Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Immunol ; 23(9): 1365-1378, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35999394

RESUMEN

CD28 and CTLA-4 (CD152) play essential roles in regulating T cell immunity, balancing the activation and inhibition of T cell responses, respectively. Although both receptors share the same ligands, CD80 and CD86, the specific requirement for two distinct ligands remains obscure. In the present study, we demonstrate that, although CTLA-4 targets both CD80 and CD86 for destruction via transendocytosis, this process results in separate fates for CTLA-4 itself. In the presence of CD80, CTLA-4 remained ligand bound, and was ubiquitylated and trafficked via late endosomes and lysosomes. In contrast, in the presence of CD86, CTLA-4 detached in a pH-dependent manner and recycled back to the cell surface to permit further transendocytosis. Furthermore, we identified clinically relevant mutations that cause autoimmune disease, which selectively disrupted CD86 transendocytosis, by affecting either CTLA-4 recycling or CD86 binding. These observations provide a rationale for two distinct ligands and show that defects in CTLA-4-mediated transendocytosis of CD86 are associated with autoimmunity.


Asunto(s)
Antígenos CD , Antígenos CD28 , Antígenos CD/metabolismo , Antígenos de Diferenciación/metabolismo , Antígeno B7-1 , Antígeno B7-2/genética , Antígenos CD28/metabolismo , Antígeno CTLA-4/genética , Moléculas de Adhesión Celular , Ligandos , Activación de Linfocitos
2.
Immunother Adv ; 2(1): ltab022, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35072168

RESUMEN

OBJECTIVES: We assessed the safety of ustekinumab (a monoclonal antibody used in psoriasis to target the IL-12 and IL-23 pathways) in a small cohort of recent-onset (<100 days of diagnosis) adults with type 1 diabetes (T1D) by conducting a pilot open-label dose-finding and mechanistic study (NCT02117765) at the University of British Columbia. METHODS: We sequentially enrolled 20 participants into four subcutaneous dosing cohorts: (i) 45 mg loading weeks 0/4/16, (ii) 45 mg maintenance weeks 0/4/16/28/40, (iii) 90 mg loading weeks 0/4/16, and (iv) 90 mg maintenance weeks 0/4/16/28/40. The primary endpoint was safety as assessed by an independent data and safety monitoring board (DSMB) but we also measured mixed meal tolerance test C-peptide, insulin use/kg, and HbA1c. Immunophenotyping was performed to assess immune cell subsets and islet antigen-specific T cell responses. RESULTS: Although several adverse events were reported, only two (bacterial vaginosis and hallucinations) were thought to be possibly related to drug administration by the study investigators. At 1 year, the 90 mg maintenance dosing cohort had the smallest mean decline in C-peptide area under the curve (AUC) (0.1 pmol/ml). Immunophenotyping showed that ustekinumab reduced the percentage of circulating Th17, Th1, and Th17.1 cells and proinsulin-specific T cells that secreted IFN-γ and IL-17A. CONCLUSION: Ustekinumab was deemed safe to progress to efficacy studies by the DSMB at doses used to treat psoriasis in adults with T1D. A 90 mg maintenance dosing schedule reduced proinsulin-specific IFN-γ and IL-17A-producing T cells. Further studies are warranted to determine if ustekinumab can prevent C-peptide AUC decline and induce a clinical response.

3.
Cell Rep ; 36(5): 109494, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34348163

RESUMEN

Regulatory T cell (Treg) therapy is a promising curative approach for a variety of immune-mediated conditions. CRISPR-based genome editing allows precise insertion of transgenes through homology-directed repair, but its use in human Tregs has been limited. We report an optimized protocol for CRISPR-mediated gene knockin in human Tregs with high-yield expansion. To establish a benchmark of human Treg dysfunction, we target the master transcription factor FOXP3 in naive and memory Tregs. Although FOXP3-ablated Tregs upregulate cytokine expression, effects on suppressive capacity in vitro manifest slowly and primarily in memory Tregs. Moreover, FOXP3-ablated Tregs retain their characteristic protein, transcriptional, and DNA methylation profile. Instead, FOXP3 maintains DNA methylation at regions enriched for AP-1 binding sites. Thus, although FOXP3 is important for human Treg development, it has a limited role in maintaining mature Treg identity. Optimized gene knockin with human Tregs will enable mechanistic studies and the development of tailored, next-generation Treg cell therapies.


Asunto(s)
Sistemas CRISPR-Cas/genética , Factores de Transcripción Forkhead/metabolismo , Técnicas de Sustitución del Gen , Linfocitos T Reguladores/inmunología , Secuencia de Bases , Metilación de ADN/genética , Reparación del ADN , Dependovirus/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Terapia de Inmunosupresión , Interleucina-2/metabolismo , Subgrupos Linfocitarios/inmunología , Fenotipo , Plásmidos/metabolismo , Factores de Tiempo , Transcripción Genética , Transgenes
4.
Front Immunol ; 11: 600000, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33363541

RESUMEN

CD80 and CD86 are expressed on antigen presenting cells and are required to engage their shared receptor, CD28, for the costimulation of CD4 T cells. It is unclear why two stimulatory ligands with overlapping roles have evolved. CD80 and CD86 also bind the regulatory molecule CTLA-4. We explored the role of CD80 and CD86 in the homeostasis and proliferation of CD4+FoxP3+ regulatory T cells (Treg), which constitutively express high levels of CTLA-4 yet are critically dependent upon CD28 signals. We observed that CD86 was the dominant ligand for Treg proliferation, survival, and maintenance of a regulatory phenotype, with higher expression of CTLA-4, ICOS, and OX40. We also explored whether CD80-CD28 interactions were specifically compromised by CTLA-4 and found that antibody blockade, clinical deficiency of CTLA-4 and CRISPR-Cas9 deletion of CTLA-4 all improved Treg survival following CD80 stimulation. Taken together, our data suggest that CD86 is the dominant costimulatory ligand for Treg homeostasis, despite its lower affinity for CD28, because CD80-CD28 interactions are selectively impaired by the high levels of CTLA-4. These data suggest a cell intrinsic role for CTLA-4 in regulating CD28 costimulation by direct competition for CD80, and indicate that that CD80 and CD86 have discrete roles in CD28 costimulation of CD4 T cells in the presence of high levels of CTLA-4.


Asunto(s)
Antígeno B7-2/inmunología , Antígenos CD28/inmunología , Antígeno CTLA-4/inmunología , Homeostasis/inmunología , Linfocitos T Reguladores/inmunología , Antígeno B7-2/genética , Antígenos CD28/genética , Antígeno CTLA-4/genética , Homeostasis/genética , Humanos , Linfocitos T Reguladores/citología
5.
JCI Insight ; 4(23)2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31671072

RESUMEN

At diagnosis, most people with type 1 diabetes (T1D) produce measurable levels of endogenous insulin, but the rate at which insulin secretion declines is heterogeneous. To explain this heterogeneity, we sought to identify a composite signature predictive of insulin secretion, using a collaborative assay evaluation and analysis pipeline that incorporated multiple cellular and serum measures reflecting ß cell health and immune system activity. The ability to predict decline in insulin secretion would be useful for patient stratification for clinical trial enrollment or therapeutic selection. Analytes from 12 qualified assays were measured in shared samples from subjects newly diagnosed with T1D. We developed a computational tool (DIFAcTO, Data Integration Flexible to Account for different Types of data and Outcomes) to identify a composite panel associated with decline in insulin secretion over 2 years following diagnosis. DIFAcTO uses multiple filtering steps to reduce data dimensionality, incorporates error estimation techniques including cross-validation and sensitivity analysis, and is flexible to assay type, clinical outcome, and disease setting. Using this novel analytical tool, we identified a panel of immune markers that, in combination, are highly associated with loss of insulin secretion. The methods used here represent a potentially novel process for identifying combined immune signatures that predict outcomes relevant for complex and heterogeneous diseases like T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Progresión de la Enfermedad , Secreción de Insulina/fisiología , Adolescente , Adulto , Niño , Biología Computacional , Femenino , Humanos , Hipoglucemiantes/farmacología , Inmunoterapia/métodos , Células Secretoras de Insulina/metabolismo , Masculino , Adulto Joven
6.
J Immunol ; 202(8): 2195-2209, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30850479

RESUMEN

Regulatory T cell (Treg) therapy is a potential curative approach for a variety of immune-mediated conditions, including autoimmunity and transplantation, in which there is pathological tissue damage. In mice, IL-33R (ST2)-expressing Tregs mediate tissue repair by producing the growth factor amphiregulin, but whether similar tissue-reparative Tregs exist in humans remains unclear. We show that human Tregs in blood and multiple tissue types produced amphiregulin, but this was neither a unique feature of Tregs nor selectively upregulated in tissues. Human Tregs in blood, tonsil, synovial fluid, colon, and lung tissues did not express ST2, so ST2+ Tregs were engineered via lentiviral-mediated overexpression, and their therapeutic potential for cell therapy was examined. Engineered ST2+ Tregs exhibited TCR-independent, IL-33-stimulated amphiregulin expression and a heightened ability to induce M2-like macrophages. The finding that amphiregulin-producing Tregs have a noneffector phenotype and are progressively lost upon TCR-induced proliferation and differentiation suggests that the tissue repair capacity of human Tregs may be an innate function that operates independently from their classical suppressive function.


Asunto(s)
Proliferación Celular , Inmunidad Innata/fisiología , Linfocitos T Reguladores/inmunología , Adulto , Femenino , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-33 , Macrófagos/citología , Macrófagos/inmunología , Masculino , Especificidad de Órganos , Linfocitos T Reguladores/citología
7.
Front Immunol ; 10: 46, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30740105

RESUMEN

Regulatory T cells (Tregs) are believed to be dysfunctional in autoimmunity. Juvenile idiopathic arthritis (JIA) and juvenile dermatomyositis (JDM) result from a loss of normal immune regulation in specific tissues such as joints or muscle and skin, respectively. Here, we discuss recent findings in regard to Treg biology in oligo-/polyarticular JIA and JDM, as well as what we can learn about Treg-related disease mechanism, treatment and biomarkers in JIA/JDM from studies of other diseases. We explore the potential use of Treg immunoregulatory markers and gene signatures as biomarkers for disease course and/or treatment success. Further, we discuss how Tregs are affected by several treatment strategies already employed in the therapy of JIA and JDM and by alternative immunotherapies such as anti-cytokine or co-receptor targeting. Finally, we review recent successes in using Tregs as a treatment target with low-dose IL-2 or cellular immunotherapy. Thus, this mini review will highlight our current understanding and identify open questions in regard to Treg biology, and how recent findings may advance biomarkers and new therapies for JIA and JDM.


Asunto(s)
Artritis Juvenil/inmunología , Artritis Juvenil/terapia , Dermatomiositis/inmunología , Dermatomiositis/terapia , Inmunoterapia Adoptiva/efectos adversos , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Autoinmunidad , Biomarcadores , Citocinas/antagonistas & inhibidores , Humanos , Ratones , Resultado del Tratamiento
8.
JCI Insight ; 4(6)2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30730852

RESUMEN

BACKGROUND: Multiple therapeutic strategies to restore immune regulation and slow type 1 diabetes (T1D) progression are in development and testing. A major challenge has been defining biomarkers to prospectively identify subjects likely to benefit from immunotherapy and/or measure intervention effects. We previously found that, compared with healthy controls, Tregs from children with new-onset T1D have an altered Treg gene signature (TGS), suggesting that this could be an immunoregulatory biomarker. METHODS: nanoString was used to assess the TGS in sorted Tregs (CD4+CD25hiCD127lo) or peripheral blood mononuclear cells (PBMCs) from individuals with T1D or type 2 diabetes, healthy controls, or T1D recipients of immunotherapy. Biomarker discovery pipelines were developed and applied to various sample group comparisons. RESULTS: Compared with controls, the TGS in isolated Tregs or PBMCs was altered in adult new-onset and cross-sectional T1D cohorts, with sensitivity or specificity of biomarkers increased by including T1D-associated SNPs in algorithms. The TGS was distinct in T1D versus type 2 diabetes, indicating disease-specific alterations. TGS measurement at the time of T1D onset revealed an algorithm that accurately predicted future rapid versus slow C-peptide decline, as determined by longitudinal analysis of placebo arms of START and T1DAL trials. The same algorithm stratified participants in a phase I/II clinical trial of ustekinumab (αIL-12/23p40) for future rapid versus slow C-peptide decline. CONCLUSION: These data suggest that biomarkers based on measuring TGSs could be a new approach to stratify patients and monitor autoimmune activity in T1D. FUNDING: JDRF (1-PNF-2015-113-Q-R, 2-PAR-2015-123-Q-R, 3-SRA-2016-209-Q-R, 3-PDF-2014-217-A-N), the JDRF Canadian Clinical Trials Network, the National Institute of Allergy and Infectious Diseases of the National Institutes of Health (UM1AI109565 and FY15ITN168), and BCCHRI.


Asunto(s)
Biomarcadores/análisis , Diabetes Mellitus Tipo 1/inmunología , Linfocitos T Reguladores/inmunología , Adolescente , Adulto , Algoritmos , Canadá , Biología Computacional , Estudios Transversales , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2 , Regulación de la Expresión Génica , Genotipo , Humanos , Inmunoterapia , Leucocitos Mononucleares , ARN Mensajero/metabolismo , Adulto Joven
9.
Eur J Immunol ; 49(2): 336-347, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30566246

RESUMEN

Obesity-associated visceral adipose tissue (AT) inflammation promotes insulin resistance and type 2 diabetes (T2D). In mice, lean visceral AT is populated with anti-inflammatory cells, notably regulatory T cells (Tregs) expressing the IL-33 receptor ST2. Conversely, obese AT contains fewer Tregs and more proinflammatory cells. In humans, however, there is limited evidence for a similar pattern of obesity-associated immunomodulation. We used flow cytometry and mRNA quantification to characterize human omental AT in 29 obese subjects, 18 of whom had T2D. Patients with T2D had increased proportions of inflammatory cells, including M1 macrophages, with positive correlations to body mass index. In contrast, Treg frequencies negatively correlated to body mass index but were comparable between T2D and non-T2D individuals. Compared to human thymic Tregs, omental AT Tregs expressed similar levels of FOXP3, CD25, IKZF2, and CTLA4, but higher levels of PPARG, CCR4, PRDM1, and CXCL2. ST2, however, was not detectable on omental AT Tregs from lean or obese subjects. This is the first comprehensive investigation into how omental AT immunity changes with obesity and T2D in humans, revealing important similarities and differences to paradigms in mice. These data increase our understanding of how pathways of immune regulation could be targeted to ameliorate AT inflammation in humans.


Asunto(s)
Tejido Adiposo/inmunología , Diabetes Mellitus Tipo 2/inmunología , Obesidad/inmunología , Paniculitis/inmunología , Linfocitos T Reguladores/inmunología , Tejido Adiposo/patología , Adulto , Antígenos de Diferenciación/inmunología , Diabetes Mellitus Tipo 2/patología , Femenino , Humanos , Inflamación/inmunología , Inflamación/patología , Masculino , Obesidad/patología , Paniculitis/patología , Linfocitos T Reguladores/patología
10.
Eur J Immunol ; 48(8): 1415-1419, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29676458

RESUMEN

We optimized a method to detect FOXP3 by mass cytometry and compared the resulting data to conventional flow cytometry. We also demonstrated the utility of the protocol to profile antigen-specific Tregs from whole blood, or Tregs from tissues such as cord blood, thymus and synovial fluid.


Asunto(s)
Recuento de Linfocito CD4/métodos , Citometría de Flujo/métodos , Factores de Transcripción Forkhead/metabolismo , Linfocitos T Reguladores/citología , Antígenos/inmunología , Humanos , Coloración y Etiquetado/métodos , Linfocitos T Reguladores/inmunología
11.
Curr Opin Immunol ; 43: 16-23, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27544816

RESUMEN

FOXP3 controls the development and function of T regulatory cells (Tregs). Autoimmunity is linked to changes in FOXP3 activity that can occur at multiple levels and lead to Treg dysfunction. For example, changes in IL-2 signaling, FOXP3 transcription and/or post-translational modifications can all contribute to loss of self-tolerance. As additional pathways of FOXP3 regulation are elucidated, new therapeutic approaches to increase Treg activity either by cell therapy or pharmacological intervention are being tested. Early success from pioneering studies of Treg-based therapy in transplantation has promoted the undertaking of similar studies in autoimmunity, with emerging evidence for the effectiveness of these approaches, particularly in the context of type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Factores de Transcripción Forkhead/metabolismo , Rechazo de Injerto/prevención & control , Inmunoterapia/métodos , Linfocitos T Reguladores/inmunología , Empalme Alternativo , Animales , Autoinmunidad , Diabetes Mellitus Tipo 1/terapia , Factores de Transcripción Forkhead/genética , Rechazo de Injerto/inmunología , Humanos , Inmunomodulación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Autotolerancia , Transducción de Señal , Linfocitos T Reguladores/trasplante , Trasplante
12.
J Clin Invest ; 126(3): 1039-51, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26854929

RESUMEN

T regulatory cells (Tregs) control immune homeostasis by preventing inappropriate responses to self and nonharmful foreign antigens. Tregs use multiple mechanisms to control immune responses, all of which require these cells to be near their targets of suppression; however, it is not known how Treg-to-target proximity is controlled. Here, we found that Tregs attract CD4+ and CD8+ T cells by producing chemokines. Specifically, Tregs produced both CCL3 and CCL4 in response to stimulation, and production of these chemokines was critical for migration of target T cells, as Tregs from Ccl3-/- mice, which are also deficient for CCL4 production, did not promote migration. Moreover, CCR5 expression by target T cells was required for migration of these cells to supernatants conditioned by Tregs. Tregs deficient for expression of CCL3 and CCL4 were impaired in their ability to suppress experimental autoimmune encephalomyelitis or islet allograft rejection in murine models. Moreover, Tregs from subjects with established type 1 diabetes were impaired in their ability to produce CCL3 and CCL4. Together, these results demonstrate a previously unappreciated facet of Treg function and suggest that chemokine secretion by Tregs is a fundamental aspect of their therapeutic effect in autoimmunity and transplantation.


Asunto(s)
Quimiocina CCL3/biosíntesis , Quimiocina CCL4/biosíntesis , Linfocitos T Reguladores/fisiología , Adolescente , Traslado Adoptivo , Animales , Proliferación Celular , Células Cultivadas , Quimiocina CCL3/metabolismo , Quimiocina CCL4/metabolismo , Quimiotaxis de Leucocito , Niño , Preescolar , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Femenino , Humanos , Lactante , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores CCR5/fisiología
13.
Diabetes ; 65(4): 1031-9, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26786322

RESUMEN

Type 1 diabetes (T1D) is caused by immune-mediated destruction of insulin-producing ß-cells. Insufficient control of autoreactive T cells by regulatory T cells (Tregs) is believed to contribute to disease pathogenesis, but changes in Treg function are difficult to quantify because of the lack of Treg-exclusive markers in humans and the complexity of functional experiments. We established a new way to track Tregs by using a gene signature that discriminates between Tregs and conventional T cells regardless of their activation states. The resulting 31-gene panel was validated with the NanoString nCounter platform and then measured in sorted CD4(+)CD25(hi)CD127(lo) Tregs from children with T1D and age-matched control subjects. By using biomarker discovery analysis, we found that expression of a combination of six genes, including TNFRSF1B (CD120b) and FOXP3, was significantly different between Tregs from subjects with new-onset T1D and control subjects, resulting in a sensitive (mean ± SD 0.86 ± 0.14) and specific (0.78 ± 0.18) biomarker algorithm. Thus, although the proportion of Tregs in peripheral blood is similar between children with T1D and control subjects, significant changes in gene expression can be detected early in disease process. These findings provide new insight into the mechanisms underlying the failure to control autoimmunity in T1D and might lead to a biomarker test to monitor Tregs throughout disease progression.


Asunto(s)
Biomarcadores , Diabetes Mellitus Tipo 1/diagnóstico , Tamizaje Masivo/métodos , Linfocitos T Reguladores/metabolismo , Transcriptoma , Adulto , Edad de Inicio , Autoinmunidad/genética , Biomarcadores/análisis , Estudios de Casos y Controles , Células Cultivadas , Niño , Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Progresión de la Enfermedad , Humanos , Masculino , Sensibilidad y Especificidad
14.
Eur J Immunol ; 45(2): 333-43, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25378065

RESUMEN

Treg cells control immune responses to self and nonharmful foreign antigens. Emerging data from animal models indicate that Treg cells function in both secondary lymphoid organs and tissues, and that these different microenvironments may contain specialized subsets of Treg cells with distinct mechanisms of action. The design of therapies for the restoration of tissue-localized immune homeostasis is dependent upon understanding how local immune responses are influenced by Treg cells in health versus disease. Here we review the current state of knowledge about human Treg cells in four locations: the skin, lung, intestine, and joint. Despite the distinct biology of these tissues, there are commonalities in the biology of their resident Treg cells, including phenotypic and functional differences from circulating Treg cells, and the presence of cytokine-producing (e.g. IL-17(+)) FOXP3(+) cells. We also highlight the challenges to studying tissue Treg cells in humans, and opportunities to use new technologies for the detailed analysis of Treg cells at the single-cell level. As emerging biological therapies are increasingly targeted toward tissue-specific effects, it is critical to understand their potential impact on local immune regulation.


Asunto(s)
Inmunidad Innata , Intestinos/inmunología , Articulaciones/inmunología , Pulmón/inmunología , Piel/inmunología , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Expresión Génica , Homeostasis/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-17/genética , Interleucina-17/inmunología , Intestinos/citología , Articulaciones/citología , Pulmón/citología , Especificidad de Órganos , Análisis de la Célula Individual , Piel/citología , Linfocitos T Reguladores/citología
15.
J Immunol ; 193(6): 2699-708, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25092890

RESUMEN

The maintenance of FOXP3 expression in CD25(hi) regulatory T cells (Tregs) is crucial to the control of inflammation and essential for successful Treg transfer therapies. Coexpression of CD25 and FOXP3 in combination with a hypomethylated region within the FOXP3 gene, called the Treg-specific demethylated region (TSDR), is considered the hallmark of stable Tregs. The TSDR is an epigenetic motif that is important for stable FOXP3 expression and is used as a biomarker to measure Treg lineage commitment. In this study, we report that, unlike in peripheral blood, CD4(+) T cell expression of CD25 and FOXP3 is frequently dissociated at the inflamed site in patients with juvenile idiopathic arthritis, which led us to question the stability of human Tregs in chronic inflammatory environments. We describe a novel CD4(+)CD127(lo)CD25(hi) human T cell population that exhibits extensive TSDR and promoter demethylation in the absence of stable FOXP3 expression. This population expresses high levels of CTLA-4 and can suppress T conventional cell proliferation in vitro. These data collectively suggest that this population may represent a chronically activated FOXP3(lo) Treg population. We show that these cells have defects in IL-2 signaling and reduced expression of a deubiquitinase important for FOXP3 stability. Clinically, the proportions of these cells within the CD25(hi) T cell subset are increased in patients with the more severe courses of disease. Our study demonstrates, therefore, that hypomethylation at the TSDR can be decoupled from FOXP3 expression in human T cells and that environment-specific breakdown in FOXP3 stability may compromise the resolution of inflammation in juvenile idiopathic arthritis.


Asunto(s)
Artritis Juvenil/inmunología , Factores de Transcripción Forkhead/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Linfocitos T Reguladores/inmunología , Adulto , Biomarcadores , Antígeno CTLA-4/biosíntesis , Proliferación Celular , Niño , Femenino , Humanos , Inflamación/inmunología , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/química , Masculino , Metilación , Transducción de Señal/inmunología , Linfocitos T Reguladores/citología , Factor de Necrosis Tumoral alfa/biosíntesis , Proteasas Ubiquitina-Específicas/genética
16.
Arthritis Rheumatol ; 66(7): 1955-60, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24692225

RESUMEN

OBJECTIVE: Granulocyte-macrophage colony stimulating factor (GM-CSF) is a potent inflammatory mediator that is responsible for recruitment and activation of innate immune cells. Recent data from murine studies have identified Th17 cells as a key source of GM-CSF and suggest that T cell-derived GM-CSF is instrumental in the induction of autoimmune disease. The present study was undertaken to analyze the expression of T cell-derived GM-CSF in the joints of patients with juvenile idiopathic arthritis (JIA) and to investigate the differentiation of Th17 cells and how this relates to GM-CSF+ T helper cells. METHODS: Synovial fluid (SF) and peripheral blood (PB) samples from 24 patients with JIA were analyzed, by flow cytometry and reverse transcription-polymerase chain reaction, for expression of GM-CSF and the Th17 marker CD161. A cytokine capture assay was used to purify Th17 cells and test the plasticity of cytokine production in response to interleukin-12 (IL-12) and IL-23. RESULTS: The frequency of GM-CSF-producing T helper cells was significantly enriched in SF mononuclear cells compared to PB mononuclear cells from the patients with JIA (24.1% of CD4+ T cells versus 2.9%) and closely correlated with the erythrocyte sedimentation rate (r(2) = 0.91, P < 0.001). Synovial GM-CSF+ T cells were predominantly CD161+ and coexpressed interferon-γ (IFNγ), but not IL-17. Culture of Th17 cells in the presence of IL-12 led to rapid up-regulation of GM-CSF and IFNγ, recapitulating the phenotype of GM-CSF-expressing cells within the joint. CONCLUSION: Our results identify a novel outcome of Th17 plasticity in humans that may account for the enrichment of GM-CSF-expressing T cells in the joints of patients with JIA. The association of GM-CSF expression with systemic inflammation highlights the potential role of Th17-related cytokines in the pathology of JIA.


Asunto(s)
Artritis Juvenil/inmunología , Artritis Juvenil/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunidad Innata/inmunología , Células Th17/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Niño , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Masculino , Líquido Sinovial/inmunología , Líquido Sinovial/metabolismo , Células Th17/citología , Células Th17/inmunología
17.
Immunology ; 142(2): 227-36, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24405357

RESUMEN

Children with systemic Juvenile Idiopathic Arthritis (sJIA), the most severe subtype of JIA, are at risk from destructive polyarthritis and growth failure, and corticosteroids as part of conventional treatment can result in osteoporosis and growth delay. In children where there is failure or toxicity from drug therapies, disease has been successfully controlled by T-cell-depleted autologous stem cell transplantation (ASCT). At present, the immunological basis underlying remission after ASCT is unknown. Immune reconstitution of T cells, B cells, natural killer cells, natural killer T cells and monocytes, in parallel with T-cell receptor (TCR) diversity by analysis of the ß variable region (TCRVb) complementarity determining region-3 (CDR3) using spectratyping and sequencing, were studied in five children with sJIA before and after ASCT. At time of follow up (mean 11.5 years), four patients remain in complete remission, while one child relapsed within 1 month of transplant. The CD8(+) TCRVb repertoire was highly oligoclonal early in immune reconstitution and re-emergence of pre-transplant TCRVb CDR3 dominant peaks was observed after transplant in certain TCRVb families. Further, re-emergence of pre-ASCT clonal sequences in addition to new sequences was identified after transplant. These results suggest that a chimeric TCR repertoire, comprising T-cell clones developed before and after transplant, can be associated with clinical remission from severe arthritis.


Asunto(s)
Artritis Juvenil/inmunología , Trasplante de Células Madre Hematopoyéticas , Depleción Linfocítica , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Artritis Juvenil/terapia , Niño , Células Clonales/inmunología , Células Clonales/metabolismo , Femenino , Humanos , Masculino , Linfocitos T/citología , Trasplante Autólogo
18.
Expert Rev Mol Med ; 15: e13, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24294966

RESUMEN

In recent years, there have been many new developments in the field of regulatory T cells (Treg), challenging the consensus on their behaviour, classification and role(s) in disease. The role Treg might play in autoimmune disease appears to be more complex than previously thought. Here, we discuss the current knowledge of regulatory T cells through animal and human research and illustrate the recent developments in childhood autoimmune arthritis (juvenile idiopathic arthritis (JIA)). Furthermore, this review summarises our understanding of the fields and assesses current and future implications for Treg in the treatment of JIA.


Asunto(s)
Artritis Juvenil/inmunología , Linfocitos T Reguladores/inmunología , Adolescente , Traslado Adoptivo , Animales , Artritis Juvenil/metabolismo , Artritis Juvenil/terapia , Niño , Citocinas/genética , Citocinas/metabolismo , Humanos , Linfocitos T Reguladores/clasificación , Linfocitos T Reguladores/metabolismo
19.
Blood ; 121(14): 2647-58, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23355538

RESUMEN

Regulatory FoxP3+CD4+ T cells (Treg) are vital for maintaining the balance between tolerance, adequate immune response, and autoimmunity. Despite this immunoregulatory role, it has been shown that Treg may also produce proinflammatory cytokines. Here we present a distinct population of Treg, defined by CD161 expression, as the major source of FoxP3+ Treg-derived proinflammatory cytokines. CD161+ Treg can be followed throughout development, from thymus and cord blood to healthy child and adult samples. CD161+ Treg display anergy, are suppressive in cocultures with conventional T cells (Tconv), and possess a predominantly demethylated Treg-specific demethylated region of the FOXP3 locus. In addition to the production of interleukin (IL) 17A, interferon γ, and IL-2, CD161+FoxP3+ cells share markers with Tconv, including expression of the transcription factors retinoic acid-related orphan receptor Cv2 (RORCv2) and T-cell-specific T-box transcription factor (Tbet). Expression of CD161 and enrichment for cytokine production are stable characteristics of CD161+ Treg upon both short- and longer-term culture in vitro. Additionally, CD161+ Treg are highly enriched within the inflammatory environment of childhood arthritis, suggesting a role in disease. Our data therefore demonstrate that CD161+FoxP3+ T cells are a novel Treg subset, found in health and disease, which display high proinflammatory potential but also exhibit hallmark Treg characteristics.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Inmunofenotipificación , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Adolescente , Antígenos CD4/inmunología , Antígenos CD4/metabolismo , Linaje de la Célula/inmunología , Niño , Preescolar , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Humanos , Lactante , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-2/inmunología , Interleucina-2/metabolismo , Masculino , Subfamilia B de Receptores Similares a Lectina de Células NK/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Células Th17/citología , Células Th17/inmunología , Células Th17/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...