Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 175(7): 1731-1743.e13, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30503213

RESUMEN

Checkpoint inhibitors have revolutionized cancer treatment. However, only a minority of patients respond to these immunotherapies. Here, we report that blocking the inhibitory NKG2A receptor enhances tumor immunity by promoting both natural killer (NK) and CD8+ T cell effector functions in mice and humans. Monalizumab, a humanized anti-NKG2A antibody, enhanced NK cell activity against various tumor cells and rescued CD8+ T cell function in combination with PD-x axis blockade. Monalizumab also stimulated NK cell activity against antibody-coated target cells. Interim results of a phase II trial of monalizumab plus cetuximab in previously treated squamous cell carcinoma of the head and neck showed a 31% objective response rate. Most common adverse events were fatigue (17%), pyrexia (13%), and headache (10%). NKG2A targeting with monalizumab is thus a novel checkpoint inhibitory mechanism promoting anti-tumor immunity by enhancing the activity of both T and NK cells, which may complement first-generation immunotherapies against cancer.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Carcinoma de Células Escamosas , Cetuximab/uso terapéutico , Inmunidad Celular/efectos de los fármacos , Inmunoterapia , Células Asesinas Naturales/inmunología , Subfamília C de Receptores Similares a Lectina de Células NK , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Ensayos Clínicos Fase II como Asunto , Humanos , Células Asesinas Naturales/patología , Ratones , Subfamília C de Receptores Similares a Lectina de Células NK/antagonistas & inhibidores , Subfamília C de Receptores Similares a Lectina de Células NK/inmunología
2.
Annu Rev Immunol ; 28: 367-88, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20192808

RESUMEN

The immune system has evolved to respond not only to pathogens, but also to signals released from dying cells. Cell death through necrosis induces inflammation, whereas apoptotic cell death provides an important signal for tolerance induction. High mobility group box 1 (HMGB1) is a DNA-binding nuclear protein, released actively following cytokine stimulation as well as passively during cell death; it is the prototypic damage-associated molecular pattern (DAMP) molecule and has been implicated in several inflammatory disorders. HMGB1 can associate with other molecules, including TLR ligands and cytokines, and activates cells through the differential engagement of multiple surface receptors including TLR2, TLR4, and RAGE. RAGE is a multiligand receptor that binds structurally diverse molecules, including not only HMGB1, but also S100 family members and amyloid-beta. RAGE activation has been implicated in sterile inflammation as well as in cancer, diabetes, and Alzheimer's disease. While HMGB1 through interactions with TLRs may also be important, this review focuses on the role of the HMGB1-RAGE axis in inflammation and cancer.


Asunto(s)
Proteína HMGB1/inmunología , Inflamación/inmunología , Neoplasias/inmunología , Receptores Inmunológicos/inmunología , Animales , Proteína HMGB1/química , Humanos , Inflamación/metabolismo , Ligandos , Neoplasias/metabolismo , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/metabolismo , Transducción de Señal
3.
Nat Immunol ; 17(2): 196-203, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26692173

RESUMEN

Canonically, immunoglobulin E (IgE) mediates allergic immune responses by triggering mast cells and basophils to release histamine and type 2 helper cytokines. Here we found that in human systemic lupus erythematosus (SLE), IgE antibodies specific for double-stranded DNA (dsDNA) activated plasmacytoid dendritic cells (pDCs), a type of cell of the immune system linked to viral defense, which led to the secretion of substantial amounts of interferon-α (IFN-α). The concentration of dsDNA-specific IgE found in patient serum correlated with disease severity and greatly potentiated pDC function by triggering phagocytosis via the high-affinity FcɛRI receptor for IgE, followed by Toll-like receptor 9 (TLR9)-mediated sensing of DNA in phagosomes. Our findings expand the known pathogenic mechanisms of IgE-mediated inflammation beyond those found in allergy and demonstrate that IgE can trigger interferon responses capable of exacerbating self-destructive autoimmune responses.


Asunto(s)
Autoanticuerpos/inmunología , Autoinmunidad , Inmunoglobulina E/inmunología , Interferones/metabolismo , Anticuerpos Antinucleares/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Citocinas/sangre , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Masculino , Fagocitosis/inmunología , Fagosomas/metabolismo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Receptor Toll-Like 9/metabolismo
4.
Immunity ; 46(1): 106-119, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28099860

RESUMEN

A hallmark of autoimmunity in murine models of lupus is the formation of germinal centers (GCs) in lymphoid tissues where self-reactive B cells expand and differentiate. In the host response to foreign antigens, follicular dendritic cells (FDCs) maintain GCs through the uptake and cycling of complement-opsonized immune complexes. Here, we examined whether FDCs retain self-antigens and the impact of this process in autoantibody secretion in lupus. We found that FDCs took up and retained self-immune complexes composed of ribonucleotide proteins, autoantibody, and complement. This uptake, mediated through CD21, triggered endosomal TLR7 and led to the secretion of interferon (IFN) α via an IRF5-dependent pathway. Blocking of FDC secretion of IFN-α restored B cell tolerance and reduced the amount of GCs and pathogenic autoantibody. Thus, FDCs are a critical source of the IFN-α driving autoimmunity in this lupus model. This pathway is conserved in humans, suggesting that it may be a viable therapeutic target in systemic lupus erythematosus.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos B/inmunología , Células Dendríticas Foliculares/inmunología , Lupus Eritematoso Sistémico/inmunología , Animales , Autoantígenos/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Interferón-alfa/biosíntesis , Interferón-alfa/inmunología , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Reacción en Cadena de la Polimerasa , Receptor Toll-Like 7/inmunología , Transcriptoma
5.
Nature ; 578(7793): 177, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32025017

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
Nature ; 546(7659): 539-543, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28614301

RESUMEN

Systemic lupus erythematosus (SLE) is an incurable autoimmune disease characterized by autoantibody deposition in tissues such as kidney, skin and lungs. Notably, up to 75% of patients with SLE experience neuropsychiatric symptoms that range from anxiety, depression and cognitive impairment to seizures and, in rare cases, psychosis-collectively this is referred to as central nervous system (CNS) lupus. In some cases, certain autoantibodies, such as anti-NMDAR or anti-phospholipid antibodies, promote CNS lupus. However, in most patients, the mechanisms that underlie these symptoms are unknown. CNS lupus typically presents at lupus diagnosis or within the first year, suggesting that early factors contributing to peripheral autoimmunity may promote CNS lupus symptoms. Here we report behavioural phenotypes and synapse loss in lupus-prone mice that are prevented by blocking type I interferon (IFN) signalling. Furthermore, we show that type I IFN stimulates microglia to become reactive and engulf neuronal and synaptic material in lupus-prone mice. These findings and our observation of increased type I IFN signalling in post-mortem hippocampal brain sections from patients with SLE may instruct the evaluation of ongoing clinical trials of anifrolumab, a type I IFN-receptor antagonist. Moreover, identification of IFN-driven microglia-dependent synapse loss, along with microglia transcriptome data, connects CNS lupus with other CNS diseases and provides an explanation for the neurological symptoms observed in some patients with SLE.


Asunto(s)
Interferón Tipo I/inmunología , Vasculitis por Lupus del Sistema Nervioso Central/inmunología , Vasculitis por Lupus del Sistema Nervioso Central/patología , Microglía/inmunología , Microglía/patología , Sinapsis/patología , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Conducta Animal , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Interferón Tipo I/antagonistas & inhibidores , Vasculitis por Lupus del Sistema Nervioso Central/psicología , Masculino , Ratones , Microglía/metabolismo , Fenotipo , Transducción de Señal , Sinapsis/inmunología , Transcriptoma
7.
J Immunol ; 205(1): 102-112, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32434940

RESUMEN

To maintain homeostasis, macrophages must be capable of assuming either an inflammatory or an anti-inflammatory phenotype. To better understand the latter, we stimulated human macrophages in vitro with TLR ligands in the presence of high-density immune complexes (IC). This combination of stimuli resulted in a broad suppression of inflammatory mediators and an upregulation of molecules involved in tissue remodeling and angiogenesis. Transcriptomic analysis of TLR stimulation in the presence of IC predicted the downstream activation of AKT and the inhibition of GSK3. Consequently, we pretreated LPS-stimulated human macrophages with small molecule inhibitors of GSK3 to partially phenocopy the regulatory effects of stimulation in the presence of IC. The upregulation of DC-STAMP and matrix metalloproteases was observed on these cells and may represent potential biomarkers for this regulatory activation state. To demonstrate the presence of these anti-inflammatory, growth-promoting macrophages in a human infectious disease, biopsies from patients with leprosy (Hanseniasis) were analyzed. The lepromatous form of this disease is characterized by hypergammaglobulinemia and defective cell-mediated immunity. Lesions in lepromatous leprosy contained macrophages with a regulatory phenotype expressing higher levels of DC-STAMP and lower levels of IL-12, relative to macrophages in tuberculoid leprosy lesions. Therefore, we propose that increased signaling by FcγR cross-linking on TLR-stimulated macrophages can paradoxically promote the resolution of inflammation and initiate processes critical to tissue growth and repair. It can also contribute to infectious disease progression.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Lepra Lepromatosa/inmunología , Lepra Tuberculoide/inmunología , Macrófagos/inmunología , Biopsia , Diferenciación Celular/inmunología , Línea Celular , Progresión de la Enfermedad , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Lepra Lepromatosa/patología , Lepra Tuberculoide/patología , Activación de Macrófagos , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Neovascularización Fisiológica/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , RNA-Seq , Receptores de IgG/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Piel/citología , Piel/inmunología , Piel/patología , Receptores Toll-Like/metabolismo , Adulto Joven
8.
Ann Rheum Dis ; 78(2): 228-237, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30459279

RESUMEN

OBJECTIVE: Immune complexes (ICs) play a critical role in the pathology of autoimmune diseases. The aim of this study was to generate and characterise a first-in-class anti-FcγRIIA antibody (Ab) VIB9600 (previously known as MEDI9600) that blocks IgG immune complex-mediated cellular activation for clinical development. METHODS: VIB9600 was humanised and optimised from the IV.3 Ab. Binding affinity and specificity were determined by Biacore and ELISA. Confocal microscopy, Flow Cytometry-based assays and binding competition assays were used to assess the mode of action of the antibody. In vitro cell-based assays were used to demonstrate suppression of IC-mediated inflammatory responses. In vivo target suppression and efficacy was demonstrated in FcγRIIA-transgenic mice. Single-dose pharmacokinetic (PK)/pharmacodynamic study multiple dose Good Laboratory Practice (GLP) toxicity studies were conducted in non-human primates. RESULTS: We generated a humanised effector-deficient anti-FcγRIIA antibody (VIB9600) that potently blocks autoantibody and IC-mediated proinflammatory responses. VIB9600 suppresses FcγRIIA activation by blocking ligand engagement and by internalising FcγRIIA from the cell surface. VIB9600 inhibits IC-induced type I interferons from plasmacytoid dendritic cells (involved in SLE), antineutrophil cytoplasmic antibody (ANCA)-induced production of reactive oxygen species by neutrophils (involved in ANCA-associated vasculitis) and IC-induced tumour necrosis factor α and interleukin-6 production (involved in rheumatoid arthritis). In FcγRIIA transgenic mice, VIB9600 suppressed antiplatelet antibody-induced thrombocytopaenia, acute anti-GBM Ab-induced nephritis and anticollagen Ab-induced arthritis. VIB9600 also exhibited favourable PK and safety profiles in cynomolgus monkey studies. CONCLUSIONS: VIB9600 is a specific humanised antibody antagonist of FcγRIIA with null effector function that warrants further clinical development for the treatment of IC-mediated diseases.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Complejo Antígeno-Anticuerpo/efectos de los fármacos , Enfermedades Autoinmunes/tratamiento farmacológico , Factores Inmunológicos/farmacología , Receptores de IgG/inmunología , Animales , Anticuerpos Anticitoplasma de Neutrófilos/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Enfermedades Autoinmunes/inmunología , Células Dendríticas/inmunología , Humanos , Inmunoglobulina G/inmunología , Interleucina-6/inmunología , Macaca fascicularis , Ratones , Ratones Transgénicos , Neutrófilos/inmunología , Especies Reactivas de Oxígeno/inmunología , Factor de Necrosis Tumoral alfa/inmunología
9.
J Transl Med ; 17(1): 357, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31684954

RESUMEN

BACKGROUND: The 18-gene tumor inflammation signature (TIS) is a clinical research assay that enriches for clinical benefit to immune checkpoint blockade. We evaluated its ability to predict clinical benefit of immunotherapy in cancer patients treated with PD-1 checkpoint inhibitors in routine clinical care. METHODS: The CERTIM cohort is a prospective cohort which includes patients receiving immune checkpoint inhibitors in Cochin University hospital. RNA extracted from 58 archival formalin fixed paraffin embedded tumor blocks (including 38 lung cancers, 5 melanomas, 10 renal carcinomas, 4 urothelial carcinomas and 1 colon carcinoma) was hybridized to a beta version of the NanoString® PanCancer IO360™ CodeSet using nCounter® technology. Gene expression signatures were correlated with tumor responses (by RECIST criteria) and overall survival. PD-L1 immunostaining on tumor cells was assessed in 37 non-small cell lung cancer (NSCLC) samples and tumor mutational burden (TMB) measured by whole exome sequencing in 19 of these. RESULTS: TIS scores were significantly associated with complete or partial response to anti-PD-1 treatment in the whole cohort (odds ratio = 2.64, 95% CI [1.4; 6.0], p = 0.008), as well as in the NSCLC population (odds ratio = 3.27, 95% CI [1.2; 11.6], p = 0.03). Patients whose tumor had a high TIS score (upper tertile) showed prolonged overall survival compared to patients whose tumor had lower TIS scores, both in the whole cohort (hazard ratio = 0.37, 95% CI [0.18, 0.76], p = 0.005) and in the NSCLC population (hazard ratio = 0.36, 95% CI [0.14, 0.90], p = 0.02). In the latter, the TIS score was independent from either PD-L1 staining on tumor cells (spearman coefficient 0.2) and TMB (spearman coefficient - 0.2). CONCLUSIONS: These results indicate that validated gene expression assay measuring the level of tumor microenvironment inflammation such as TIS, are accurate and independent predictive biomarkers and can be easily implemented in the clinical practice.


Asunto(s)
Inflamación/genética , Inflamación/terapia , Neoplasias/genética , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/terapia , Estudios de Cohortes , Femenino , Humanos , Inmunoterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Mutación , Estudios Prospectivos , Transcriptoma , Investigación Biomédica Traslacional , Resultado del Tratamiento
10.
J Immunol ; 199(9): 3316-3325, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28939759

RESUMEN

Dysfunction of the epithelial barrier is a hallmark of inflammatory intestinal diseases. The intestinal epithelial barrier is maintained by expression of tight junctions that connect adjacent epithelial cells and seal the paracellular space. IL-22 is critical for the maintenance of intestinal barrier function through promoting antipathogen responses and regeneration of epithelial tissues in the gut. However, little is known about the effects of IL-22 on the regulation of tight junctions in the intestinal epithelium. In this study we report that IL-22 signals exclusively through the basolateral side of polarized Caco-2 cell monolayers. IL-22 treatment does not affect the flux of uncharged macromolecules across cell monolayers but significantly reduces transepithelial electrical resistance (TEER), indicating an increase of paracellular permeability for ions. IL-22 treatment on Caco-2 monolayers and on primary human intestinal epithelium markedly induces the expression of Claudin-2, a cation-channel-forming tight junction protein. Furthermore, treatment of IL-22 in mice upregulates Claudin-2 protein in colonic epithelial cells. Knocking down Claudin-2 expression with small interfering RNA reverses the reduction of TEER in IL-22-treated cells. Moreover, IL-22-mediated upregulation of Claudin-2 and loss of TEER can be suppressed with the treatment of JAK inhibitors. In summary, our results reveal that IL-22 increases intestinal epithelial permeability by upregulating Claudin-2 expression through the JAK/STAT pathway. These results provide novel mechanistic insights into the role of IL-22 in the regulation and maintenance of the intestinal epithelial barrier.


Asunto(s)
Claudinas/inmunología , Interleucinas/inmunología , Mucosa Intestinal/inmunología , Transducción de Señal/inmunología , Uniones Estrechas/inmunología , Regulación hacia Arriba/inmunología , Animales , Células CACO-2 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Permeabilidad , Interleucina-22
11.
Am J Respir Crit Care Med ; 198(7): 928-940, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29518341

RESUMEN

RATIONALE: Patients with chronic obstructive pulmonary disease (COPD) have a higher prevalence of lung cancer. The chronic inflammation associated with COPD probably promotes the earliest stages of carcinogenesis. However, once tumors have progressed to malignancy, the impact of COPD on the tumor immune microenvironment remains poorly defined, and its effects on immune-checkpoint blockers' efficacy are still unknown. OBJECTIVES: To study the impact of COPD on the immune contexture of non-small cell lung cancer. METHODS: We performed in-depth immune profiling of lung tumors by immunohistochemistry and we determined its impact on patient survival (n = 435). Tumor-infiltrating T lymphocyte (TIL) exhaustion by flow cytometry (n = 50) was also investigated. The effectiveness of an anti-PD-1 (programmed cell death-1) treatment (nivolumab) was evaluated in 39 patients with advanced-stage non-small cell lung cancer. All data were analyzed according to patient COPD status. MEASUREMENTS AND MAIN RESULTS: Remarkably, COPD severity is positively correlated with the coexpression of PD-1/TIM-3 (T-cell immunoglobulin and mucin domain-containing molecule-3) by CD8 T cells. In agreement, we observed a loss of CD8 T cell-associated favorable clinical outcome in COPD+ patients. Interestingly, a negative prognostic value of PD-L1 (programmed cell death ligand 1) expression by tumor cells was observed only in highly CD8 T cell-infiltrated tumors of COPD+ patients. Finally, data obtained on 39 patients with advanced-stage non-small cell lung cancer treated by an anti-PD-1 antibody showed longer progression-free survival in COPD+ patients, and also that the association between the severity of smoking and the response to nivolumab was preferentially observed in COPD+ patients. CONCLUSIONS: COPD is associated with an increased sensitivity of CD8 tumor-infiltrating T lymphocytes to immune escape mechanisms developed by tumors, thus suggesting a higher sensitivity to PD-1 blockade in patients with COPD.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Neoplasias Pulmonares/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Microambiente Tumoral/inmunología , Anciano , Análisis de Varianza , Biopsia con Aguja , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Estudios de Cohortes , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Enfermedad Pulmonar Obstructiva Crónica/mortalidad , Enfermedad Pulmonar Obstructiva Crónica/patología , Estudios Retrospectivos , Medición de Riesgo , Análisis de Supervivencia
12.
PLoS Genet ; 12(4): e1005895, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27093186

RESUMEN

Small cell lung cancer (SCLC) is an aggressive disease with poor survival. A few sequencing studies performed on limited number of samples have revealed potential disease-driving genes in SCLC, however, much still remains unknown, particularly in the Asian patient population. Here we conducted whole exome sequencing (WES) and transcriptomic sequencing of primary tumors from 99 Chinese SCLC patients. Dysregulation of tumor suppressor genes TP53 and RB1 was observed in 82% and 62% of SCLC patients, respectively, and more than half of the SCLC patients (62%) harbored TP53 and RB1 mutation and/or copy number loss. Additionally, Serine/Arginine Splicing Factor 1 (SRSF1) DNA copy number gain and mRNA over-expression was strongly associated with poor survival using both discovery and validation patient cohorts. Functional studies in vitro and in vivo demonstrate that SRSF1 is important for tumorigenicity of SCLC and may play a key role in DNA repair and chemo-sensitivity. These results strongly support SRSF1 as a prognostic biomarker in SCLC and provide a rationale for personalized therapy in SCLC.


Asunto(s)
Carcinoma de Células Pequeñas/genética , Neoplasias Pulmonares/genética , Proteínas Oncogénicas/genética , Factores de Empalme Serina-Arginina/genética , Adulto , Anciano , Variaciones en el Número de Copia de ADN , Daño del ADN , Femenino , Silenciador del Gen , Humanos , Masculino , Persona de Mediana Edad , Mutación
13.
Ann Neurol ; 82(5): 795-812, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29059494

RESUMEN

OBJECTIVE: Biomarkers aid diagnosis, allow inexpensive screening of therapies, and guide selection of patient-specific therapeutic regimens in most internal medicine disciplines. In contrast, neurology lacks validated measurements of the physiological status, or dysfunction(s) of cells of the central nervous system (CNS). Accordingly, patients with chronic neurological diseases are often treated with a single disease-modifying therapy without understanding patient-specific drivers of disability. Therefore, using multiple sclerosis (MS) as an example of a complex polygenic neurological disease, we sought to determine whether cerebrospinal fluid (CSF) biomarkers are intraindividually stable, cell type-, disease- and/or process-specific, and responsive to therapeutic intervention. METHODS: We used statistical learning in a modeling cohort (n = 225) to develop diagnostic classifiers from DNA-aptamer-based measurements of 1,128 CSF proteins. An independent validation cohort (n = 85) assessed the reliability of derived classifiers. The biological interpretation resulted from in vitro modeling of primary or stem cell-derived human CNS cells and cell lines. RESULTS: The classifier that differentiates MS from CNS diseases that mimic MS clinically, pathophysiologically, and on imaging achieved a validated area under the receiver operating characteristic curve (AUROC) of 0.98, whereas the classifier that differentiates relapsing-remitting from progressive MS achieved a validated AUROC of 0.91. No classifiers could differentiate primary progressive from secondary progressive MS better than random guessing. Treatment-induced changes in biomarkers greatly exceeded intraindividual and technical variabilities of the assay. INTERPRETATION: CNS biological processes reflected by CSF biomarkers are robust, stable, disease specific, or even disease stage specific. This opens opportunities for broad utilization of CSF biomarkers in drug development and precision medicine for CNS disorders. Ann Neurol 2017;82:795-812.


Asunto(s)
Proteínas del Líquido Cefalorraquídeo/metabolismo , Esclerosis Múltiple Crónica Progresiva/líquido cefalorraquídeo , Esclerosis Múltiple Crónica Progresiva/diagnóstico , Esclerosis Múltiple Recurrente-Remitente/líquido cefalorraquídeo , Esclerosis Múltiple Recurrente-Remitente/diagnóstico , Adolescente , Adulto , Anciano , Biomarcadores/líquido cefalorraquídeo , Estudios de Casos y Controles , Línea Celular , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
14.
J Immunol ; 197(2): 491-503, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27296665

RESUMEN

ICOS, a member of the CD28 family, represents a key molecule that regulates adaptive responses to foreign Ags. ICOS is prominently expressed on T follicular helper (TFH) cells, a specialized CD4(+) T cell subset that orchestrates B cell differentiation within the germinal centers and humoral response. However, the contribution of ICOS and TFH cells to autoantibody profiles under pathological conditions has not been thoroughly investigated. We used the Sle1 lupus-prone mouse model to examine the role of ICOS in the expansion and function of pathogenic TFH cells. Genetic deletion of ICOS impacted the expansion of TFH cells in B6.Sle1 mice and inhibited the differentiation of B lymphocytes into plasma cells. The phenotypic changes observed in B6.Sle1-ICOS-knockout mice were also associated with a significant reduction in class-switched IgG, and anti-nucleosomal IgG-secreting B cells compared with B6.Sle1 animals. The level of vascular cell adhesion protein 1, a molecule that was shown to be elevated in patients with SLE and in lupus models, was also increased in an ICOS-dependent manner in Sle1 mice and correlated with autoantibody levels. The elimination of ICOS-expressing CD4(+) T cells in B6.Sle1 mice, using a glyco-engineered anti-ICOS-depleting Ab, resulted in a significant reduction in anti-nucleosomal autoantibodies. Our results indicate that ICOS regulates the ontogeny and homeostasis of B6.Sle1 TFH cells and influences the function of TFH cells during aberrant germinal center B cell responses. Therapies targeting the ICOS signaling pathway may offer new opportunities for the treatment of lupus and other autoimmune diseases.


Asunto(s)
Tolerancia Inmunológica/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Lupus Eritematoso Sistémico/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Ensayo de Immunospot Ligado a Enzimas , Citometría de Flujo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Inmunohistoquímica , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Factores de Transcripción/genética , Factores de Transcripción/inmunología
15.
J Immunol ; 196(4): 1541-9, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26764035

RESUMEN

The contribution of autoantibody-producing plasma cells in multiple sclerosis (MS) remains unclear. Anti-CD20 B cell depletion effectively reduces disease activity in MS patients, but it has a minimal effect on circulating autoantibodies and oligoclonal bands in the cerebrospinal fluid. Recently we reported that MEDI551, an anti-CD19 mAb, therapeutically ameliorates experimental autoimmune encephalomyelitis (EAE), the mouse model of MS. MEDI551 potently inhibits pathogenic adaptive immune responses, including depleting autoantibody-producing plasma cells. In the present study, we demonstrated that CD19 mAb treatment ameliorates EAE more effectively than does CD20 mAb. Myelin oligodendrocyte glycoprotein-specific Abs and short-lived and long-lived autoantibody-secreting cells were nearly undetectable in the CD19 mAb-treated mice, but they remained detectable in the CD20 mAb-treated mice. Interestingly, residual disease severity in the CD20 mAb-treated animals positively correlated with the frequency of treatment-resistant plasma cells in the bone marrow. Of note, treatment-resistant plasma cells contained a substantial proportion of CD19(+)CD20(-) plasma cells, which would have otherwise been targeted by CD19 mAb. These data suggested that CD19(+)CD20(-) plasma cells spared by anti-CD20 therapy likely contribute to residual EAE severity by producing autoreactive Abs. In patients with MS, we also identified a population of CD19(+)CD20(-) B cells in the cerebrospinal fluid that would be resistant to CD20 mAb treatment.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD19/inmunología , Antígenos CD20/inmunología , Linfocitos B/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Células Plasmáticas/inmunología , Adulto , Animales , Anticuerpos Monoclonales/inmunología , Autoanticuerpos/análisis , Líquido Cefalorraquídeo/química , Líquido Cefalorraquídeo/citología , Líquido Cefalorraquídeo/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/fisiopatología , Encefalomielitis Autoinmune Experimental/terapia , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Esclerosis Múltiple/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología
16.
Mol Ther ; 25(8): 1917-1932, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28578991

RESUMEN

Vesicular stomatitis virus encoding the IFNß transgene (VSV-IFNß) is a mediator of potent oncolytic activity and is undergoing clinical evaluation for the treatment of solid tumors. Emerging preclinical and clinical data suggest treatment of tumors with oncolytic viruses may sensitize tumors to checkpoint inhibitors and increase the anti-tumor immune response. New generations of immuno-oncology molecules including T cell agonists are entering clinical development and could be hypothesized to enhance the activity of oncolytic viruses, including VSV-IFNß. Here, we show that VSV-IFNß exhibits multiple mechanisms of action, including direct cell killing, stimulation of an innate immune response, recruitment of CD8 T cells, and depletion of T regulatory cells. Moreover, VSV-IFNß promotes the establishment of a CD8 T cell response to endogenous tumor antigens. Our data demonstrate a significant enhancement of anti-tumor function for VSV-IFNß when combined with checkpoint inhibitors, but not OX40 agonists. While the addition of checkpoint inhibitors to VSV-IFNß generated robust tumor growth inhibition, it resulted in no increase in viral replication, transgene expression, or immunophenotypic changes beyond treatment with VSV-IFNß alone. We hypothesize that tumor-specific T cells generated by VSV-IFNß retain activity due to a lack of immune exhaustion when checkpoint inhibitors were used.


Asunto(s)
Terapia Genética , Vectores Genéticos/genética , Inmunoterapia , Neoplasias/genética , Neoplasias/inmunología , Viroterapia Oncolítica , Virus Oncolíticos/genética , Virus de la Estomatitis Vesicular Indiana/genética , Animales , Anticuerpos Monoclonales/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Biomarcadores de Tumor , Terapia Combinada , Modelos Animales de Enfermedad , Expresión Génica , Terapia Genética/métodos , Humanos , Inmunomodulación , Inmunoterapia/métodos , Interferón beta/genética , Interferón beta/metabolismo , Interferones/genética , Interferones/metabolismo , Melanoma Experimental , Ratones , Neoplasias/patología , Neoplasias/terapia , Receptores OX40/agonistas , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Transducción Genética , Transgenes , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Eur J Immunol ; 45(6): 1706-15, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25754933

RESUMEN

Interactions between ICOS and ICOS ligand (ICOSL) are essential for the development of T follicular helper (Tfh) cells and thus the formation and maintenance of GC reactions. Given the conflicting reports on the requirement of other CD4(+) T-cell populations for ICOS signals, we have employed a range of in vivo approaches to dissect requirements for ICOS signals in mice during an endogenous CD4(+) T-cell response and contrasted this with CD28 signals. Genetic absence of ICOSL only modestly reduced the total number of antigen-specific CD4(+) T cells at the peak of the primary response, but resulted in a severely diminished number of both T central memory and T effector memory cells. Treatment with blocking anti-ICOS mAb during the primary response recapitulated these effects and caused a more substantial reduction than blocking CD28 signals with CTLA4Ig. During the memory phase of the response further signals through ICOS or CD28 were not required for survival. However, upon secondary challenge only Tfh cell expansion remained heavily ICOS-dependent, while CD28 signals were required for optimal expansion of all subsets. These data demonstrate the importance of ICOS signals specifically for memory CD4(+) T-cell formation, while highlighting the potential of therapeutically targeting this pathway.


Asunto(s)
Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Memoria Inmunológica , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Infecciones Bacterianas/genética , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Supervivencia Celular/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/antagonistas & inhibidores , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Ratones , Ratones Noqueados , Transducción de Señal
18.
J Immunol ; 193(10): 4823-32, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25281717

RESUMEN

Plasma cells and the autoreactive Abs they produce are suspected to contribute to the pathogenesis of multiple sclerosis, but recent attempts to target these components of humoral immunity have failed. MEDI551, an anti-CD19 Ab that depletes mature B cells including plasma cells may offer a compelling alternative that reduces pathogenic adaptive immune responses while sparing regulatory mechanisms. Indeed, our data demonstrate that a single dose of MEDI551, given before or during ongoing experimental autoimmune encephalomyelitis, disrupts development of the disease. Leukocyte infiltration into the spinal cord is significantly reduced, as well as short-lived and long-lived autoreactive CD138(+) plasma cells in the spleen and bone marrow, respectively. In addition, potentially protective CD1d(hi)CD5(+) regulatory B cells show resistance to depletion, and myelin-specific Foxp3(+) regulatory T cells are expanded. Taken together, these results demonstrate that MEDI551 disrupts experimental autoimmune encephalomyelitis by inhibiting multiple proinflammatory components whereas preserving regulatory populations.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Anticuerpos/farmacología , Antígenos CD19/inmunología , Médula Ósea/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Médula Espinal/inmunología , Animales , Antígenos CD19/genética , Antígenos CD1d/genética , Antígenos CD1d/inmunología , Linfocitos B Reguladores/inmunología , Linfocitos B Reguladores/patología , Médula Ósea/patología , Supervivencia Celular , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Expresión Génica , Depleción Linfocítica , Masculino , Ratones , Ratones Transgénicos , Glicoproteína Mielina-Oligodendrócito , Células Plasmáticas/inmunología , Células Plasmáticas/patología , Ingeniería de Proteínas , Transducción de Señal , Médula Espinal/patología , Bazo/inmunología , Bazo/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
19.
J Immunol ; 192(4): 1480-90, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24442430

RESUMEN

B cell activation is regulated by a variety of signals. CD19 positively regulates B cell activation, augmenting signals delivered through the BCR complex. In contrast, CD32b contains an ITIM and negatively regulates BCR signaling. Importantly, there are drugs currently in clinical trials and preclinical development that cross-link CD32b to molecules within the BCR complex. We wanted to address how single engagement versus cotargeting these molecules affects human B cell function. When B cells from healthy individuals were activated by signals that mimic a T cell response (IL-21 costimulation), ligation of CD32b, but not CD19, inhibited B cell expansion and plasma cell (PC) differentiation. In contrast, when B cells were activated through TLR, anti-CD19, but not anti-CD32b, blunted the response. However, when both CD19 and CD32b were coengaged by a bispecific anti-CD19×CD32b Ab, both types of stimuli were potently inhibited. Cross-linking CD19 with CD32b also inhibited Ab-independent functions of B cells, such as HLA upregulation, cytokine production, and the ability of B cells to prime CD4(+) T cells. Finally, although cross-linking CD19 and CD32b inhibited PC differentiation of primary B cells, it did not alter Ig production from pre-established PCs. These data elucidate the mechanism by which a complex set of signals determines the fate of B cell responsiveness. Although signals through CD19 influence TLR-driven activation, CD32b impacts the magnitude of the response following IL-21 costimulation. Therefore, simultaneous targeting of multiple surface molecules may be a necessary approach to comprehensively modulate B cell activation in vivo.


Asunto(s)
Antígenos CD19/metabolismo , Linfocitos B/inmunología , Activación de Linfocitos/inmunología , Células Plasmáticas/metabolismo , Receptores de IgG/metabolismo , Anticuerpos/inmunología , Antígenos CD19/biosíntesis , Antígenos CD19/inmunología , Antígenos de Diferenciación de Linfocitos B/inmunología , Enfermedades Autoinmunes/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Muerte Celular/inmunología , Diferenciación Celular , Células Cultivadas , Reactivos de Enlaces Cruzados , Humanos , Memoria Inmunológica/inmunología , Interleucinas/metabolismo , Unión Proteica/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de IgG/biosíntesis , Receptores de IgG/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/metabolismo
20.
Am J Respir Crit Care Med ; 192(6): 706-18, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26266827

RESUMEN

RATIONALE: B cell-activating factor (BAFF) plays a major role in activation of B cells and in adaptive humoral immune responses. In chronic obstructive pulmonary disease (COPD), lymphoid follicles have been associated with disease severity, and overexpression of BAFF has been demonstrated within lymphoid follicles of patients with severe COPD. OBJECTIVES: To investigate expression and localization of BAFF in the lungs of patients with COPD and to study the role of BAFF in COPD by antagonizing BAFF in a mouse model of chronic cigarette smoke (CS) exposure. METHODS: We quantified and localized BAFF expression in lungs of never-smokers, smokers without COPD, and patients with COPD and in lungs of air- or CS-exposed mice by reverse-transcriptase polymerase chain reaction, ELISA, immunohistochemistry, and confocal imaging. Next, to investigate the role of BAFF in COPD, we antagonized BAFF by prophylactic or therapeutic administration of a soluble fusion protein of the BAFF-receptor, BAFFR-Fc, in mice exposed to air or CS for 24 weeks and evaluated several hallmarks of COPD and polarization of lung macrophages. MEASUREMENTS AND MAIN RESULTS: BAFF expression was significantly increased in lungs of patients with COPD and CS-exposed mice. BAFF staining in lymphoid follicles was observed around B cells, CD4(+) cells, dendritic cells, follicular dendritic cells, and fibroblastic reticular cells. Prophylactic and therapeutic administration of BAFFR-Fc in mice reduced pulmonary B-cell numbers and prevented CS-induced formation of lymphoid follicles and increases in immunoglobulin levels. Interestingly, prophylactic BAFFR-Fc administration significantly attenuated pulmonary inflammation and destruction of alveolar walls. Moreover, antagonizing BAFF altered the phenotype of alveolar and interstitial macrophages. CONCLUSIONS: BAFF is significantly increased in lungs of patients with COPD and is present around both immune and stromal cells within lymphoid follicles. Antagonizing BAFF in CS-exposed mice attenuates pulmonary inflammation and alveolar destruction.


Asunto(s)
Factor Activador de Células B/metabolismo , Pulmón/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Inmunidad Adaptativa , Anciano , Animales , Factor Activador de Células B/antagonistas & inhibidores , Factor Activador de Células B/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunohistoquímica , Pulmón/inmunología , Tejido Linfoide/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Humo/efectos adversos , Fumar/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA