high serum exposure, and significantly decreased serum IgM, IgA, and IgG levels after a single dose. CONCLUSION: Enhanced APRIL and BAFF inhibition by povetacicept led to greater inhibition of B cell populations critical for autoantibody production compared to WT TACI-Fc and CD20-, APRIL-, or BAFF-selective inhibitors. Potent, dual inhibition by povetacicept has the potential to significantly improve clinical outcomes in autoantibody-related autoimmune diseases.


Asunto(s)
Lupus Eritematoso Sistémico , Nefritis Lúpica , Ratones , Animales , Humanos , Autoanticuerpos , Factor Activador de Células B/genética , Linfocitos B , Ratones Endogámicos
5.
J Immunother Cancer ; 8(1)2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32345627

RESUMEN

BACKGROUND: CD47 is a broadly expressed cell surface glycoprotein associated with immune evasion. Interaction with the inhibitory receptor signal regulatory protein alpha (SIRPα), primarily expressed on myeloid cells, normally serves to restrict effector function (eg, phagocytosis and immune cell homeostasis). CD47/SIRPα antagonists, commonly referred to as 'macrophage checkpoint' inhibitors, are being developed as cancer interventions. SRF231 is an investigational fully human IgG4 anti-CD47 antibody that is currently under evaluation in a phase 1 clinical trial. The development and preclinical characterization of SRF231 are reported here. METHODS: SRF231 was characterized in assays designed to probe CD47/SIRPα blocking potential and effects on red blood cell (RBC) phagocytosis and agglutination. Additionally, SRF231-mediated phagocytosis and cell death were assessed in macrophage:tumor cell in vitro coculture systems. Further mechanistic studies were conducted within these coculture systems to ascertain the dependency of SRF231-mediated antitumor activity on Fc receptor engagement vs CD47/SIRPα blockade. In vivo, SRF231 was evaluated in a variety of hematologic xenograft models, and the mechanism of antitumor activity was assessed using cytokine and macrophage infiltration analyses following SRF231 treatment. RESULTS: SRF231 binds CD47 and disrupts the CD47/SIRPα interaction without causing hemagglutination or RBC phagocytosis. SRF231 exerts antitumor activity in vitro through both phagocytosis and cell death in a manner dependent on the activating Fc-gamma receptor (FcγR), CD32a. Through its Fc domain, SRF231 engagement with macrophage-derived CD32a serves dual purposes by eliciting FcγR-mediated phagocytosis of cancer cells and acting as a scaffold to drive CD47-mediated death signaling into tumor cells. Robust antitumor activity occurs across multiple hematologic xenograft models either as a single agent or in combination with rituximab. In tumor-bearing mice, SRF231 increases tumor macrophage infiltration and induction of the macrophage cytokines, mouse chemoattractant protein 1 and macrophage inflammatory protein 1 alpha. Macrophage depletion results in diminished SRF231 antitumor activity, underscoring a mechanistic role for macrophage engagement by SRF231. CONCLUSION: SRF231 elicits antitumor activity via apoptosis and phagocytosis involving macrophage engagement in a manner dependent on the FcγR, CD32a.


Asunto(s)
Antígeno CD47/metabolismo , Neoplasias/genética , Receptores de IgG/metabolismo , Animales , Humanos , Ratones , Neoplasias/patología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cancer Cell ; 26(2): 177-89, 2014 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-25043603

RESUMEN

Death receptor agonist therapies have exhibited limited clinical benefit to date. Investigations into why Apo2L/TRAIL and AMG 655 preclinical data were not predictive of clinical response revealed that coadministration of Apo2L/TRAIL with AMG 655 leads to increased antitumor activity in vitro and in vivo. The combination of Apo2L/TRAIL and AMG 655 results in enhanced signaling and can sensitize Apo2L/TRAIL-resistant cells. Structure determination of the Apo2L/TRAIL-DR5-AMG 655 ternary complex illustrates how higher order clustering of DR5 is achieved when both agents are combined. Enhanced agonism generated by combining Apo2L/TRAIL and AMG 655 provides insight into the limited efficacy observed in previous clinical trials and suggests testable hypotheses to reconsider death receptor agonism as a therapeutic strategy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Animales , Anticuerpos Monoclonales/química , Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular , Cristalografía por Rayos X , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Ratones , Modelos Moleculares , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/química , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/química , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cytokine Growth Factor Rev ; 25(2): 185-93, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24418173

RESUMEN

The activation of cell-surface death receptors represents an attractive therapeutic strategy to promote apoptosis of tumor cells. Several investigational therapeutics that target this extrinsic pathway, including recombinant human Apo2L/TRAIL and monoclonal agonist antibodies directed against death receptors-4 (DR4) or -5 (DR5), have been evaluated in the clinic. Although Phase 1/1b studies provided encouraging preliminary results, findings from randomized Phase 2 studies failed to demonstrate significant clinical benefit. This has raised multiple questions as to why pre-clinical data were not predictive of clinical response. Results from clinical studies and insight into why current agents have failed to yield robust responses are discussed. In addition, new strategies for the development of next generation death receptor agonists are reviewed.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Receptores de Muerte Celular/agonistas , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Anticuerpos Monoclonales/inmunología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Humanos , Neoplasias/tratamiento farmacológico , Proteínas Recombinantes/farmacología , Insuficiencia del Tratamiento
8.
Cancer Lett ; 332(2): 156-62, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-21220186

RESUMEN

The early observation that Apo2L/TRAIL preferentially triggers apoptosis in tumor cells over normal cells highlighted its potential as a candidate therapeutic in cancer. Since its identification in the mid-1990s, our increased understanding of Apo2L/TRAIL and Apo2L/TRAIL receptor signaling has led to the development of several agonists designed to promote tumor cell apoptosis through death receptor engagement. Recombinant human Apo2L/TRAIL/dulanermin is unique in that it is the only agonist which binds both Apo2L/TRAIL death receptors. In pre-clinical studies dulanermin demonstrates broad spectrum anti-tumor activity and the ability to cooperate with multiple conventional and targeted therapies. Results from early stage clinical trials indicate that dulanermin is well tolerated and shows some evidence of clinical activity. Not all tumors are likely to be equally sensitive to apoptosis induction by Apo2L/TRAIL. Therefore, an increased understanding of the regulation of Apo2L/TRAIL signaling should aid in the identification of molecular signatures that define a patient population likely to respond. In this review, current knowledge and new insights about Apo2L/TRAIL signaling is discussed with the focus on the development of Apo2L/TRAIL as a cancer therapeutic.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/tratamiento farmacológico , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores de Tumor/metabolismo , Humanos , Neoplasias/patología , Proteínas Recombinantes/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico
9.
Cancer Biol Ther ; 9(7): 539-50, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20150760

RESUMEN

In bone metastases, tumor cells interact with the bone microenvironment to induce osteoclastogenesis, leading to bone destruction and the growth factor release.  RANK ligand (RANKL) is essential for osteoclast formation, function, and survival.  Tumor cell-mediated osteolysis is thought to occur ultimately via induction of RANKL within the bone stroma, and inhibition of RANKL in models of breast cancer bone metastases blocks tumor-induced osteolysis and reduces skeletal tumor burden.  In addition, the skeleton is co-opted by tumor cells and functions as a supportive tumor microenvironment.  Inhibition of RANKL, by reducing tumor-induced osteoclastogenesis, may reduce the local release of growth factors and calcium which may potentially enhance the anti-tumor activity of cytotoxic or direct tumor apoptotic agents.  Recombinant human Apo2 ligand/ TNF-related apoptosis-inducing ligand (rhApo2L/TRAIL/dulanermin) is a dual pro-apoptotic receptor agonist that preferentially induces apoptosis in cancer cells versus normal cells.  We therefore examined RANKL inhibition (using RANK-Fc) in combination with rhApo2L/TRAIL on tumor-induced osteolysis and skeletal tumor burden in a murine intracardiac injection model of MDA-MB-231 breast carcinoma bone metastasis.  rhApo2L/TRAIL treatment resulted in a rapid reduction of skeletal tumor burden. Treatment with RANK-Fc prevented osteolytic lesions and reduced skeletal tumor burden. Combining RANK-Fc with rhApo2L/TRAIL was superior to either rhApo2L/TRAIL or RANK-Fc alone at reducing skeletal tumor burden in the bone metastasis model.  Our findings show that RANKL inhibition effectively inhibits pathologic osteolysis induced by human breast adenocarcinoma MDA-MB-231 cells in animals with established tumors, and also enhances the ability of rhApo2L/TRAIL to reduce skeletal tumor burden in vivo.


Asunto(s)
Adenocarcinoma/terapia , Neoplasias Óseas/terapia , Neoplasias de la Mama/terapia , Terapia Genética , Ligando RANK/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Recombinantes/genética , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Adenocarcinoma/genética , Adenocarcinoma/secundario , Animales , Apoptosis , Western Blotting , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Quimioterapia Combinada , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Ratones , Ratones SCID , Osteólisis , Carga Tumoral
10.
J Invest Dermatol ; 130(1): 102-12, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19626033

RESUMEN

The receptor-interacting protein (RIP) family kinase RIP4 interacts with protein kinase C (PKC) isoforms and is implicated in PKC-dependent signaling pathways. RIP4(-/-) mice die at birth with epidermal differentiation defects, causing fusions of all external orifices and loss of the esophageal lumen. To further understand RIP4 function in the skin, we generated transgenic mice with epidermal-specific expression of RIP4 using the human keratin-14 promoter (K14-RIP4). The K14-RIP4 transgene rescued the epidermal phenotype of RIP4(-/-) mice, showing that RIP4 acts autonomously in the epidermis to regulate differentiation. Although RIP4(-/-) mice share many phenotypic similarities with inhibitor kappaB kinase (IKK)alpha(-/-) mice and stratifin repeated epilation (Sfn(Er/Er)) mice, the K14-RIP4 transgene failed to promote epidermal differentiation in these mutant backgrounds. Unexpectedly, topical treatment of K14-RIP4 mice with 12-O-tetradecanoylphorbol-13-acetate (TPA) induced dramatic, neutrophilic inflammation, an effect that was independent of tumor necrosis factor type 1 receptor (TNFR1/p55) function. Despite their enhanced sensitivity to TPA, K14-RIP4 mice did not have an altered frequency of tumor formation in TPA-promoted skin cancer initiated with 7,12-dimethylbenz[a]anthracene (DMBA). These data suggest that RIP4 functions in the epidermis through PKC-specific signaling pathways to regulate differentiation and inflammation.


Asunto(s)
Dermatitis por Contacto/inmunología , Dermatitis por Contacto/fisiopatología , Epidermis/inmunología , Epidermis/patología , Proteínas Quinasas , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Carcinógenos/toxicidad , Diferenciación Celular/fisiología , Dermatitis por Contacto/patología , Femenino , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Queratina-14/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Regiones Promotoras Genéticas/fisiología , Proteína Quinasa C/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/inmunología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/inmunología , Acetato de Tetradecanoilforbol/toxicidad
11.
Apoptosis ; 12(8): 1465-78, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17440816

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) selectively induces apoptosis in transformed cells. Normal cells and certain tumor cells can evade Apo2L/TRAIL induced cell death, but the determinants of Apo2L/TRAIL sensitivity are poorly understood. To better understand the factors that contribute to Apo2L/TRAIL resistance, we characterized two colon carcinoma lines with pronounced differences in Apo2L/TRAIL sensitivity. Colo205 cells are highly sensitive to Apo2L/TRAIL whereas Colo320 cells are unresponsive. Components of the DISC (death inducing signaling complex) could be immunoprecipitated from both cell lines in response to Apo2L/TRAIL. Sensitizing agents including a proteasome inhibitor conferred Apo2L/TRAIL sensitivity in Colo320 cells, indicating that the apoptotic machinery was intact and functional. We specifically suppressed the expression of Bcl-2, FLIP or XIAP in Colo320 cells. Downregulation of either FLIP or XIAP but not Bcl-2 restored sensitivity of Colo320 cells to Apo2L/TRAIL. Moreover, stable knockdown of XIAP expression in Colo320 subcutaneous tumors resulted in suppression of tumor growth and sensitivity to Apo2L/TRAIL in vivo. Our results indicate that only a specific subset of anti-apoptotic proteins can confer resistance to Apo2L/TRAIL in Colo320 cells. Elucidation of the factors that contribute to Apo2L/TRAIL resistance in tumor cells may provide insight into combination therapies with Apo2L/TRAIL in a clinical setting.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Carcinoma/genética , Neoplasias del Colon/genética , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Animales , Apoptosis/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Carcinoma/patología , Neoplasias del Colon/patología , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones SCID , Receptores de Muerte Celular/metabolismo , Células Tumorales Cultivadas , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Mol Immunol ; 43(10): 1694-9, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16256200

RESUMEN

Protein kinase C-associated kinase (PKK; DIK/RIP4) is an ankyrin-repeat containing serine/threonine receptor-interacting protein (RIP)-family kinase that can activate NFkappaB, and is required for keratinocyte development. In earlier studies, the expression of a catalytically inactive mutant of PKK in the B cell lineage resulted in a marked decrease in peripheral B cells in the spleen and a severe reduction of B-1 B cells. Here we explore the consequences of a null mutation in PKK with respect to the generation of peripheral B cell lineages and the activation of NFkappaB. We show that PKK is not required for the production of B cells in the bone marrow or for the development and maintenance of all mature B lymphocyte populations. We also show that PKK is not required for the activation of NFkappaB downstream of the BCR, CD40, or TLR-4 in B cells. Taken together, these data demonstrate that the loss of this RIP-family kinase does not compromise B lymphocyte development and maintenance, but leaves open the possibility that PKK may have a redundant role in these processes.


Asunto(s)
Linfocitos B/citología , Linfocitos B/enzimología , Proteínas Quinasas/fisiología , Animales , Médula Ósea/inmunología , Antígenos CD40/metabolismo , Linaje de la Célula , Activación Enzimática , Ratones , Ratones Mutantes , Mutación , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Receptor Toll-Like 4/metabolismo , Quinasa de Factor Nuclear kappa B
13.
J Biol Chem ; 277(18): 16229-40, 2002 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-11864968

RESUMEN

We describe the isolation, cloning, and characterization of human Nek8, a new mammalian NIMA-related kinase, and its candidate substrate Bicd2. Nek8 was isolated as a beta-casein kinase activity in rabbit lung and has an N-terminal catalytic domain homologous to the Nek family of protein kinases. Nek8 also contains a central domain with homology to RCC1, a guanine nucleotide exchange factor for the GTPase Ran, and a C-terminal coiled-coil domain. Like Nek2, Nek8 prefers beta-casein over other exogenous substrates, has shared biochemical requirements for kinase activity, and is capable of autophosphorylation and oligomerization. Nek8 activity is not cell cycle regulated, but like Nek3, levels are consistently higher in G(0)-arrested cells. During the purification of Nek8 a second protein co-chromatographed with Nek8 activity. This protein, Bicd2, is a human homolog of the Drosophila protein Bicaudal D, a coiled-coil protein. Bicd2 is phosphorylated by Nek8 in vitro, and the endogenous proteins associate in vivo. Bicd2 localizes to cytoskeletal structures, and its subcellular localization is dependent on microtubule morphology. Treatment of cells with nocodazole leads to dramatic reorganization of Bicd2, and correlates with Nek8 phosphorylation. This may be indicative of a role for Nek8 and Bicd2 associated with cell cycle independent microtubule dynamics.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Proteínas de la Membrana/metabolismo , Proteínas Nucleares , Proteínas Quinasas , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Dominio Catalítico , Ciclo Celular , Clonación Molecular , Drosophila/enzimología , Citometría de Flujo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Pulmón/enzimología , Proteínas de la Membrana/química , Proteínas Asociadas a Microtúbulos , Datos de Secuencia Molecular , Quinasa 1 Relacionada con NIMA , Quinasas Relacionadas con NIMA , Fragmentos de Péptidos/química , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/aislamiento & purificación , Conejos , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA