Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Lancet Oncol ; 18(1): 42-51, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27932068

RESUMEN

BACKGROUND: Rovalpituzumab tesirine is a first-in-class antibody-drug conjugate directed against delta-like protein 3 (DLL3), a novel target identified in tumour-initiating cells and expressed in more than 80% of patients with small-cell lung cancer. We aimed to assess the safety and activity of rovalpituzumab tesirine in patients who progressed after one or more previous regimen. METHODS: We conducted a phase 1 open-label study at ten cancer centres in the USA. Eligible patients were aged 18 years or older and had histologically or cytologically confirmed small-cell lung cancer or large-cell neuroendocrine tumours with progressive measurable disease (according to Response Evaluation Criteria in Solid Tumors [RECIST], version 1.1) previously treated with one or two chemotherapeutic regimens, including a platinum-based regimen. We assigned patients to dose-escalation or expansion cohorts, ranging from 0·05 mg/kg to 0·8 mg/kg rovalpituzumab tesirine intravenously every 3 weeks or every 6 weeks, followed by investigation of the dose schedules 0·3 mg/kg and 0·4 mg/kg every 6 weeks and 0·2 mg/kg every 3 weeks. Primary objectives were to assess the safety of rovalpituzumab tesirine, including the maximum tolerated dose and dose-limiting toxic effects. The primary activity endpoint was objective response by intention-to-treat analysis. This study is registered with ClinicalTrials.gov, number NCT01901653. The study is closed to enrolment; this report focuses on the cohort with small-cell lung cancer. FINDINGS: Between July 22, 2013, and Aug 10, 2015, 82 patients were enrolled, including 74 patients with small-cell lung cancer and eight with large-cell neuroendocrine carcinoma, all of whom received at least one dose of rovalpituzumab tesirine. Dose-limiting toxic effects of rovalpituzumab tesirine occurred at a dose of 0·8 mg/kg every 3 weeks, including grade 4 thrombocytopenia (in two of two patients at that dose level) and grade 4 liver function test abnormalities (in one patient). The most frequent grade 3 or worse treatment-related adverse events in 74 patients with small-cell lung cancer were thrombocytopenia (eight [11%]), pleural effusion (six [8%]), and increased lipase (five [7%]). Drug-related serious adverse events occurred in 28 (38%) of 74 patients. The maximum tolerated dose of rovalpituzumab tesirine was 0·4 mg/kg every 3 weeks; the recommended phase 2 dose and schedule is 0·3 mg/kg every 6 weeks. At active doses of rovalpituzumab tesirine (0·2 mg/kg or 0·4 mg/kg every 3 weeks or 0·3 mg/kg or 0·4 mg/kg every 6 weeks), 11 (18%) of 60 assessable patients had a confirmed objective response. 11 (18%) of 60 assessable patients had a confirmed objective response, including ten (38%) of 26 patients confirmed to have high DLL3 expression (expression in 50% or more of tumour cells). INTERPRETATION: Rovalpituzumab tesirine shows encouraging single-agent antitumour activity with a manageable safety profile. Further development of rovalpituzumab tesirine in DLL3-expressing malignant diseases is warranted. FUNDING: Stemcentrx Inc.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Benzodiazepinonas/uso terapéutico , Carcinoma de Células Grandes/tratamiento farmacológico , Carcinoma Neuroendocrino/tratamiento farmacológico , Inmunoconjugados/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas de la Membrana/inmunología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Anciano , Carcinoma de Células Grandes/inmunología , Carcinoma de Células Grandes/patología , Carcinoma Neuroendocrino/inmunología , Carcinoma Neuroendocrino/patología , Relación Dosis-Respuesta a Droga , Femenino , Estudios de Seguimiento , Humanos , Inmunoconjugados/farmacología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Pronóstico , Carcinoma Pulmonar de Células Pequeñas/inmunología , Carcinoma Pulmonar de Células Pequeñas/patología , Tasa de Supervivencia
2.
J Immunol ; 190(3): 1066-75, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23277488

RESUMEN

CD8(+) T cells undergo rapid expansion during infection with intracellular pathogens, which is followed by swift and massive culling of primed CD8(+) T cells. The mechanisms that govern the massive contraction and maintenance of primed CD8(+) T cells are not clear. We show in this study that the transcription factor, FoxO3a, does not influence Ag presentation and the consequent expansion of CD8(+) T cell response during Listeria monocytogenes infection, but plays a key role in the maintenance of memory CD8(+) T cells. The effector function of primed CD8(+) T cells as revealed by cytokine secretion and CD107a degranulation was not influenced by inactivation of FoxO3a. Interestingly, FoxO3a-deficient CD8(+) T cells displayed reduced expression of proapoptotic molecules BIM and PUMA during the various phases of response, and underwent reduced apoptosis in comparison with wild-type cells. A higher number of memory precursor effector cells and memory subsets was detectable in FoxO3a-deficient mice compared with wild-type mice. Furthermore, FoxO3a-deficient memory CD8(+) T cells upon transfer into normal or RAG1-deficient mice displayed enhanced survival. These results suggest that FoxO3a acts in a cell-intrinsic manner to regulate the survival of primed CD8(+) T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Factores de Transcripción Forkhead/inmunología , Memoria Inmunológica/inmunología , Listeriosis/inmunología , Activación de Linfocitos/inmunología , Subgrupos Linfocitarios/inmunología , Animales , Presentación de Antígeno , Antígenos Bacterianos/inmunología , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Linfocitos T CD8-positivos/metabolismo , Citocinas/sangre , Citotoxicidad Inmunológica , Femenino , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/deficiencia , Proteínas de Homeodominio/genética , Selectina L/biosíntesis , Selectina L/genética , Listeria monocytogenes/inmunología , Listeriosis/sangre , Subgrupos Linfocitarios/metabolismo , Linfocinas/metabolismo , Proteínas de Membrana de los Lisosomas/inmunología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/genética , Ovalbúmina/inmunología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Receptores de Interleucina-7/biosíntesis , Receptores de Interleucina-7/genética , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética
3.
PLoS Pathog ; 8(2): e1002533, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22359505

RESUMEN

CD8 T cell responses have three phases: expansion, contraction, and memory. Dynamic alterations in proliferation and apoptotic rates control CD8 T cell numbers at each phase, which in turn dictate the magnitude of CD8 T cell memory. Identification of signaling pathways that control CD8 T cell memory is incomplete. The PI3K/Akt signaling pathway controls cell growth in many cell types by modulating the activity of FOXO transcription factors. But the role of FOXOs in regulating CD8 T cell memory remains unknown. We show that phosphorylation of Akt, FOXO and mTOR in CD8 T cells occurs in a dynamic fashion in vivo during an acute viral infection. To elucidate the potentially dynamic role for FOXO3 in regulating homeostasis of activated CD8 T cells in lymphoid and non-lymphoid organs, we infected global and T cell-specific FOXO3-deficient mice with Lymphocytic Choriomeningitis Virus (LCMV). We found that FOXO3 deficiency induced a marked increase in the expansion of effector CD8 T cells, preferentially in the spleen, by T cell-intrinsic mechanisms. Mechanistically, the enhanced accumulation of proliferating CD8 T cells in FOXO3-deficient mice was not attributed to an augmented rate of cell division, but instead was linked to a reduction in cellular apoptosis. These data suggested that FOXO3 might inhibit accumulation of growth factor-deprived proliferating CD8 T cells by reducing their viability. By virtue of greater accumulation of memory precursor effector cells during expansion, the numbers of memory CD8 T cells were strikingly increased in the spleens of both global and T cell-specific FOXO3-deficient mice. The augmented CD8 T cell memory was durable, and FOXO3 deficiency did not perturb any of the qualitative attributes of memory T cells. In summary, we have identified FOXO3 as a critical regulator of CD8 T cell memory, and therapeutic modulation of FOXO3 might enhance vaccine-induced protective immunity against intracellular pathogens.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Factores de Transcripción Forkhead/inmunología , Memoria Inmunológica/inmunología , Transducción de Señal/inmunología , Animales , Infecciones por Arenaviridae/inmunología , Western Blotting , Linfocitos T CD8-positivos/metabolismo , Separación Celular , Citometría de Flujo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
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
5.
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
6.
Nat Med ; 11(6): 666-71, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15895074

RESUMEN

In inflammatory arthridities such as rheumatoid arthritis, cognate lymphocytes have long been considered instigators of autoimmunity, but accumulating evidence indicates that innate immune cells such as neutrophils and mast cells are responsible for a vast majority of acute and ongoing inflammation; however, the molecular mechanisms that govern them remain largely unknown. Here we show that such inflammation requires the forkhead transcription factor Foxo3a: Foxo3a-deficient mice are resistant to two models of neutrophilic inflammation, immune complex-mediated inflammatory arthritis and thioglycollate-induced peritonitis. This reflects a need for Foxo3a to maintain neutrophil vitality during inflammation by suppressing Fas ligand; because Foxo3a can bind and suppress the Fasl promoter, Foxo3a-deficient neutrophils upregulate Fas ligand and undergo apoptosis in response to TNF-alpha and IL-1, and Fas ligand blockade renders Foxo3a-deficient mice susceptible to both arthritis and peritonitis. Thus, Foxo3a ensures neutrophil survival during inflammation, identifying Foxo3a as therapeutic target in inflammation.


Asunto(s)
Apoptosis/fisiología , Artritis/inmunología , Glicoproteínas de Membrana/fisiología , Neutrófilos/fisiología , Factores de Transcripción/fisiología , Animales , Proteína Ligando Fas , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead , Inflamación/inmunología , Ratones , Ratones Noqueados , Factores de Tiempo , Factores de Transcripción/genética
7.
Nat Commun ; 13(1): 1790, 2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35379805

RESUMEN

Despite the recent clinical success of T cell checkpoint inhibition targeting the CTLA-4 and PD-1 pathways, many patients either fail to achieve objective responses or they develop resistance to therapy. In some cases, poor responses to checkpoint blockade have been linked to suboptimal CD28 costimulation and the inability to generate and maintain a productive adaptive anti-tumor immune response. To address this, here we utilize directed evolution to engineer a CD80 IgV domain with increased PD-L1 affinity and fuse this to an immunoglobulin Fc domain, creating a therapeutic (ALPN-202, davoceticept) capable of providing CD28 costimulation in a PD-L1-dependent fashion while also antagonizing PD-1 - PD-L1 and CTLA-4-CD80/CD86 interactions. We demonstrate that by combining CD28 costimulation and dual checkpoint inhibition, ALPN-202 enhances T cell activation and anti-tumor efficacy in cell-based assays and mouse tumor models more potently than checkpoint blockade alone and thus has the potential to generate potent, clinically meaningful anti-tumor immunity in humans.


Asunto(s)
Antígenos CD28 , Neoplasias , Animales , Antígeno B7-1/metabolismo , Antígenos CD28/metabolismo , Humanos , Activación de Linfocitos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Linfocitos T
8.
Eur J Immunol ; 40(7): 1890-6, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20449867

RESUMEN

A fundamental component of signaling initiated by the BCR and CD19 is the activation of phosphoinositide 3-kinase. Downstream of phosphoinositide 3-kinase, the protein kinase AKT phosphorylates several substrates, including members of the forkhead box subgroup O (Foxo) transcription factor family. Among the Foxo proteins, Foxo1 has unique functions in bone marrow B-cell development and peripheral B-cell function. Here, we report a previously unrecognized role for Foxo1 in controlling the ratio of mature B-cell subsets in the spleen. Conditional deletion of Foxo1 in B cells resulted in an increased percentage of marginal zone B cells and a decrease in follicular (FO) B cells. In addition, Foxo1 deficiency corrected the absence of marginal zone B cells that occurs in CD19-deficient mice. These findings show that Foxo1 regulates the balance of mature B-cell subsets and is required for the marginal zone B-cell deficiency phenotype of mice lacking CD19.


Asunto(s)
Antígenos CD19/metabolismo , Linfocitos B/metabolismo , Factores de Transcripción Forkhead/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Bazo/inmunología , Animales , Antígenos CD19/genética , Antígenos CD19/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Diferenciación Celular , Proliferación Celular , Separación Celular , Células Cultivadas , Citometría de Flujo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Regulación del Desarrollo de la Expresión Génica/inmunología , Cambio de Clase de Inmunoglobulina/genética , Ratones , Ratones Noqueados , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/patología , Bazo/embriología , Bazo/patología
9.
Clin Transl Sci ; 14(4): 1314-1326, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33503289

RESUMEN

ALPN-101 (ICOSL vIgD-Fc) is an Fc fusion protein of a human inducible T cell costimulatory ligand (ICOSL) variant immunoglobulin domain (vIgD) designed to inhibit the cluster of differentiation 28 (CD28) and inducible T cell costimulator (ICOS) pathways simultaneously. A first-in-human study evaluated the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of ALPN-101 in healthy adult subjects. ALPN-101 was generally well-tolerated with no evidence of cytokine release, clinically significant immunogenicity, or severe adverse events following single subcutaneous (SC) doses up to 3 mg/kg or single intravenous (IV) doses up to 10 mg/kg or up to 4 weekly IV doses of up to 1 mg/kg. ALPN-101 exhibited a dose-dependent increase in exposure with an estimated terminal half-life of 4.3-8.6 days and SC bioavailability of 60.6% at 3 mg/kg. Minimal to modest accumulation in exposure was observed with repeated IV dosing. ALPN-101 resulted in a dose-dependent increase in maximum target saturation and duration of high-level target saturation. Consistent with its mechanism of action, ALPN-101 inhibited cytokine production in whole blood stimulated by Staphylococcus aureus enterotoxin B ex vivo, as well as antibody responses to keyhole limpet hemocyanin immunization, reflecting immunomodulatory effects upon T cell and T-dependent B cell responses, respectively. In conclusion, ALPN-101 was well-tolerated in healthy subjects with dose-dependent PK and PD consistent with the known biology of the CD28 and ICOS costimulatory pathways. Further clinical development of ALPN-101 in inflammatory and/or autoimmune diseases is therefore warranted.


Asunto(s)
Antígenos CD28 , Inmunosupresores , Proteína Coestimuladora de Linfocitos T Inducibles , Adolescente , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Administración Intravenosa , Antígenos CD28/antagonistas & inhibidores , Antígenos CD28/metabolismo , Voluntarios Sanos , Inmunosupresores/administración & dosificación , Inmunosupresores/efectos adversos , Inmunosupresores/farmacocinética , Proteína Coestimuladora de Linfocitos T Inducibles/antagonistas & inhibidores , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo
10.
J Exp Med ; 200(1): 115-22, 2004 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-15226356

RESUMEN

B cell terminal differentiation involves development into an antibody-secreting plasma cell, reflecting the concerted activation of proplasma cell transcriptional regulators, such as Blimp-1, IRF-4, and Xbp-1. Here, we show that the microphthalmia-associated transcription factor (Mitf) is highly expressed in naive B cells, where it antagonizes the process of terminal differentiation through the repression of IRF-4. Defective Mitf activity results in spontaneous B cell activation, antibody secretion, and autoantibody production. Conversely, ectopic Mitf expression suppresses the expression of IRF-4, the plasma cell marker CD138, and antibody secretion. Thus, Mitf regulates B cell homeostasis by suppressing the antibody-secreting fate.


Asunto(s)
Linfocitos B/fisiología , Diferenciación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Células Plasmáticas/fisiología , Factores de Transcripción/metabolismo , Animales , Anticuerpos/metabolismo , Linfocitos B/inmunología , Proteínas de Unión al ADN/genética , Homeostasis , Factores Reguladores del Interferón , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción Asociado a Microftalmía , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética
11.
J Exp Med ; 200(2): 261-6, 2004 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-15263031

RESUMEN

The signaling lymphocytic activation molecule (SLAM)/CD150 family includes a family of chromosome 1-encoded cell surface molecules with costimulatory functions mediated in part by the adaptor protein SH2D1A (SLAM-associated protein, SAP). Deficiency in SH2D1A protects mice from an experimental model of lupus, including the development of hypergammaglobulinemia, autoantibodies including anti-double stranded DNA, and renal disease. This protection did not reflect grossly defective T or B cell function per se because SH2D1A-deficient mice were susceptible to experimental autoimmune encephalomyelitis, a T cell-dependent disease, and they were capable of mounting normal T-independent antigen-specific immunoglobulin responses. Instead, T-dependent antibody responses were impaired in SH2D1A-deficient mice, reflecting defective germinal center formation. These findings demonstrate a specific role for the SLAM-SH2D1A system in the regulation of T-dependent humoral immune responses, implicating members of the CD150-SH2D1A family as targets in the pathogenesis and therapy of antibody-mediated autoimmune and allergic diseases.


Asunto(s)
Proteínas Portadoras/fisiología , Péptidos y Proteínas de Señalización Intracelular , Linfocitos T/inmunología , Animales , Formación de Anticuerpos , Antígenos CD , Enfermedades Autoinmunes/inmunología , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Predisposición Genética a la Enfermedad , Glicoproteínas/biosíntesis , Inmunoglobulinas/biosíntesis , Lupus Vulgar/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Superficie Celular , Transducción de Señal , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Factores de Tiempo
12.
Eur J Immunol ; 39(11): 2991-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19658095

RESUMEN

Forkhead transcription factors play critical roles in leukocyte homeostasis. To study further the immunological functions of Foxo1, we generated mice that selectively lack Foxo1 in T cells (Foxo1(flox/flox) Lck.cre(+)conditional knockout mice (cKO)). Although thymocyte development appeared relatively normal, Foxo1 cKO mice harbored significantly increased percentages of mature single positive T cells in the thymus as compared with WT mice, yet possessed smaller lymph nodes and spleens that contained fewer T cells. Foxo1 cKO T cells were not more prone to apoptosis, but instead were characterized by a CD62L(lo) CCR7(lo) CD44(hi) surface phenotype, a poorly populated lymphoid compartment in the periphery, and were relatively refractory to TCR stimulation, all of which were associated with reduced expression of Sell, Klf2, Ccr7, and S1pr1. Thus, Foxo1 is critical for naïve T cells to populate the peripheral lymphoid organs by coordinating a molecular program that maintains homeostasis and regulates trafficking.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Factores de Transcripción Forkhead/inmunología , Tejido Linfoide/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Apoptosis/inmunología , Proliferación Celular , Citometría de Flujo , Proteína Forkhead Box O1 , Tejido Linfoide/citología , Ratones , Ratones Noqueados , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/citología , Linfocitos T/citología
13.
J Immunol ; 181(4): 2732-40, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18684964

RESUMEN

Complement activation is tightly regulated to avoid excessive inflammatory and immune responses. Crry(-/-) is an embryonic lethal phenotype secondary to the maternal complement alternative pathway (AP) attacking a placenta deficient in this inhibitor. In this study, we demonstrate that Crry(-/-) mice could be rescued on a partial as well as on a complete factor B (fB)- or C3-deficient maternal background. The C3 and fB protein concentrations in Crry(-/-)C3(+/-) and Crry(-/-)fB(+/-) mice were substantially reduced for gene dosage secondary to enhanced AP turnover. Based on these observations, a breeding strategy featuring reduced maternal AP-activating capacity rescued the lethal phenotype. It led to a novel, stable line of Crry SKO mice carrying normal alleles for C3 and fB. Crry SKO mice also had accelerated C3 and fB turnover and therefore reduced AP- activating potential. These instructive results represent an example of a membrane regulatory protein being responsible for homeostasis of the complement system. They imply that there is constant turnover on cells of the AP pathway which functions as an immune surveillance system for pathogens and altered self.


Asunto(s)
Vía Alternativa del Complemento/inmunología , Homeostasis/inmunología , Proteínas de la Membrana/fisiología , Receptores de Complemento/fisiología , Animales , Línea Celular , Complemento C3/biosíntesis , Complemento C3/deficiencia , Complemento C3/metabolismo , Factor B del Complemento/biosíntesis , Factor B del Complemento/deficiencia , Factor B del Complemento/genética , Vía Alternativa del Complemento/genética , Pérdida del Embrión/genética , Pérdida del Embrión/inmunología , Femenino , Genotipo , Homeostasis/genética , Humanos , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Receptores de Complemento 3b
14.
Sci Transl Med ; 12(564)2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-33028709

RESUMEN

Acute graft-versus-host disease (aGVHD) remains a major complication of allogeneic hematopoietic cell transplantation (HCT). CD146 and CCR5 are proteins that mark activated T helper 17 (Th17) cells. The Th17 cell phenotype is promoted by the interaction of the receptor ICOS on T cells with ICOS ligand (ICOSL) on dendritic cells (DCs). We performed multiparametric flow cytometry in a cohort of 156 HCT recipients and conducted experiments with aGVHD murine models to understand the role of ICOSL+ DCs. We observed an increased frequency of ICOSL+ plasmacytoid DCs, correlating with CD146+CCR5+ T cell frequencies, in the 64 HCT recipients with gastrointestinal aGVHD. In murine models, donor bone marrow cells from ICOSL-deficient mice compared to those from wild-type mice reduced aGVHD-related mortality. Reduced aGVHD resulted from lower intestinal infiltration of pDCs and pathogenic Th17 cells. We transplanted activated human ICOSL+ pDCs along with human peripheral blood mononuclear cells into immunocompromised mice and observed infiltration of intestinal CD146+CCR5+ T cells. We found that prophylactic administration of a dual human ICOS/CD28 antagonist (ALPN-101) prevented aGVHD in this model better than did the clinically approved belatacept (CTLA-4-Fc), which binds CD80 (B7-1) and CD86 (B7-2) and interferes with the CD28 T cell costimulatory pathway. When started at onset of aGVHD signs, ALPN-101 treatment alleviated symptoms of ongoing aGVHD and improved survival while preserving antitumoral cytotoxicity. Our data identified ICOSL+-pDCs as an aGVHD biomarker and suggest that coinhibition of the ICOSL/ICOS and B7/CD28 axes with one biologic drug may represent a therapeutic opportunity to prevent or treat aGVHD.


Asunto(s)
Antígenos CD28 , Enfermedad Injerto contra Huésped , Abatacept , Animales , Células Dendríticas , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Proteína Coestimuladora de Linfocitos T Inducibles , Leucocitos Mononucleares , Ratones
15.
Front Immunol ; 10: 3086, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32038630

RESUMEN

Immunoglobulin superfamily member (IgSF) proteins play a significant role in regulating immune responses with surface expression on all immune cell subsets, making the IgSF an attractive family of proteins for therapeutic targeting in human diseases. We have developed a directed evolution platform capable of engineering IgSF domains to increase affinities for cognate ligands and/or introduce binding to non-cognate ligands. Using this scientific platform, ICOSL domains have been derived with enhanced binding to ICOS and with additional high-affinity binding to the non-cognate receptor, CD28. Fc-fusion proteins containing these engineered ICOSL domains significantly attenuate T cell activation in vitro and in vivo and can inhibit development of inflammatory diseases in mouse models. We also present evidence that engineered ICOSL domains can be formatted to selectively provide costimulatory signals to augment T cell responses. Our scientific platform thus provides a system for developing therapeutic protein candidates with selective biological impact for treatments of a wide array of human disorders including cancer and autoimmune/inflammatory diseases.


Asunto(s)
Inmunoglobulinas/química , Inmunoglobulinas/genética , Familia de Multigenes , Animales , Antígenos CD28/genética , Antígenos CD28/inmunología , Evolución Molecular Dirigida , Femenino , Humanos , Inmunoglobulinas/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Dominios Proteicos , Linfocitos T/inmunología
16.
Front Biosci ; 13: 4218-40, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508507

RESUMEN

Studies on the pathogenesis of autoimmune diseases have begun to delve into the molecular and cellular mechanisms, and transcription factors, as key regulators of immune effector cell development and function, have received growing attention. Their involvement has been investigated in immune cells, such as T and B lymphocytes, macrophages and neutrophils, but also end-organ tissues, such as synoviocytes, keratinocytes and epithelial cells, and has revealed particularly dominant roles for NF-kappaB, STAT and AP-1 family members. This review summarizes recent findings and current knowledge regarding the roles of transcription factors in autoimmunity, focusing on their role in pathogenesis, as evidenced by both biological and genetic studies, as well as the implications of these findings for anti-inflammatory therapies.


Asunto(s)
Enfermedades Autoinmunes/fisiopatología , Factores de Transcripción/fisiología , Artritis/genética , Artritis/fisiopatología , Enfermedades Autoinmunes/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Inflamación/genética , Inflamación/fisiopatología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/fisiopatología , Esclerosis Múltiple/genética , Esclerosis Múltiple/fisiopatología , Proteínas Proto-Oncogénicas c-ets/fisiología , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/fisiología , Transcripción Genética
17.
J Pharmacol Exp Ther ; 327(3): 610-9, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18776065

RESUMEN

P38alpha is a protein kinase that regulates the expression of inflammatory cytokines, suggesting a role in the pathogenesis of diseases such as rheumatoid arthritis (RA) or systemic lupus erythematosus. Here, we describe the preclinical pharmacology of pamapimod, a novel p38 mitogen-activated protein kinase inhibitor. Pamapimod inhibited p38alpha and p38beta enzymatic activity, with IC(50) values of 0.014 +/- 0.002 and 0.48 +/- 0.04 microM, respectively. There was no activity against p38delta or p38gamma isoforms. When profiled across 350 kinases, pamapimod bound only to four kinases in addition to p38. Cellular potency was assessed using phosphorylation of heat shock protein-27 and c-Jun as selective readouts for p38 and c-Jun NH(2)-terminal kinase (JNK), respectively. Pamapimod inhibited p38 (IC(50), 0.06 microM), but inhibition of JNK was not detected. Pamapimod also inhibited lipopolysaccharide (LPS)-stimulated tumor necrosis factor (TNF) alpha production by monocytes, interleukin (IL)-1beta production in human whole blood, and spontaneous TNFalpha production by synovial explants from RA patients. LPS- and TNFalpha-stimulated production of TNFalpha and IL-6 in rodents also was inhibited by pamapimod. In murine collagen-induced arthritis, pamapimod reduced clinical signs of inflammation and bone loss at 50 mg/kg or greater. In a rat model of hyperalgesia, pamapimod increased tolerance to pressure in a dose-dependent manner, suggesting an important role of p38 in pain associated with inflammation. Finally, an analog of pamapimod that has equivalent potency and selectivity inhibited renal disease in lupus-prone MRL/lpr mice. Our study demonstrates that pamapimod is a potent, selective inhibitor of p38alpha with the ability to inhibit the signs and symptoms of RA and other autoimmune diseases.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridonas/farmacología , Pirimidinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Artritis Reumatoide/tratamiento farmacológico , Evaluación Preclínica de Medicamentos , Humanos , Inflamación/tratamiento farmacológico , Concentración 50 Inhibidora , Interleucina-1beta/antagonistas & inhibidores , Interleucina-6/antagonistas & inhibidores , Enfermedades Renales/prevención & control , Ratones , Monocitos/inmunología , Monocitos/metabolismo , Osteoporosis/prevención & control , Isoformas de Proteínas , Piridonas/uso terapéutico , Pirimidinas/uso terapéutico , Líquido Sinovial/inmunología , Líquido Sinovial/metabolismo , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
18.
Curr Opin Immunol ; 17(3): 230-6, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15886111

RESUMEN

Toll-like receptors (TLRs) and their ligands have emerged as important regulators of immunity, relevant to a wide range of effector responses from vaccination to autoimmunity. The most well-studied ligands of TLRs expressed on B cells include the lipopolysaccharides (for TLR4) and CpG-containing DNAs (for TLR9), which induce and/or co-stimulate B cells to undergo proliferation, class switching and differentiation into antibody-secreting cells. Recent developments in this area include advancements into our understanding of the role of these receptor pathways in B cells, and in particular the relevance of TLR9, which has received substantial attention as both a Th1-like inflammatory immunomodulator and a pathogenic co-stimulator of autoreactive B cell responses.


Asunto(s)
Linfocitos B/inmunología , Glicoproteínas de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Animales , Linfocitos B/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Activación de Linfocitos/inmunología , Ratones , Receptor Toll-Like 4 , Receptor Toll-Like 9 , Receptores Toll-Like
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA