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










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 3148, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672305

RESUMEN

Checkpoint blockade with Pembrolizumab, has demonstrated durable clinical responses in advanced non-small cell lung cancer, however, treatment is offset by the development of high-grade immune related adverse events (irAEs) in some patients. Here, we show that in these patients a deficient Breg checkpoint fails to limit self-reactive T cell enhanced activity and auto-antibody formation enabled by PD-1/PD-L1 blockade, leading to severe auto-inflammatory sequelae. Principally a failure of IL-10 producing regulatory B cells as demonstrated through functional ex vivo assays and deep phenotyping mass cytometric analysis, is a major and significant finding in patients who develop high-grade irAEs when undergoing treatment with anti-PD1/PD-L1 checkpoint blockade. There is currently a lack of biomarkers to identify a priori those patients at greatest risk of developing severe auto-inflammatory syndrome. Pre-therapy B cell profiling could provide an important tool to identify lung cancer patients at high risk of developing severe irAEs on checkpoint blockade.


Asunto(s)
Linfocitos B Reguladores , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Antígeno B7-H1/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Receptor de Muerte Celular Programada 1/genética
2.
Immunol Lett ; 244: 1-11, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35227735

RESUMEN

The formation of high-affinity antibodies that protect against infection requires the formation of germinal centres (GCs), where specialized T follicular helper cells (Tfh) provide help to B cells. Those T-B interactions are critical in supporting isotype switching and affinity maturation of B cells. However, GC responses need to be tightly regulated by specialized Foxp3-expressing T follicular regulatory cells (Tfr). It has been shown that the failure of Tfr cells to regulate GC responses can lead to antibody-mediated autoimmunity. Hence, the balance between protection against infection versus tolerance towards self requires an appropriate regulation of cellular and molecular events within secondary lymphoid tissue. Here, we review the development and biology of these T follicular cell subsets, with special emphasis on the metabolic regulation of Tfh cells, thus contributing to a greater understanding of GC responses.


Asunto(s)
Linfocitos T Colaboradores-Inductores , Linfocitos T Reguladores , Linfocitos B , Centro Germinal , Homeostasis
3.
PLoS One ; 16(9): e0256813, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34525109

RESUMEN

There is a worldwide need for reagents to perform SARS-CoV-2 detection. Some laboratories have implemented kit-free protocols, but many others do not have the capacity to develop these and/or perform manual processing. We provide multiple workflows for SARS-CoV-2 nucleic acid detection in clinical samples by comparing several commercially available RNA extraction methods: QIAamp Viral RNA Mini Kit (QIAgen), RNAdvance Blood/Viral (Beckman) and Mag-Bind Viral DNA/RNA 96 Kit (Omega Bio-tek). We also compared One-step RT-qPCR reagents: TaqMan Fast Virus 1-Step Master Mix (FastVirus, ThermoFisher Scientific), qPCRBIO Probe 1-Step Go Lo-ROX (PCR Biosystems) and Luna® Universal Probe One-Step RT-qPCR Kit (Luna, NEB). We used primer-probes that detect viral N (EUA CDC) and RdRP. RNA extraction methods provided similar results, with Beckman performing better with our primer-probe combinations. Luna proved most sensitive although overall the three reagents did not show significant differences. N detection was more reliable than that of RdRP, particularly in samples with low viral titres. Importantly, we demonstrated that heat treatment of nasopharyngeal swabs at 70°C for 10 or 30 min, or 90°C for 10 or 30 min (both original variant and B 1.1.7) inactivated SARS-CoV-2 employing plaque assays, and had minimal impact on the sensitivity of the qPCR in clinical samples. These findings make SARS-CoV-2 testing portable in settings that do not have CL-3 facilities. In summary, we provide several testing pipelines that can be easily implemented in other laboratories and have made all our protocols and SOPs freely available at https://osf.io/uebvj/.


Asunto(s)
Prueba de COVID-19/métodos , COVID-19/diagnóstico , Calor , ARN Viral/genética , SARS-CoV-2/genética , Inactivación de Virus , COVID-19/epidemiología , COVID-19/virología , Epidemias/prevención & control , Humanos , Nasofaringe/virología , Juego de Reactivos para Diagnóstico , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , SARS-CoV-2/fisiología , Sensibilidad y Especificidad , Manejo de Especímenes/métodos , Flujo de Trabajo
4.
Clin Sci (Lond) ; 135(11): 1389-1408, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34086048

RESUMEN

The immune system protects the body against harm by inducing inflammation. During the immune response, cells of the immune system get activated, divided and differentiated in order to eliminate the danger signal. This process relies on the metabolic reprogramming of both catabolic and anabolic pathways not only to produce energy in the form of ATP but also to generate metabolites that exert key functions in controlling the response. Equally important to mounting an appropriate effector response is the process of immune resolution, as uncontrolled inflammation is implicated in the pathogenesis of many human diseases, including allergy, chronic inflammation and cancer. In this review, we aim to introduce the reader to the field of cholesterol immunometabolism and discuss how both metabolites arising from the pathway and cholesterol homeostasis are able to impact innate and adaptive immune cells, staging cholesterol homeostasis at the centre of an adequate immune response. We also review evidence that demonstrates the clear impact that cholesterol metabolism has in both the induction and the resolution of the inflammatory response. Finally, we propose that emerging data in this field not only increase our understanding of immunometabolism but also provide new tools for monitoring and intervening in human diseases, where controlling and/or modifying inflammation is desirable.


Asunto(s)
Colesterol/metabolismo , Sistema Inmunológico/inmunología , Inflamación/metabolismo , Metabolismo de los Lípidos/inmunología , Animales , Colesterol/inmunología , Metabolismo Energético/inmunología , Metabolismo Energético/fisiología , Humanos , Inflamación/inmunología , Transducción de Señal/inmunología , Transducción de Señal/fisiología
5.
medRxiv ; 2021 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-33851184

RESUMEN

There is a worldwide need for reagents to perform SARS-CoV-2 detection. Some laboratories have implemented kit-free protocols, but many others do not have the capacity to develop these and/or perform manual processing. We provide multiple workflows for SARS-CoV-2 nucleic acid detection in clinical samples by comparing several commercially available RNA extraction methods: QIAamp Viral RNA Mini Kit (QIAgen), RNAdvance Blood/Viral (Beckman) and Mag-Bind Viral DNA/RNA 96 Kit (Omega Bio-tek). We also compared One-step RT-qPCR reagents: TaqMan Fast Virus 1-Step Master Mix (FastVirus, ThermoFisher Scientific), qPCRBIO Probe 1-Step Go Lo-ROX (PCR Biosystems) and Luna ® Universal Probe One-Step RT-qPCR Kit (Luna, NEB). We used primer-probes that detect viral N (EUA CDC) and RdRP (PHE guidelines). All RNA extraction methods provided similar results. FastVirus and Luna proved most sensitive. N detection was more reliable than that of RdRP, particularly in samples with low viral titres. Importantly, we demonstrate that treatment of nasopharyngeal swabs with 70 degrees for 10 or 30 min, or 90 degrees for 10 or 30 min (both original variant and B 1.1.7) inactivates SARS-CoV-2 employing plaque assays, and that it has minimal impact on the sensitivity of the qPCR in clinical samples. These findings make SARS-CoV-2 testing portable to settings that do not have CL-3 facilities. In summary, we provide several testing pipelines that can be easily implemented in other laboratories and have made all our protocols and SOPs freely available at https://osf.io/uebvj/ .

6.
Nat Commun ; 11(1): 3412, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32641742

RESUMEN

Regulatory B cells restrict immune and inflammatory responses across a number of contexts. This capacity is mediated primarily through the production of IL-10. Here we demonstrate that the induction of a regulatory program in human B cells is dependent on a metabolic priming event driven by cholesterol metabolism. Synthesis of the metabolic intermediate geranylgeranyl pyrophosphate (GGPP) is required to specifically drive IL-10 production, and to attenuate Th1 responses. Furthermore, GGPP-dependent protein modifications control signaling through PI3Kδ-AKT-GSK3, which in turn promote BLIMP1-dependent IL-10 production. Inherited gene mutations in cholesterol metabolism result in a severe autoinflammatory syndrome termed mevalonate kinase deficiency (MKD). Consistent with our findings, B cells from MKD patients induce poor IL-10 responses and are functionally impaired. Moreover, metabolic supplementation with GGPP is able to reverse this defect. Collectively, our data define cholesterol metabolism as an integral metabolic pathway for the optimal functioning of human IL-10 producing regulatory B cells.


Asunto(s)
Linfocitos B Reguladores/metabolismo , Colesterol/metabolismo , Interleucina-10/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Animales , Antígenos CD19/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Técnicas de Cocultivo , Enfermedades Autoinflamatorias Hereditarias/metabolismo , Humanos , Macrófagos/metabolismo , Síndrome Metabólico/metabolismo , Deficiencia de Mevalonato Quinasa/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Análisis de Componente Principal , Transducción de Señal , Células TH1/metabolismo , Receptor Toll-Like 9/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
Rheumatology (Oxford) ; 58(Suppl 7): vii17-vii28, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31816081

RESUMEN

Immune check point inhibitor (CPI) therapy has revolutionized treatment paradigms for several cancers, but at the cost of triggering a diverse spectrum of immune-mediated injury to non-cancer tissues. The complex biology of these toxicities remains incompletely understood, partly because tissue acquisition from affected areas can be challenging to retrieve, thus hindering development of targeted therapy. Here, we review the literature describing pathology of immune-mediated tissue lesions including gastrointestinal, skin, rheumatic, pulmonary, cardiac, renal and hepatic lesions and highlight key immunological insights.


Asunto(s)
Antineoplásicos Inmunológicos/efectos adversos , Factores Inmunológicos/efectos adversos , Inmunoterapia/efectos adversos , Neoplasias/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Erupciones por Medicamentos/etiología , Erupciones por Medicamentos/patología , Enfermedades Gastrointestinales/inducido químicamente , Enfermedades Gastrointestinales/patología , Humanos , Inmunoterapia/métodos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/patología , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/patología , Miocarditis/inducido químicamente , Miocarditis/patología , Enfermedades Reumáticas/inducido químicamente , Enfermedades Reumáticas/patología
8.
Nat Commun ; 10(1): 498, 2019 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-30700717

RESUMEN

The mechanisms controlling CD4+ T cell switching from an effector to an anti-inflammatory (IL-10+) phenotype play an important role in the persistence of chronic inflammatory diseases. Here, we identify the cholesterol biosynthesis pathway as a key regulator of this process. Pathway analysis of cultured cytokine-producing human T cells reveals a significant association between IL-10 and cholesterol metabolism gene expression. Inhibition of the cholesterol biosynthesis pathway with atorvastatin or 25-hydroxycholesterol during switching from IFNγ+ to IL-10+ shows a specific block in immune resolution, defined as a significant decrease in IL-10 expression. Mechanistically, the master transcriptional regulator of IL10 in T cells, c-Maf, is significantly decreased by physiological levels of 25-hydroxycholesterol. Strikingly, progression to rheumatoid arthritis is associated with altered expression of cholesterol biosynthesis genes in synovial biopsies of predisposed individuals. Our data reveal a link between sterol metabolism and the regulation of the anti-inflammatory response in human CD4+ T cells.


Asunto(s)
Colesterol/biosíntesis , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Células TH1/metabolismo , Atorvastatina/farmacología , Humanos , Hidroxicolesteroles/farmacología , Células TH1/efectos de los fármacos
9.
Am J Transplant ; 18(6): 1370-1379, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29392897

RESUMEN

Improvements in immunosuppression have modified short-term survival of deceased-donor allografts, but not their rate of long-term failure. Mismatches between donor and recipient HLA play an important role in the acute and chronic allogeneic immune response against the graft. Perfect matching at clinically relevant HLA loci does not obviate the need for immunosuppression, suggesting that additional genetic variation plays a critical role in both short- and long-term graft outcomes. By combining patient data and samples from supranational cohorts across the United Kingdom and European Union, we performed the first large-scale genome-wide association study analyzing both donor and recipient DNA in 2094 complete renal transplant-pairs with replication in 5866 complete pairs. We studied deceased-donor grafts allocated on the basis of preferential HLA matching, which provided some control for HLA genetic effects. No strong donor or recipient genetic effects contributing to long- or short-term allograft survival were found outside the HLA region. We discuss the implications for future research and clinical application.


Asunto(s)
Estudio de Asociación del Genoma Completo , Trasplante de Riñón , Donantes de Tejidos , Receptores de Trasplantes , Adulto , Replicación del ADN , Femenino , Genotipo , Supervivencia de Injerto/inmunología , Prueba de Histocompatibilidad , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Trasplante Homólogo
10.
PLoS Genet ; 13(2): e1006587, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28187197

RESUMEN

The polarization of CD4+ T cells into distinct T helper cell lineages is essential for protective immunity against infection, but aberrant T cell polarization can cause autoimmunity. The transcription factor T-bet (TBX21) specifies the Th1 lineage and represses alternative T cell fates. Genome-wide association studies have identified single nucleotide polymorphisms (SNPs) that may be causative for autoimmune diseases. The majority of these polymorphisms are located within non-coding distal regulatory elements. It is considered that these genetic variants contribute to disease by altering the binding of regulatory proteins and thus gene expression, but whether these variants alter the binding of lineage-specifying transcription factors has not been determined. Here, we show that SNPs associated with the mucosal inflammatory diseases Crohn's disease, ulcerative colitis (UC) and celiac disease, but not rheumatoid arthritis or psoriasis, are enriched at T-bet binding sites. Furthermore, we identify disease-associated variants that alter T-bet binding in vitro and in vivo. ChIP-seq for T-bet in individuals heterozygous for the celiac disease-associated SNPs rs1465321 and rs2058622 and the IBD-associated SNPs rs1551398 and rs1551399, reveals decreased binding to the minor disease-associated alleles. Furthermore, we show that rs1465321 is an expression quantitative trait locus (eQTL) for the neighboring gene IL18RAP, with decreased T-bet binding associated with decreased expression of this gene. These results suggest that genetic polymorphisms may predispose individuals to mucosal autoimmune disease through alterations in T-bet binding. Other disease-associated variants may similarly act by modulating the binding of lineage-specifying transcription factors in a tissue-selective and disease-specific manner.


Asunto(s)
Enfermedad Celíaca/genética , Colitis Ulcerosa/genética , Enfermedad de Crohn/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple , Proteínas de Dominio T Box/genética , Animales , Sitios de Unión/genética , Western Blotting , Linfocitos T CD4-Positivos/metabolismo , Enfermedad Celíaca/metabolismo , Células Cultivadas , Colitis Ulcerosa/metabolismo , Enfermedad de Crohn/metabolismo , Expresión Génica , Estudio de Asociación del Genoma Completo/métodos , Humanos , Subunidad beta del Receptor de Interleucina-18/genética , Subunidad beta del Receptor de Interleucina-18/metabolismo , Ratones Noqueados , Unión Proteica/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas de Dominio T Box/metabolismo , Células TH1/metabolismo
11.
Gastroenterology ; 149(2): 456-67.e15, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25917784

RESUMEN

BACKGROUND & AIMS: Innate lymphoid cells (ILCs) are a heterogeneous group of mucosal inflammatory cells that participate in chronic intestinal inflammation. We investigated the role of interleukin 6 (IL6) in inducing activation of ILCs in mice and in human beings with chronic intestinal inflammation. METHODS: ILCs were isolated from colons of Tbx21(-/-) × Rag2(-/-) mice (TRUC), which develop colitis; patients with inflammatory bowel disease (IBD); and patients without colon inflammation (controls). ILCs were characterized by flow cytometry; cytokine production was measured by enzyme-linked immunosorbent assay and cytokine bead arrays. Mice were given intraperitoneal injections of depleting (CD4, CD90), neutralizing (IL6), or control antibodies. Isolated colon tissues were analyzed by histology, explant organ culture, and cell culture. Bacterial DNA was extracted from mouse fecal samples to assess the intestinal microbiota. RESULTS: IL17A- and IL22-producing, natural cytotoxicity receptor-negative, ILC3 were the major subset of ILCs detected in colons of TRUC mice. Combinations of IL23 and IL1α induced production of cytokines by these cells, which increased further after administration of IL6. Antibodies against IL6 reduced colitis in TRUC mice without significantly affecting the structure of their intestinal microbiota. Addition of IL6 increased production of IL17A, IL22, and interferon-γ by human intestinal CD3-negative, IL7-receptor-positive cells, in a dose-dependent manner. CONCLUSIONS: IL6 contributes to activation of colonic natural cytotoxicity receptor-negative, CD4-negative, ILC3s in mice with chronic intestinal inflammation (TRUC mice) by increasing IL23- and IL1α-induced production of IL17A and IL22. This pathway might be targeted to treat patients with IBD because IL6, which is highly produced in colonic tissue by some IBD patients, also increased the production of IL17A, IL22, and interferon-γ by cultured human colon CD3-negative, IL7-receptor-positive cells.


Asunto(s)
Antígenos CD4/metabolismo , Citocinas/metabolismo , Inmunidad Innata/efectos de los fármacos , Enfermedades Inflamatorias del Intestino/inmunología , Interleucina-6/farmacología , Linfocitos/efectos de los fármacos , Animales , Complejo CD3/metabolismo , Técnicas de Cultivo de Célula , Colon/citología , Colon/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-23/metabolismo , Interleucina-6/administración & dosificación , Interleucinas/metabolismo , Linfocitos/inmunología , Ratones , Ratones Noqueados , Receptores Gatillantes de la Citotoxidad Natural/metabolismo , Interleucina-22
12.
J Immunol ; 191(12): 5925-32, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24249732

RESUMEN

The complex relationship between Th1 and Th17 cells is incompletely understood. The transcription factor T-bet is best known as the master regulator of Th1 lineage commitment. However, attention is now focused on the repression of alternate T cell subsets mediated by T-bet, particularly the Th17 lineage. It has recently been suggested that pathogenic Th17 cells express T-bet and are dependent on IL-23. However, T-bet has previously been shown to be a negative regulator of Th17 cells. We have taken an unbiased approach to determine the functional impact of T-bet on Th17 lineage commitment. Genome-wide analysis of functional T-bet binding sites provides an improved understanding of the transcriptional regulation mediated by T-bet, and suggests novel mechanisms by which T-bet regulates Th cell differentiation. Specifically, we show that T-bet negatively regulates Th17 lineage commitment via direct repression of the transcription factor IFN regulatory factor-4 (IRF4). An in vivo analysis of the pathogenicity of T-bet-deficient T cells demonstrated that mucosal Th17 responses were augmented in the absence of T-bet, and we have demonstrated that the roles of T-bet in enforcing Th1 responses and suppressing Th17 responses are separable. The interplay of the two key transcription factors T-bet and IRF4 during the determination of T cell fate choice significantly advances our understanding of the mechanisms underlying the development of pathogenic T cells.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Factores Reguladores del Interferón/antagonistas & inhibidores , Linfopoyesis/genética , Proteínas de Dominio T Box/fisiología , Células Th17/citología , Transcripción Genética , Traslado Adoptivo , Animales , Sitios de Unión , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Células Cultivadas , Quimera , Colitis/inmunología , Proteínas de Unión al ADN/deficiencia , Femenino , Genes Reporteros , Vectores Genéticos , Estudio de Asociación del Genoma Completo , Factores Reguladores del Interferón/biosíntesis , Factores Reguladores del Interferón/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Dominio T Box/genética
13.
Blood ; 122(20): 3473-81, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-23878142

RESUMEN

Interleukin-1ß (IL-1ß) is a proinflammatory cytokine and a therapeutic target in several chronic autoimmune states. Monocytes and macrophages are the major sources of IL-1ß. IL-1ß production by these cells requires Toll-like receptor (TLR) and adenosine triphosphate (ATP)-mediated P2X purinoceptor 7 (P2X7) signals, which together activate the inflammasome. However, how TLR signals and ATP availability are regulated during monocyte activation is unclear and the involvement of another danger signal system has been proposed. Here, we demonstrate that both lipopolysaccharide (LPS) and the anaphylatoxin C3a are needed for IL-1ß production in human macrophages and dendritic cells, while in monocytes, C3a enhanced the secretion of LPS-induced IL-1ß. C3a and LPS-stimulated monocytes increased T helper 17 (Th17) cell induction in vitro, and human rejecting, but not nonrejecting, kidney transplant biopsies were characterized by local generation of C3a and monocyte and Th17 cell infiltration. Mechanistically, C3a drives IL-1ß production in monocytes by controlling the release of intracellular ATP into the extracellular space via regulation of as-yet unidentified ATP-releasing channels in an extracellular signal-regulated kinase 1/2-dependent fashion. These data define a novel function for complement in inflammasome activation in monocytes and suggest that C3aR-mediated signaling is a vital component of the IL-1ß-Th17 axis.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteínas Portadoras/fisiología , Complemento C3/fisiología , Inflamasomas/fisiología , Interleucina-1beta/metabolismo , Monocitos/metabolismo , Células Cultivadas , Complemento C3/agonistas , Citocinas/biosíntesis , Citocinas/genética , Células Dendríticas/metabolismo , Activación Enzimática , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Humanos , Trasplante de Riñón , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas , Antígeno de Macrófago-1/efectos de los fármacos , Antígeno de Macrófago-1/fisiología , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores Purinérgicos P2X7/fisiología , Proteínas Recombinantes/farmacología , Células Th17/metabolismo , Receptores Toll-Like/fisiología
14.
PLoS One ; 8(7): e65485, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23843942

RESUMEN

Discovery of novel immune biomarkers for monitoring of disease prognosis and response to therapy in immune-mediated inflammatory diseases is an important unmet clinical need. Here, we establish a novel framework for immunological biomarker discovery, comparing a conventional (liquid) flow cytometry platform (CFP) and a unique lyoplate-based flow cytometry platform (LFP) in combination with advanced computational data analysis. We demonstrate that LFP had higher sensitivity compared to CFP, with increased detection of cytokines (IFN-γ and IL-10) and activation markers (Foxp3 and CD25). Fluorescent intensity of cells stained with lyophilized antibodies was increased compared to cells stained with liquid antibodies. LFP, using a plate loader, allowed medium-throughput processing of samples with comparable intra- and inter-assay variability between platforms. Automated computational analysis identified novel immunophenotypes that were not detected with manual analysis. Our results establish a new flow cytometry platform for standardized and rapid immunological biomarker discovery with wide application to immune-mediated diseases.


Asunto(s)
Biomarcadores , Biología Computacional/métodos , Citometría de Flujo/métodos , Inmunofenotipificación/métodos , Inflamación/inmunología , Inflamación/metabolismo , Adulto , Anticuerpos , Femenino , Citometría de Flujo/normas , Humanos , Inmunofenotipificación/normas , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Adulto Joven
15.
Cell Metab ; 17(4): 520-33, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23562076

RESUMEN

Low-grade inflammation in fat is associated with insulin resistance, although the mechanisms are unclear. We report that mice deficient in the immune cell transcription factor T-bet have lower energy expenditure and increased visceral fat compared with wild-type mice, yet paradoxically are more insulin sensitive. This striking phenotype, present in young T-bet(-/-) mice, persisted with high-fat diet and increasing host age and was associated with altered immune cell numbers and cytokine secretion specifically in visceral adipose tissue. However, the favorable metabolic phenotype observed in T-bet-deficient hosts was lost in T-bet(-/-) mice also lacking adaptive immunity (T-bet(-/-)xRag2(-/-)), demonstrating that T-bet expression in the adaptive rather than the innate immune system impacts host glucose homeostasis. Indeed, adoptive transfer of T-bet-deficient, but not wild-type, CD4(+) T cells to Rag2(-/-) mice improved insulin sensitivity. Our results reveal a role for T-bet in metabolic physiology and obesity-associated insulin resistance.


Asunto(s)
Resistencia a la Insulina , Grasa Intraabdominal/metabolismo , Proteínas de Dominio T Box/metabolismo , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dieta Alta en Grasa , Metabolismo Energético , Sistema Inmunológico/metabolismo , Técnicas In Vitro , Interferón gamma/deficiencia , Interferón gamma/genética , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Obesidad/metabolismo , Obesidad/patología , Fenotipo , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética
16.
Nat Commun ; 3: 1268, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23232398

RESUMEN

T-bet and GATA3 regulate the CD4+ T cell Th1/Th2 cell fate decision but little is known about the interplay between these factors outside of the murine Ifng and Il4/Il5/Il13 loci. Here we show that T-bet and GATA3 bind to multiple distal sites at immune regulatory genes in human effector T cells. These sites display markers of functional elements, act as enhancers in reporter assays and are associated with a requirement for T-bet and GATA3. Furthermore, we demonstrate that both factors bind distal sites at Tbx21 and that T-bet directly activates its own expression. We also show that in Th1 cells, GATA3 is distributed away from Th2 genes, instead occupying T-bet binding sites at Th1 genes, and that T-bet is sufficient to induce GATA3 binding at these sites. We propose these aspects of T-bet and GATA3 function are important for Th1/Th2 differentiation and for understanding transcription factor interactions in other T cell lineage decisions.


Asunto(s)
Factor de Transcripción GATA3/fisiología , Proteínas de Dominio T Box/fisiología , Células TH1/fisiología , Células Th2/fisiología , Animales , Sitios de Unión/fisiología , Linfocitos T CD4-Positivos/fisiología , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Ratones , Ratones Endogámicos BALB C , Análisis de Secuencia por Matrices de Oligonucleótidos , Secuencias Reguladoras de Ácidos Nucleicos
17.
Cytometry A ; 79(7): 496-506, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21638766

RESUMEN

Asymmetric cell division is an important mechanism for generating cellular diversity, however, techniques for measuring the distribution of fate-regulating molecules during mitosis have been hampered by a lack of objectivity, quantitation, and statistical robustness. Here we describe a novel imaging flow cytometric approach that is able to report a cells proliferative history and cell cycle position using dye dilution, pH3, and PI staining to then measure the spatial distribution of fluorescent signals during mitosis using CCD-derived imagery. Using Jurkat cells, resolution of the fluorescently labeled populations was comparable to traditional PMT based cytometers thus eliminating the need to sort cells with specific division histories for microscopy. Subdividing mitotic stages by morphology allowed us to determine the time spent in each cell cycle phase using mathematical modeling approaches. Furthermore high sample throughput allowed us to collect statistically relevant numbers of cells without the need to use blocking agents that artificially enrich for mitotic events. The fluorescent imagery was used to measure PKCζ protein and EEA-1+ endosome distribution during different mitotic phases in Jurkat cells. While telophase cells represented the favorable population for measuring asymmetry, asynchronously dividing cells spent approximately 43 seconds in this stage, explaining why they were present at such low frequencies. This necessitated the acquisition of large cell numbers. Interestingly we found that PKCζ was inherited asymmetrically in 2.5% of all telophasic events whereas endosome inheritance was significantly more symmetrical. Furthermore, molecular polarity at early mitotic phases was a poor indicator of asymmetry during telophase highlighting that, though rare, telophasic events represented the best candidates for asymmetry studies. In summary, this technique combines the spatial information afforded by fluorescence microscopy with the statistical wealth and objectivity of traditional flow cytometry, overcoming the key limitations of existing approaches for studying asymmetry during mitosis.


Asunto(s)
División Celular/fisiología , Citometría de Flujo/métodos , Citometría de Imagen/métodos , Mitosis/fisiología , Animales , Citometría de Flujo/instrumentación , Humanos , Citometría de Imagen/instrumentación , Células Jurkat/citología , Microscopía Fluorescente/métodos , Proteína Quinasa C/metabolismo
18.
Kidney Int Suppl (2011) ; 1(2): 40-46, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25018902

RESUMEN

Induction of transplantation tolerance remains the ideal long-term clinical and logistic solution to the current challenges facing the management of renal allograft recipients. In this review, we describe the recent studies and advances made in identifying biomarkers of renal transplant tolerance, from study inceptions, to the lessons learned and their implications for current and future studies with the same goal. With the age of biomarker discovery entering a new dimension of high-throughput technologies, here we also review the current approaches, developments, and pitfalls faced in the subsequent statistical analysis required to identify valid biomarker candidates.

19.
J Clin Invest ; 120(6): 1848-61, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20501943

RESUMEN

Identifying transplant recipients in whom immunological tolerance is established or is developing would allow an individually tailored approach to their posttransplantation management. In this study, we aimed to develop reliable and reproducible in vitro assays capable of detecting tolerance in renal transplant recipients. Several biomarkers and bioassays were screened on a training set that included 11 operationally tolerant renal transplant recipients, recipient groups following different immunosuppressive regimes, recipients undergoing chronic rejection, and healthy controls. Highly predictive assays were repeated on an independent test set that included 24 tolerant renal transplant recipients. Tolerant patients displayed an expansion of peripheral blood B and NK lymphocytes, fewer activated CD4+ T cells, a lack of donor-specific antibodies, donor-specific hyporesponsiveness of CD4+ T cells, and a high ratio of forkhead box P3 to alpha-1,2-mannosidase gene expression. Microarray analysis further revealed in tolerant recipients a bias toward differential expression of B cell-related genes and their associated molecular pathways. By combining these indices of tolerance as a cross-platform biomarker signature, we were able to identify tolerant recipients in both the training set and the test set. This study provides an immunological profile of the tolerant state that, with further validation, should inform and shape drug-weaning protocols in renal transplant recipients.


Asunto(s)
Biomarcadores/metabolismo , Tolerancia Inmunológica/inmunología , Inmunosupresores/inmunología , Trasplante de Riñón/inmunología , Humanos , Tolerancia Inmunológica/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Donantes de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...