Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 2194, 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38467629

RESUMO

The regulation of thymocyte development by RNA-binding proteins (RBPs) is largely unexplored. We identify 642 RBPs in the thymus and focus on Arpp21, which shows selective and dynamic expression in early thymocytes. Arpp21 is downregulated in response to T cell receptor (TCR) and Ca2+ signals. Downregulation requires Stim1/Stim2 and CaMK4 expression and involves Arpp21 protein phosphorylation, polyubiquitination and proteasomal degradation. Arpp21 directly binds RNA through its R3H domain, with a preference for uridine-rich motifs, promoting the expression of target mRNAs. Analysis of the Arpp21-bound transcriptome reveals strong interactions with the Rag1 3'-UTR. Arpp21-deficient thymocytes show reduced Rag1 expression, delayed TCR rearrangement and a less diverse TCR repertoire. This phenotype is recapitulated in Rag1 3'-UTR mutant mice harboring a deletion of the Arpp21 response region. These findings show how thymocyte-specific Arpp21 promotes Rag1 expression to enable TCR repertoire diversity until signals from the TCR terminate Arpp21 and Rag1 activities.


Assuntos
Receptores de Antígenos de Linfócitos T , Timócitos , Animais , Camundongos , Diferenciação Celular/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Timócitos/metabolismo , Timo/metabolismo
2.
Cell Metab ; 36(2): 229-239, 2024 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-38218187

RESUMO

Tissue regulatory T cells (Tregs) exert pivotal functions in both immune and metabolic regulation, maintaining local tissue homeostasis, integrity, and function. Accordingly, Tregs play a crucial role in controlling obesity-induced inflammation and supporting efficient muscle function and repair. Depending on the tissue context, Tregs are characterized by unique transcriptomes, growth, and survival factors and T cell receptor (TCR) repertoires. This functional specialization offers the potential to selectively target context-specific Treg populations, tailoring therapeutic strategies to specific niches, thereby minimizing potential side effects. Here, we discuss challenges and perspectives for niche-specific Treg targeting, which holds promise for highly efficient and precise medical interventions to combat metabolic disease.


Assuntos
Doenças Metabólicas , Linfócitos T Reguladores , Humanos , Homeostase , Doenças Metabólicas/metabolismo
3.
Cell Metab ; 35(10): 1736-1751.e7, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37734370

RESUMO

Muscle-residing regulatory T cells (Tregs) control local tissue integrity and function. However, the molecular interface connecting Treg-based regulation with muscle function and regeneration remains largely unexplored. Here, we show that exercise fosters a stable induction of highly functional muscle-residing Tregs with increased expression of amphiregulin (Areg), EGFR, and ST2. Mechanistically, we find that mice lacking IL6Rα on T cells (TKO) harbor significant reductions in muscle Treg functionality and satellite and fibro-adipogenic progenitor cells, which are required for muscle regeneration. Using exercise and sarcopenia models, IL6Rα TKO mice demonstrate deficits in Tregs, their functional maturation, and a more pronounced decline in muscle mass. Muscle injury models indicate that IL6Rα TKO mice have significant disabilities in muscle regeneration. Treg gain of function restores impaired muscle repair in IL6Rα TKO mice. Of note, pharmacological IL6R blockade in WT mice phenocopies deficits in muscle function identified in IL6Rα TKO mice, thereby highlighting the clinical implications of the findings.


Assuntos
Músculo Esquelético , Linfócitos T Reguladores , Camundongos , Animais , Linfócitos T Reguladores/metabolismo , Músculo Esquelético/metabolismo , Transdução de Sinais , Adipogenia , Receptores de Interleucina-6/metabolismo
4.
Science ; 379(6636): 1023-1030, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36893254

RESUMO

Cell-cell interactions in the central nervous system play important roles in neurologic diseases. However, little is known about the specific molecular pathways involved, and methods for their systematic identification are limited. Here, we developed a forward genetic screening platform that combines CRISPR-Cas9 perturbations, cell coculture in picoliter droplets, and microfluidic-based fluorescence-activated droplet sorting to identify mechanisms of cell-cell communication. We used SPEAC-seq (systematic perturbation of encapsulated associated cells followed by sequencing), in combination with in vivo genetic perturbations, to identify microglia-produced amphiregulin as a suppressor of disease-promoting astrocyte responses in multiple sclerosis preclinical models and clinical samples. Thus, SPEAC-seq enables the high-throughput systematic identification of cell-cell communication mechanisms.


Assuntos
Anfirregulina , Astrócitos , Comunicação Autócrina , Testes Genéticos , Técnicas Analíticas Microfluídicas , Microglia , Astrócitos/fisiologia , Testes Genéticos/métodos , Ensaios de Triagem em Larga Escala , Técnicas Analíticas Microfluídicas/métodos , Microglia/fisiologia , Anfirregulina/genética , Comunicação Autócrina/genética , Expressão Gênica , Humanos
5.
Mol Metab ; 64: 101565, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35944899

RESUMO

BACKGROUND: The highly complex pathogenesis of Type 1 Diabetes is driven by several immune cell types with both effector and regulatory characteristics, which ultimately ends in the destruction of the insulin-producing beta cells. There are multiple layers of interaction between these immune cell populations and the pancreatic islets. SCOPE OF REVIEW: In this review article, we aim to discuss important recent insights into the multiple layers of interaction between immune cell populations and the pancreatic islets. Specifically, we discuss the environment where immune and beta cell interactions occur, the key cell types and molecules involved, and the outcomes of these interactions. MAJOR CONCLUSIONS: Most of the molecular mechanisms underlying aberrant immune cell activation and impaired immune tolerance remain insufficiently understood, which hinders the development of efficient prevention and treatment strategies. In order to overcome this knowledge gap, a better understanding of the complex interactions of immune cells and beta cells, including both the underlying protective and pathogenic mechanisms is urgently required.


Assuntos
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Ilhotas Pancreáticas , Autoimunidade , Comunicação Celular , Humanos , Células Secretoras de Insulina/patologia
6.
Front Immunol ; 12: 712870, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34367177

RESUMO

Regulatory T cells (Tregs) are key mediators of peripheral self-tolerance and alterations in their frequencies, stability, and function have been linked to autoimmunity. The antigen-specific induction of Tregs is a long-envisioned goal for the treatment of autoimmune diseases given reduced side effects compared to general immunosuppressive therapies. However, the translation of antigen-specific Treg inducing therapies for the treatment or prevention of autoimmune diseases into the clinic remains challenging. In this mini review, we will discuss promising results for antigen-specific Treg therapies in allergy and specific challenges for such therapies in autoimmune diseases, with a focus on type 1 diabetes (T1D). We will furthermore discuss opportunities for antigen-specific Treg therapies in T1D, including combinatorial strategies and tissue-specific Treg targeting. Specifically, we will highlight recent advances in miRNA-targeting as a means to foster Tregs in autoimmunity. Additionally, we will discuss advances and perspectives of computational strategies for the detailed analysis of tissue-specific Tregs on the single-cell level.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Epitopos de Linfócito T/imunologia , Imunoterapia Adotiva , Linfócitos T Reguladores/imunologia , Animais , Doenças Autoimunes , Autoimunidade , Biomarcadores , Gerenciamento Clínico , Suscetibilidade a Doenças , Humanos , Imunoterapia Adotiva/métodos , Especificidade de Órgãos/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Linfócitos T Reguladores/metabolismo
7.
Front Immunol ; 12: 643544, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33679808

RESUMO

Immunodeficient mice engrafted with a functional human immune system [Human immune system (HIS) mice] have paved the way to major advances for personalized medicine and translation of immune-based therapies. One prerequisite for advancing personalized medicine is modeling the immune system of individuals or disease groups in a preclinical setting. HIS mice engrafted with peripheral blood mononuclear cells have provided fundamental insights in underlying mechanisms guiding immune activation vs. regulation in several diseases including cancer. However, the development of Graft-vs.-host disease restrains relevant long-term studies in HIS mice. Alternatively, engraftment with hematopoietic stem cells (HSCs) enables mimicking different disease stages, however, low frequencies of HSCs in peripheral blood of adults impede engraftment efficacy. One possibility to overcome those limitations is the use of patient-derived induced pluripotent stem cells (iPSCs) reprogrammed into HSCs, a challenging process which has recently seen major advances. Personalized HIS mice bridge research in mice and human diseases thereby facilitating the translation of immunomodulatory therapies. Regulatory T cells (Tregs) are important mediators of immune suppression and thereby contribute to tumor immune evasion, which has made them a central target for cancer immunotherapies. Importantly, studying Tregs in the human immune system in vivo in HIS mice will help to determine requirements for efficient Treg-targeting. In this review article, we discuss advances on personalized HIS models using reprogrammed iPSCs and review the use of HIS mice to study requirements for efficient targeting of human Tregs for personalized cancer immunotherapies.


Assuntos
Tolerância Imunológica , Imunoterapia , Modelos Imunológicos , Neoplasias , Linfócitos T Reguladores/imunologia , Evasão Tumoral , Animais , Humanos , Camundongos , Neoplasias/imunologia , Neoplasias/terapia
8.
Med ; 2(10): 1120-1137, 2021 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-34993499

RESUMO

In this review, we bring our personal experiences to showcase insulin from its breakthrough discovery as a life-saving drug 100 years ago to its uncovering as the autoantigen and potential cause of type 1 diabetes and eventually as an opportunity to prevent autoimmune diabetes. The work covers the birth of insulin to treat patients, which is now 100 years ago, the development of human insulin, insulin analogues, devices, and the way into automated insulin delivery, the realization that insulin is the primary autoimmune target of type 1 diabetes in children, novel approaches of immunotherapy using insulin for immune tolerance induction, the possible limitations of insulin immunotherapy, and an outlook how modern vaccines could remove the need for another 100 years of insulin therapy.


Assuntos
Diabetes Mellitus Tipo 1 , Insulina , Autoantígenos , Autoimunidade , Criança , Diabetes Mellitus Tipo 1/tratamento farmacológico , Humanos , Tolerância Imunológica , Insulina/uso terapêutico , Insulina Regular Humana/uso terapêutico
9.
Cancers (Basel) ; 14(1)2021 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-35008253

RESUMO

BACKGROUND: Cancer is primarily a disease of high age in humans, yet most mouse studies on cancer cachexia are conducted using young adolescent mice. Given that metabolism and muscle function change with age, we hypothesized that aging may affect cachexia progression in mouse models. METHODS: We compare tumor and cachexia development in young and old mice of three different strains (C57BL/6J, C57BL/6N, BALB/c) and with two different tumor cell lines (Lewis Lung Cancer, Colon26). Tumor size, body and organ weights, fiber cross-sectional area, circulating cachexia biomarkers, and molecular markers of muscle atrophy and adipose tissue wasting are shown. We correlate inflammatory markers and body weight dependent on age in patients with cancer. RESULTS: We note fundamental differences between mouse strains. Aging aggravates weight loss in LLC-injected C57BL/6J mice, drives it in C57BL/6N mice, and does not influence weight loss in C26-injected BALB/c mice. Glucose tolerance is unchanged in cachectic young and old mice. The stress marker GDF15 is elevated in cachectic BALB/c mice independent of age and increased in old C57BL/6N and J mice. Inflammatory markers correlate significantly with weight loss only in young mice and patients. CONCLUSIONS: Aging affects cachexia development and progression in mice in a strain-dependent manner and influences the inflammatory profile in both mice and patients. Age is an important factor to consider for future cachexia studies.

10.
Front Endocrinol (Lausanne) ; 11: 606322, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329406

RESUMO

The important role of microRNAs as major modulators of various physiological processes, including immune regulation and homeostasis, has been increasingly recognized. Consequently, aberrant miRNA expression contributes to the defective regulation of T cell development, differentiation, and function. This can result in immune activation and impaired tolerance mechanisms, which exert a cardinal function for the onset of islet autoimmunity and the progression to T1D. The specific impact of miRNAs for immune regulation and how miRNAs and their downstream targets are involved in the pathogenesis of islet autoimmunity and T1D has been investigated recently. These studies revealed that increased expression of individual miRNAs is involved in several layers of tolerance impairments, such as inefficient Treg induction and Treg instability. The targeted modulation of miRNAs using specific inhibitors, resulting in improved immune homeostasis, as well as improved methods for the targeting of miRNAs, suggest that miRNAs, especially in T cells, are a promising target for the reestablishment of immune tolerance.


Assuntos
Autoimunidade/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Ilhotas Pancreáticas/imunologia , MicroRNAs/imunologia , Animais , Regulação da Expressão Gênica , Humanos , Tolerância Imunológica , Linfócitos T Reguladores/imunologia
11.
Curr Diab Rep ; 20(9): 41, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32725277

RESUMO

PURPOSE OF REVIEW: Regulatory T cells (Tregs) are critical contributors to immune homeostasis and their dysregulation can lead to the loss of immune tolerance and autoimmune diseases like type 1 diabetes (T1D). Recent studies have highlighted microRNAs (miRNAs) as important regulators of the immune system, by fine-tuning relevant genes in various immune cell types. In this review article, we discuss recent insights into miRNA regulation of immune tolerance and activation. Specifically, we discuss how the dysregulation of miRNAs in T cells contributes to their aberrant function and the onset of islet autoimmunity, as well as their potential as targets of novel intervention strategies to interfere with autoimmune activation. RECENT FINDINGS: Several studies have shown that the dysregulation of individual miRNAs in T cells can contribute to impaired immune tolerance, contributing to onset and progression of islet autoimmunity. Importantly, the targeting of these miRNAs, including miR-92a, miR-142-3p and miR-181a, resulted in relevant effects on downstream pathways, improved Treg function and reduced islet autoimmunity in murine models. miRNAs are critical regulators of immune homeostasis and the dysregulation of individual miRNAs in T cells contributes to aberrant T cell function and autoimmunity. The specific targeting of individual miRNAs could improve Treg homeostasis and therefore limit overshooting T cell activation and islet autoimmunity.


Assuntos
Doenças Autoimunes , Diabetes Mellitus Tipo 1 , MicroRNAs , Animais , Autoimunidade , Diabetes Mellitus Tipo 1/genética , Humanos , Camundongos , MicroRNAs/genética , Linfócitos T Reguladores
12.
Nat Commun ; 10(1): 5697, 2019 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-31836704

RESUMO

In type 1 diabetes, the appearance of islet autoantibodies indicates the onset of islet autoimmunity, often many years before clinical symptoms arise. While T cells play a major role in the destruction of pancreatic beta cells, molecular underpinnings promoting aberrant T cell activation remain poorly understood. Here, we show that during islet autoimmunity an miR142-3p/Tet2/Foxp3 axis interferes with the efficient induction of regulatory T (Treg) cells, resulting in impaired Treg stability in mouse and human. Specifically, we demonstrate that miR142-3p is induced in islet autoimmunity and that its inhibition enhances Treg induction and stability, leading to reduced islet autoimmunity in non-obese diabetic mice. Using various cellular and molecular approaches we identify Tet2 as a direct target of miR142-3p, thereby linking high miR142-3p levels to epigenetic remodeling in Tregs. These findings offer a mechanistic model where during islet autoimmunity miR142-3p/Tet2-mediated Treg instability contributes to autoimmune activation and progression.


Assuntos
Autoimunidade/genética , Proteínas de Ligação a DNA/genética , Diabetes Mellitus Tipo 1/imunologia , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas/genética , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Criança , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/genética , Dioxigenases , Epigênese Genética/imunologia , Regulação da Expressão Gênica/imunologia , Técnicas de Silenciamento de Genes , Humanos , Ilhotas Pancreáticas/imunologia , Masculino , Camundongos Knockout , MicroRNAs/agonistas , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Cultura Primária de Células
13.
Mol Metab ; 27S: S122-S128, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31500823

RESUMO

BACKGROUND: microRNAs (miRNAs) have emerged as critical contributors to immune regulation and homeostasis, and their dysregulation is involved in the aberrant differentiation and function of T cell subsets. In type 1 diabetes (T1D), the clinically overt disease is preceded by a presymptomatic phase which is marked by the presence of islet autoantibodies while the individual is still normoglycemic. Recent analyses revealed impaired regulatory T (Treg) cell induction from naive CD4+ T cells during this early phase of autoimmunity. SCOPE OF THE REVIEW: In this review article, we aim to discuss important recent insights into miRNA regulation of immune homeostasis and activation. Specifically, we highlight the role of miRNAs as biomarkers in autoimmunity and T1D as well as the contribution of specific miRNAs and their downstream pathways to the onset and progression of islet immunity. Furthermore, we focus on critical next steps required to establish miRNAs as biomarkers to predict disease onset and progression and as novel targets of future prevention and treatment strategies to control autoimmunity. MAJOR CONCLUSIONS: Several recent studies have provided considerable insight into the miRNA regulation of immune homeostasis and how dysregulated miRNAs contribute to onset and progression of islet autoimmunity. Specifically, high levels of individual miRNAs such as miR92a and miR181a are involved in impaired Treg induction during the onset of islet autoimmunity, thereby contributing to disease pathogenesis. The recent advancements in the field suggest miRNAs as potential biomarkers for islet autoimmunity and their direct targeting, especially in a T cell-specific manner, could contribute to the reestablishment of immune homeostasis and ultimately interfere with the onset of islet autoimmunity.


Assuntos
Autoimunidade/genética , Diabetes Mellitus Tipo 1/imunologia , Ilhotas Pancreáticas/imunologia , MicroRNAs/genética , MicroRNAs/imunologia , Linfócitos T Reguladores/imunologia , Animais , Humanos
14.
Mol Metab ; 28: 73-82, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31427184

RESUMO

OBJECTIVE: Obesity and type-2 diabetes (T2D) are metabolic diseases that represent a critical health problem worldwide. Metabolic disease is differentially associated with fat distribution, while visceral white adipose tissue (VAT) is particularly prone to obesity-associated inflammation. Next to their canonical function of immune suppression, regulatory T cells (Tregs) are key in controlling adipose tissue homeostasis. Towards understanding the molecular underpinnings of metabolic disease, we focus on how environmental-metabolic stimuli impinge on the functional interplay between Tregs and adipose tissue. Here, cold exposure or beta3-adrenergic signaling are a promising tool to increase energy expenditure by activating brown adipose tissue, as well as by reducing local inflammation within fat depots by supporting immunosuppressive Tregs. However, in humans, the underlying mechanisms that enable the environmental-immune crosstalk in the periphery and in the respective tissue remain currently unknown. METHODS: We used combinatorial approaches of next generation humanized mouse models and in vitro and in vivo experiments together with beta3-adrenergic stimulation to dissect the underlying mechanisms of human Treg induction exposed to environmental stimuli such as cold. To test the translational relevance of our findings, we analyzed samples from the FREECE study in which human subjects were exposed to individualized cooling protocols. Samples were analyzed ex vivo and after in vitro Treg induction using qRT-PCR, immunofluorescence, as well as with multicolor flow cytometry and cell sorting. RESULTS: In vivo application of the beta3-adrenergic receptor agonist mirabegron in humanized mice induced thermogenesis and improved the Treg induction capacity of naïve T cells isolated from these animals. Using samples from the human FREECE study, we demonstrate that a short-term cold stimulus supports human Treg induction in vitro and in vivo. Mechanistically, we identify BORCS6 encoding the Ragulator-interacting protein C17orf59 to be significantly induced in human CD4+ T cells upon short-term cold exposure. Strong mTOR signaling is known to limit successful Treg induction and thus likely by interfering with mTOR activation at lysosomal surfaces, C17orf59 improves the Treg induction capacity of human naïve T cells upon cold exposure. CONCLUSIONS: These novel insights into the molecular underpinnings of human Treg induction suggest an important role of Tregs in linking environmental stimuli with adipose tissue function and metabolic diseases. Moreover, these discoveries shed new light on potential approaches towards tailored anti-inflammatory concepts that support human adipose tissue homeostasis by enabling Tregs.


Assuntos
Temperatura Baixa , Linfócitos T Reguladores/imunologia , Acetanilidas/farmacologia , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/imunologia , Agonistas Adrenérgicos beta/farmacologia , Adulto , Animais , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Receptores Adrenérgicos beta/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Tiazóis/farmacologia , Adulto Jovem
15.
Cardiovasc Res ; 115(9): 1385-1392, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31228191

RESUMO

Inflammation is an important driver of atherosclerosis, and the favourable outcomes of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial revealed the large potential of anti-inflammatory drugs for the treatment of cardiovascular disease, especially in patients with a pro-inflammatory constitution. However, the complex immune reactions driving inflammation in the vascular wall in response to an atherosclerotic microenvironment are still being unravelled. Novel insights into the cellular processes driving immunity and inflammation revealed that alterations in intracellular metabolic pathways are strong drivers of survival, growth, and function of immune cells. Therefore, this position paper presents a brief overview of the recent developments in the immunometabolism field, focusing on its role in atherosclerosis. We will also highlight the potential impact of immunometabolic markers and targets in clinical cardiovascular medicine.


Assuntos
Artérias/imunologia , Aterosclerose/imunologia , Metabolismo Energético/imunologia , Sistema Imunitário/imunologia , Imunomodulação , Inflamação/imunologia , Animais , Artérias/metabolismo , Artérias/fisiopatologia , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Consenso , Humanos , Sistema Imunitário/metabolismo , Sistema Imunitário/fisiopatologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Placa Aterosclerótica , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/metabolismo
16.
Front Immunol ; 9: 1729, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30083169

RESUMO

T follicular helper (TFH) cells are an integral part of humoral immunity by providing help to B cells to produce high-affinity antibodies. The TFH precursor compartment circulates in the blood and TFH cell dysregulation is implied in various autoimmune diseases including type 1 diabetes (T1D). Symptomatic T1D is preceded by a preclinical phase (indicated by the presence of islet autoantibodies) with a highly variable progression time to the symptomatic disease. This heterogeneity points toward differences in immune activation in children with a fast versus slow progressor phenotype. In the context of T1D, previous studies on TFH cells have mainly focused on the clinically active state of the disease. In this review article, we aim to specifically discuss recent insights on TFH cells in human islet autoimmunity before the onset of symptomatic T1D. Furthermore, we will highlight advances in the field of TFH differentiation and function during human islet autoimmunity. Specifically, we will focus on the regulation of TFH cells by microRNAs (miRNAs), as well as on the potential use of miRNAs as biomarkers to predict disease progression time and as future drug targets to interfere with autoimmune activation.

17.
Sci Transl Med ; 10(422)2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29298866

RESUMO

Molecular checkpoints that trigger the onset of islet autoimmunity or progression to human type 1 diabetes (T1D) are incompletely understood. Using T cells from children at an early stage of islet autoimmunity without clinical T1D, we find that a microRNA181a (miRNA181a)-mediated increase in signal strength of stimulation and costimulation links nuclear factor of activated T cells 5 (NFAT5) with impaired tolerance induction and autoimmune activation. We show that enhancing miRNA181a activity increases NFAT5 expression while inhibiting FOXP3+ regulatory T cell (Treg) induction in vitro. Accordingly, Treg induction is improved using T cells from NFAT5 knockout (NFAT5ko) animals, whereas altering miRNA181a activity does not affect Treg induction in NFAT5ko T cells. Moreover, high costimulatory signals result in phosphoinositide 3-kinase (PI3K)-mediated NFAT5, which interferes with FoxP3+ Treg induction. Blocking miRNA181a or NFAT5 increases Treg induction in murine and humanized models and reduces murine islet autoimmunity in vivo. These findings suggest targeting miRNA181a and/or NFAT5 signaling for the development of innovative personalized medicines to limit islet autoimmunity.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , MicroRNAs/metabolismo , Fatores de Transcrição NFATC/metabolismo , Animais , Antagomirs , Linfócitos T CD4-Positivos/metabolismo , Diabetes Mellitus Tipo 1/genética , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Imunogenética , Camundongos , Camundongos Mutantes , MicroRNAs/genética , Fatores de Transcrição NFATC/genética
18.
Cell Metab ; 26(3): 475-492.e7, 2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28877454

RESUMO

Obesity and type 2 diabetes are associated with metabolic defects and adipose tissue inflammation. Foxp3+ regulatory T cells (Tregs) control tissue homeostasis by counteracting local inflammation. However, if and how T cells interlink environmental influences with adipocyte function remains unknown. Here, we report that enhancing sympathetic tone by cold exposure, beta3-adrenergic receptor (ADRB3) stimulation or a short-term high-calorie diet enhances Treg induction in vitro and in vivo. CD4+ T cell proteomes revealed higher expression of Foxp3 regulatory networks in response to cold or ADRB3 stimulation in vivo reflecting Treg induction. Specifically, Ragulator-interacting protein C17orf59, which limits mTORC1 activity, was upregulated in CD4+ T cells by either ADRB3 stimulation or cold exposure, suggesting contribution to Treg induction. By loss- and gain-of-function studies, including Treg depletion and transfers in vivo, we demonstrated that a T cell-specific Stat6/Pten axis links cold exposure or ADRB3 stimulation with Foxp3+ Treg induction and adipose tissue function. Our findings offer a new mechanistic model in which tissue-specific Tregs maintain adipose tissue function.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fator de Transcrição STAT6/metabolismo , Animais , Temperatura Baixa , Feminino , Fatores de Transcrição Forkhead/metabolismo , Camundongos Endogâmicos BALB C , Proteoma/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Linfócitos T Reguladores/metabolismo , Proteína Desacopladora 1/metabolismo
19.
Eur J Immunol ; 47(11): 1867-1874, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28849586

RESUMO

Obesity and type-2 diabetes (T2D) are associated with metabolic defects and inflammatory processes in fat depots. FoxP3+ regulatory T cells (Tregs) control immune tolerance, and have an important role in controlling tissue-specific inflammation. In this mini-review we will discuss current insights into how cross-talk between T cells and adipose tissue shapes the inflammatory environment in obesity-associated metabolic diseases, focusing on the role of CD4+ T cells and Tregs. We will also highlight potential opportunities for how the immunoregulatory properties of Tregs could be harnessed to control inflammation in obesity and T2D and emphasize the critical need for more research on humans to establish mechanisms that are conserved in both mice and humans.


Assuntos
Tecido Adiposo/imunologia , Receptor Cross-Talk/imunologia , Linfócitos T Reguladores/imunologia , Animais , Diabetes Mellitus Tipo 2/imunologia , Humanos , Camundongos , Obesidade/imunologia
20.
J Exp Med ; 214(7): 2157-2159, 2017 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-28536240

RESUMO

In this issue of JEM, Bergman et al. (https://doi.org/10.1084/jem.20160471) challenge the data published in our previous JEM paper on the preventive effect of tolerogenic vaccination with a strong agonist insulin mimetope in type 1 diabetes. Here, we provide a response to these data and suggest that appropriate subimmunogenic conditions are required to induce Foxp3+ regulatory T cell conversion.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Insulina/imunologia , Vacinação/métodos , Animais , Epitopos/imunologia , Everolimo/farmacologia , Feminino , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Microbioma Gastrointestinal/imunologia , Humanos , Tolerância Imunológica/imunologia , Imunossupressores/farmacologia , Camundongos Endogâmicos NOD , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/imunologia , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...