Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 9(5)2024 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-38341270

RESUMO

Tregs can facilitate transplant tolerance and attenuate autoimmune and inflammatory diseases. Therefore, it is clinically relevant to stimulate Treg expansion and function in vivo and to create therapeutic Treg products in vitro. We report that TNF receptor 2 (TNFR2) is a unique costimulus for naive, thymus-derived Tregs (tTregs) from human blood that promotes their differentiation into nonlymphoid tissue-resident (NLT-resident) effector Tregs, without Th-like polarization. In contrast, CD28 costimulation maintains a lymphoid tissue-resident (LT-resident) Treg phenotype. We base this conclusion on transcriptome and proteome analysis of TNFR2- and CD28-costimulated CD4+ tTregs and conventional T cells (Tconvs), followed by bioinformatic comparison with published transcriptomic Treg signatures from NLT and LT in health and disease, including autoimmunity and cancer. These analyses illuminate that TNFR2 costimulation promoted tTreg capacity for survival, migration, immunosuppression, and tissue regeneration. Functional studies confirmed improved migratory ability of TNFR2-costimulated tTregs. Flow cytometry validated the presence of the TNFR2-driven tTreg signature in effector/memory Tregs from the human placenta, as opposed to blood. Thus, TNFR2 can be exploited as a driver of NLT-resident tTreg differentiation for adoptive cell therapy or antibody-based immunomodulation in human disease.


Assuntos
Receptores Tipo II do Fator de Necrose Tumoral , Linfócitos T Reguladores , Humanos , Antígenos CD28 , Linfócitos , Timo
2.
Sci Rep ; 12(1): 20268, 2022 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-36434024

RESUMO

The CD4+ regulatory T (Treg) cell lineage, defined by FOXP3 expression, comprises thymus-derived (t)Treg cells and peripherally induced (p)Treg cells. As a model for Treg cells, studies employ TGF-ß-induced (i)Treg cells generated from CD4+ conventional T (Tconv) cells in vitro. Here, we describe how human iTreg cells relate to human blood-derived tTreg and Tconv cells according to proteomic analysis. Each of these cell populations had a unique protein expression pattern. iTreg cells had very limited overlap in protein expression with tTreg cells, regardless of cell activation status and instead shared signaling and metabolic proteins with Tconv cells. tTreg cells had a uniquely modest response to CD3/CD28-mediated stimulation. As a benchmark, we used a previously defined proteomic signature that discerns ex vivo naïve and effector Treg cells from Tconv cells and includes conserved Treg cell properties. iTreg cells largely lacked this Treg cell core signature and highly expressed e.g. STAT4 and NFATC2, which may contribute to inflammatory responses. We also used a proteomic signature that distinguishes ex vivo effector Treg cells from Tconv cells and naïve Treg cells. iTreg cells contained part of this effector Treg cell signature, suggesting acquisition of pTreg cell features. In conclusion, iTreg cells are distinct from tTreg cells and share limited features with ex vivo Treg cells at the proteomic level.


Assuntos
Linfócitos T Reguladores , Fator de Crescimento Transformador beta , Humanos , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Proteômica , Fatores de Transcrição Forkhead/metabolismo , Timo/metabolismo
3.
Front Immunol ; 13: 881166, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35844585

RESUMO

CD4+ conventional T cells (Tconvs) mediate adaptive immune responses, whereas regulatory T cells (Tregs) suppress those responses to safeguard the body from autoimmunity and inflammatory diseases. The opposing activities of Tconvs and Tregs depend on the stage of the immune response and their environment, with an orchestrating role for cytokine- and costimulatory receptors. Nutrient availability also impacts T-cell functionality via metabolic and biosynthetic processes that are largely unexplored. Many data argue that costimulation by Tumor Necrosis Factor Receptor 2 (TNFR2) favors support of Treg over Tconv responses and therefore TNFR2 is a key clinical target. Here, we review the pertinent literature on this topic and highlight the newly identified role of TNFR2 as a metabolic regulator for thymus-derived (t)Tregs. We present novel transcriptomic and metabolomic data that show the differential impact of TNFR2 on Tconv and tTreg gene expression and reveal distinct metabolic impact on both cell types.


Assuntos
Receptores Tipo II do Fator de Necrose Tumoral , Linfócitos T Reguladores , Citocinas/metabolismo , Imunidade , Contagem de Linfócitos , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo
4.
Cell Rep ; 37(7): 110013, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34788605

RESUMO

Autotaxin (ATX; ENPP2) produces lysophosphatidic acid (LPA) that regulates multiple biological functions via cognate G protein-coupled receptors LPAR1-6. ATX/LPA promotes tumor cell migration and metastasis via LPAR1 and T cell motility via LPAR2, yet its actions in the tumor immune microenvironment remain unclear. Here, we show that ATX secreted by melanoma cells is chemorepulsive for tumor-infiltrating lymphocytes (TILs) and circulating CD8+ T cells ex vivo, with ATX functioning as an LPA-producing chaperone. Mechanistically, T cell repulsion predominantly involves Gα12/13-coupled LPAR6. Upon anti-cancer vaccination of tumor-bearing mice, ATX does not affect the induction of systemic T cell responses but, importantly, suppresses tumor infiltration of cytotoxic CD8+ T cells and thereby impairs tumor regression. Moreover, single-cell data from melanoma tumors are consistent with intratumoral ATX acting as a T cell repellent. These findings highlight an unexpected role for the pro-metastatic ATX-LPAR axis in suppressing CD8+ T cell infiltration to impede anti-tumor immunity, suggesting new therapeutic opportunities.


Assuntos
Linfócitos do Interstício Tumoral/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Quimiotaxia/fisiologia , Feminino , Humanos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias , Diester Fosfórico Hidrolases/fisiologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais/fisiologia , Microambiente Tumoral
5.
Nat Metab ; 2(10): 1046-1061, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32958937

RESUMO

Following activation, conventional T (Tconv) cells undergo an mTOR-driven glycolytic switch. Regulatory T (Treg) cells reportedly repress the mTOR pathway and avoid glycolysis. However, here we demonstrate that human thymus-derived Treg (tTreg) cells can become glycolytic in response to tumour necrosis factor receptor 2 (TNFR2) costimulation. This costimulus increases proliferation and induces a glycolytic switch in CD3-activated tTreg cells, but not in Tconv cells. Glycolysis in CD3-TNFR2-activated tTreg cells is driven by PI3-kinase-mTOR signalling and supports tTreg cell identity and suppressive function. In contrast to glycolytic Tconv cells, glycolytic tTreg cells do not show net lactate secretion and shuttle glucose-derived carbon into the tricarboxylic acid cycle. Ex vivo characterization of blood-derived TNFR2hiCD4+CD25hiCD127lo effector T cells, which were FOXP3+IKZF2+, revealed an increase in glucose consumption and intracellular lactate levels, thus identifying them as glycolytic tTreg cells. Our study links TNFR2 costimulation in human tTreg cells to metabolic remodelling, providing an additional avenue for drug targeting.


Assuntos
Glicólise/efeitos dos fármacos , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Linfócitos T Reguladores/metabolismo , Complexo CD3/metabolismo , Ciclo do Ácido Cítrico/efeitos dos fármacos , Glucose/metabolismo , Glucose/farmacologia , Humanos , Ácido Láctico/sangue , Ácido Láctico/metabolismo , Metaboloma , Fosfatidilinositol 3-Quinases/metabolismo , RNA/química , Receptores Tipo II do Fator de Necrose Tumoral/efeitos dos fármacos , Análise de Sequência de RNA , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
6.
J Immunol ; 204(12): 3139-3148, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32366581

RESUMO

FOXP3-expressing regulatory T (Treg) cells safeguard immunological tolerance. Treg cells can be generated during thymic development (called thymic Treg [tTreg] cells) or derived from mature conventional CD4+ T cells that underwent TGF-ß-mediated conversion in the periphery (called peripheral Treg [pTreg] cells). Murine studies have shown that tTreg cells exhibit strong lineage fidelity, whereas pTreg cells can revert into conventional CD4+ T cells. Their stronger lineage commitment makes tTreg cells the safest cells to use in adoptive cell therapy, increasingly used to treat autoimmune and inflammatory disorders. Markers to distinguish human tTreg cells from pTreg cells have, however, not been found. Based on combined proteomic and transcriptomic approaches, we report that the Ig superfamily protein GPA33 is expressed on a subset of human Treg cells. GPA33 is acquired late during tTreg cell development but is not expressed on TGF-ß-induced Treg cells. GPA33 identifies Treg cells in human blood that lack the ability to produce effector cytokines (IL-2, IFN-γ, IL-17), regardless of differentiation stage. GPA33high Treg cells universally express the transcription factor Helios that preferentially marks tTreg cells and can robustly and stably be expanded in vitro even without rapamycin. Expanded GPA33high Treg cells are suppressive, unable to produce proinflammatory cytokines, and exhibit the epigenetic modifications of the FOXP3 gene enhancer CNS2, necessary for indelible expression of this critical transcription factor. Our findings thus suggest that GPA33 identifies human tTreg cells and provide a strategy to isolate such cells for safer and more efficacious adoptive cell therapy.


Assuntos
Biomarcadores/metabolismo , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Cultivadas , Citocinas/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Humanos , Tolerância Imunológica/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Ativação Linfocitária/imunologia , Fator de Crescimento Transformador beta/metabolismo
7.
Cell Rep ; 30(12): 4110-4123.e4, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32209472

RESUMO

Within lymph nodes (LNs), T follicular helper (TFH) cells help B cells to produce antibodies, which can either be protective or autoreactive. Here, we demonstrate that murine LN stromal cells (LNSCs) suppress the formation of autoreactive TFH cells in an antigen-specific manner, thereby significantly reducing germinal center B cell responses directed against the same self-antigen. Mechanistically, LNSCs express and present self-antigens in major histocompatibility complex (MHC) class II, leading to the conversion of naive CD4+ T cells into T regulatory (TREG) cells in an interleukin-2 (IL-2)-dependent manner. Upon blockade of TREG cells, using neutralizing IL-2 antibodies, autoreactive TFH cells are allowed to develop. We conclude that the continuous presentation of self-antigens by LNSCs is critical to generate antigen-specific TREG cells, thereby repressing the formation of TFH cells and germinal center B cell responses. Our findings uncover the ability of LNSCs to suppress the early activation of autoreactive immune cells and maintain peripheral tolerance.


Assuntos
Linfócitos B/imunologia , Epitopos/imunologia , Linfonodos/citologia , Linfócitos T Reguladores/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos CD/metabolismo , Autoantígenos/imunologia , Centro Germinativo/imunologia , Humanos , Interleucina-2/metabolismo , Camundongos Endogâmicos C57BL , Células Estromais/citologia
8.
Front Immunol ; 9: 2654, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30505306

RESUMO

Resident memory T cells (TRM) inhabit peripheral tissues and are critical for protection against localized infections. Recently, it has become evident that CD103+ TRM are not only important in combating secondary infections, but also for the elimination of tumor cells. In several solid cancers, intratumoral CD103+CD8+ tumor infiltrating lymphocytes (TILs), with TRM properties, are a positive prognostic marker. To better understand the role of TRM in tumors, we performed a detailed characterization of CD8+ and CD4+ TIL phenotype and functional properties in non-small cell lung cancer (NSCLC). Frequencies of CD8+ and CD4+ T cell infiltrates in tumors were comparable, but we observed a sharp contrast in TRM ratios compared to surrounding lung tissue. The majority of both CD4+ and CD8+ TILs expressed CD69 and a subset also expressed CD103, both hallmarks of TRM. While CD103+CD8+ T cells were enriched in tumors, CD103+CD4+ T cell frequencies were decreased compared to surrounding lung tissue. Furthermore, CD103+CD4+ and CD103+CD8+ TILs showed multiple characteristics of TRM, such as elevated expression of CXCR6 and CD49a, and decreased expression of T-bet and Eomes. In line with the immunomodulatory role of the tumor microenvironment, CD8+ and CD4+ TILs expressed high levels of inhibitory receptors 2B4, CTLA-4, and PD-1, with the highest levels found on CD103+ TILs. Strikingly, CD103+CD4+ TILs were the most potent producers of TNF-α and IFN-γ, while other TIL subsets lacked such cytokine production. Whereas, CD103+CD4+PD-1low TILs produced the most effector cytokines, CD103+CD4+PD-1++ and CD69+CD4+PD-1++ TILs produced CXCL13. Furthermore, a large proportion of TILs expressed co-stimulatory receptors CD27 and CD28, unlike lung TRM, suggesting a less differentiated phenotype. Agonistic triggering of these receptors improved cytokine production of CD103+CD4+ and CD69+CD8+ TILs. Our findings thus provide a rationale to target CD103+CD4+ TILs and add co-stimulation to current therapies to improve the efficacy of immunotherapies and cancer vaccines.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Memória Imunológica/genética , Neoplasias Pulmonares/imunologia , Linfócitos do Interstício Tumoral/imunologia , Fenótipo , Microambiente Tumoral/imunologia , Idoso , Antígenos CD/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Citocinas/metabolismo , Feminino , Granzimas/metabolismo , Humanos , Cadeias alfa de Integrinas/metabolismo , Integrina alfa1/metabolismo , Pulmão/imunologia , Neoplasias Pulmonares/metabolismo , Masculino , Pessoa de Meia-Idade , Prognóstico , Receptor de Morte Celular Programada 1/metabolismo , Receptores CXCR6/metabolismo
9.
Immunity ; 48(5): 1046-1059.e6, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29752063

RESUMO

To obtain a molecular definition of regulatory T (Treg) cell identity, we performed proteomics and transcriptomics on various populations of human regulatory and conventional CD4+ T (Tconv) cells. A protein expression signature was identified that defines all Treg cells, and another signature that defines effector Treg cells. These signatures could not be extrapolated from transcriptome data. Unique cell-biological and metabolic features in Treg cells were defined, as well as specific adaptations in cytokine, TCR, and costimulatory receptor signaling pathways. One such adaptation-selective STAT4 deficiency-prevented destabilization of Treg cell identity and function by inflammatory cytokines, while these signals could still induce critical transcription factors and homing receptors via other pathways. Furthermore, our study revealed surface markers that identify FOXP3+CD4+ T cells with distinct functional properties. Our findings suggest that adaptation in signaling pathways protect Treg cell identity and present a resource for further research into Treg cell biology.


Assuntos
Adaptação Fisiológica , Proteômica/métodos , Transdução de Sinais , Linfócitos T Reguladores/metabolismo , Western Blotting , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Citocinas/metabolismo , Citometria de Fluxo , Fatores de Transcrição Forkhead/metabolismo , Células HEK293 , Humanos , Espectrometria de Massas , Receptores de Antígenos de Linfócitos T/metabolismo
10.
J Innate Immun ; 9(6): 609-620, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28889122

RESUMO

Intestinal epithelial cells (IEC) drive regulatory T cell (Treg) responses by promoting the differentiation of aldehyde dehydrogenase (ALDH)-expressing CD103+ dendritic cells (DC). Apical stimulation of TLR9 by CpG DNA on IEC supports galectin-9 expression by IEC, which is promoted by short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides (GF). While galectin-9 can induce the maturation of monocyte-derived DC (moDC), the contribution of galectin-9 on the induction of ALDH activity in DC is not known. To this end, DC were stimulated with galectin-9, and ALDH activity and the expression of CD103 were assessed. ALDH activity was increased by moDC exposed to galectin-9, while the expression of CD103 remained unaltered. Galectin-9 secreted by IEC apically exposed to CpG DNA and GF enhanced ALDH activity, but not CD103 expression by moDC, which was abrogated upon galectin-9 neutralization. Similar observations were found in murine GM-CSF-cultured bone marrow-derived DC (BMDC). Using Flt3L-cultured BMDC and ex vivo murine splenic DC, it was observed that galectin-9 only enhanced ALDH activity in the presence of GM-CSF in CD103- cells. The induction of ALDH activity in BMDC was dependent on p38 and PI3K signaling. These data indicate a novel role for galectin-9 in modulating innate immunity by inducing ALDH activity in DC.


Assuntos
Aldeído Desidrogenase/metabolismo , Células Dendríticas/imunologia , Galectinas/metabolismo , Mucosa Intestinal/patologia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Neutralizantes/metabolismo , Antígenos CD/metabolismo , Diferenciação Celular , Ativação Enzimática , Galectinas/imunologia , Células HT29 , Humanos , Cadeias alfa de Integrinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Tirosina Quinase 3 Semelhante a fms/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
J Leukoc Biol ; 102(1): 105-115, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28495789

RESUMO

Dietary intervention with short-chain galacto-oligosaccharides (scGOS), long-chain fructo-oligosaccharides (lcFOS) and Bifidobacterium breve M-16V (Bb) (GF/Bb) suppresses food allergic symptoms in mice, potentially via intestinal epithelial cell (IEC)-derived galectin-9. Furthermore, in vitro studies showed galacto- and fructo-oligosaccharides (GF) to enhance the immunomodulatory capacity of a TLR9 ligand representing bacterial CpG DNA when exposed to IEC. In this study, we investigated whether GF/Bb modulates dendritic cells (DCs) and subsequent Th2 and regulatory T cell (Treg) frequency in the small intestinal lamina propria (SI-LP). BALB/c mice were fed GF/Bb during oral OVA sensitization. DC and T cell phenotype were determined in SI-LP mononuclear cells using flow cytometry. Murine bone marrow-derived DCs (BMDCs) were exposed to recombinant galectin-9 or human monocyte-derived DCs (moDCs) and were cultured in IEC-conditioned medium from GF and TLR9 ligand-exposed HT-29 cells. GF/Bb reduced allergic symptoms and enhanced serum galectin-9 levels, while suppressing activation, restoring phagocytic capacity, and normalizing CD103 expression of SI-LP DCs of OVA-allergic mice. In vitro, galectin-9 suppressed LPS-induced activation markers and cytokine secretion by BMDCs, and IEC-conditioned medium suppressed moDC activation in a galectin-9-dependent manner. Besides suppression of SI-LP DC activation, dietary GF/Bb also lowered the frequency of activated Th2 cells, while enhancing Treg in the SI-LP of OVA-allergic mice compared to the control diet. Dietary intervention with GF/Bb enhances galectin-9 and suppresses allergic symptoms of OVA-allergic mice in association with reduced intestinal DC and Th2 activation and increased Treg frequency in these mice.


Assuntos
Bifidobacterium breve , Células Dendríticas/imunologia , Hipersensibilidade Alimentar , Intestinos/imunologia , Oligossacarídeos/farmacologia , Administração Oral , Animais , Linhagem Celular , Células Dendríticas/patologia , Feminino , Hipersensibilidade Alimentar/imunologia , Hipersensibilidade Alimentar/patologia , Hipersensibilidade Alimentar/terapia , Intestinos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Células Th2/imunologia , Células Th2/patologia , Receptor Toll-Like 9/imunologia
12.
Clin Immunol ; 156(1): 28-35, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25450337

RESUMO

Highly active antiretroviral therapy (HAART) for the treatment of HIV infection sustains viral suppression and increases CD4(+) T cells in HIV patients. However, in 10-25% of subjects, known as immunological non-responders (INRs), HAART does not increase CD4 count. We investigated a potential role for galectin-9 and TIM-3 in INRs as galectin-9 and TIM-3 have been described to modulate NK and T cell function. PBMCs were isolated from healthy controls, HIV immunological responders (IRs, >350CD4(+) cells/mm(3)) and HIV INRs (<350CD4(+) cells/mm(3)) and TIM-3 and galectin-9 expressions on NK cell subsets and CD4(+) T cells were assessed. HIV INRs and HIV IRs showed increased galectin-9 expression on CD16(-)CD56(bright) and CD16(+)CD56(+) NK cells and CD4(+) T cells. Only HIV INRs showed a reduced frequency of TIM-3-expressing CD16(+)CD56(+), CD16(+)CD56(-) and CD4(+) cells, which correlated with low peripheral CD4 counts. These data suggest that TIM-3 expression may be characteristic for HIV INRs.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Regulação da Expressão Gênica , Infecções por HIV/imunologia , Células Matadoras Naturais/imunologia , Proteínas de Membrana/genética , Adulto , Idoso , Linfócitos T CD4-Positivos/citologia , Feminino , Citometria de Fluxo , Galectinas/genética , Galectinas/metabolismo , Receptor Celular 2 do Vírus da Hepatite A , Humanos , Células Matadoras Naturais/citologia , Masculino , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade
13.
Clin Immunol ; 154(2): 91-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25058467

RESUMO

Alterations in the gut microbiota composition are associated with food allergy. Toll-like receptors (TLRs) respond to microbial stimuli. We studied the effects of the ligation of TLRs on intestinal epithelial cells (IECs) in preventing an allergic effector response. IEC monolayers (T84 cells) were co-cultured with CD3/28-activated PBMCs from healthy controls or atopic patients and simultaneously apically exposed to TLR2, TLR4 or TLR9 ligands. The barrier integrity of T84 cell monolayers was significantly reduced upon co-culture with PBMCs of food allergic subjects compared to healthy subjects. Apical exposure of IECs to a TLR9 ligand prevented PBMC-induced epithelial barrier disruption. Using PBMCs from food allergic subjects, apical TLR9 activation on IECs increased the IFN-γ/IL-13 and IL-10/IL-13 ratio, while suppressing pro-inflammatory IL-6, IL-8 and TNF-α production in an IEC-dependent manner. Hence, the activation of apical TLR9 on IECs, potentially by microbiota-derived signals, may play an important role in the prevention of allergic inflammation.


Assuntos
Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Hipersensibilidade Alimentar/prevenção & controle , Mucosa Intestinal/citologia , Leucócitos Mononucleares/metabolismo , Receptores Toll-Like/metabolismo , Adulto , Idoso , Linhagem Celular Tumoral , Técnicas de Cocultura , Feminino , Hipersensibilidade Alimentar/imunologia , Hipersensibilidade Alimentar/metabolismo , Regulação da Expressão Gênica/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Receptores Toll-Like/imunologia , Adulto Jovem
14.
Front Immunol ; 5: 60, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24600450

RESUMO

The intestinal mucosa is constantly facing a high load of antigens including bacterial antigens derived from the microbiota and food. Despite this, the immune cells present in the gastrointestinal tract do not initiate a pro-inflammatory immune response. Toll-like receptors (TLRs) are pattern recognition receptors expressed by various cells in the gastrointestinal tract, including intestinal epithelial cells (IEC) and resident immune cells in the lamina propria. Many diseases, including chronic intestinal inflammation (e.g., inflammatory bowel disease), irritable bowel syndrome (IBS), allergic gastroenteritis (e.g., eosinophilic gastroenteritis and allergic IBS), and infections are nowadays associated with a deregulated microbiota. The microbiota may directly interact with TLR. In addition, differences in intestinal TLR expression in health and disease may suggest that TLRs play an essential role in disease pathogenesis and may be novel targets for therapy. TLR signaling in the gut is involved in either maintaining intestinal homeostasis or the induction of an inflammatory response. This mini review provides an overview of the current knowledge regarding the contribution of intestinal epithelial TLR signaling in both tolerance induction or promoting intestinal inflammation, with a focus on food allergy. We will also highlight a potential role of the microbiota in regulating gut immune responses, especially through TLR activation.

15.
Basic Clin Pharmacol Toxicol ; 114(1): 128-36, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24118847

RESUMO

Disturbed bidirectional pathways between the (central) nervous system and immune system have been implicated in various mental disorders, including depressive and neurodevelopmental disorders. In this minireview, the role of the neuro-immune axis and its targetability in relation to major depression and autism spectrum disorder will be discussed. All together, the management of these and possibly other multi-factorial mental disorders needs a new and integrated therapeutic approach. Pharmacologically bioactive molecules as well as medical nutrition targeting the (gut)-immune-brain axis could be such an approach.


Assuntos
Transtornos Mentais/tratamento farmacológico , Transtornos Mentais/imunologia , Transtornos Mentais/fisiopatologia , Animais , Anti-Inflamatórios/farmacologia , Transtorno Autístico/tratamento farmacológico , Transtorno Autístico/imunologia , Encéfalo/fisiopatologia , Sistema Nervoso Central/efeitos dos fármacos , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/imunologia , Modelos Animais de Doenças , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/inervação , Trato Gastrointestinal/fisiopatologia , Humanos
16.
J Innate Immun ; 5(6): 625-38, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23735749

RESUMO

Dietary intervention using nondigestible oligosaccharides, short-chain galacto-oligosaccharides (scGOS)/long-chain fructo-oligosaccharides (lcFOS), in combination with Bifidobacterium breve M-16V prevents allergic disease involving galectin-9. In addition, apical TLR9 signaling contributes to intestinal homeostasis. We studied the contribution of galectin-9 secreted by intestinal epithelial cells (IEC; HT-29 and T84) in Th1 and regulatory T-cell (Treg) polarization in vitro. IEC were grown in transwell filters, cocultured with CD3/CD28-activated human peripheral blood mononuclear cells (PBMC) and apically exposed to genomic DNA derived from B. breve M-16V or synthetic TLR9 ligand in the absence or presence of scGOS/lcFOS. Cytokine production and T-cell phenotype were determined and galectin expression by IEC was assessed. Galectin-9 was neutralized using lactose or a TIM-3-Fc fusion protein. IEC exposed to DNA from B. breve M-16V or TLR9 ligand in the presence of scGOS/lcFOS enhanced IFN-γ secretion by PBMC and increased the percentage of Th1 and Treg cells. Expression and secretion of galectin-9 by IEC was increased and neutralization of galectin-9 prevented the induction of IFN-γ secretion and also suppressed the production of IL-10 by PBMC. Furthermore, we show that galectin-9 induces Treg and Th1 polarization through interaction with antigen-presenting cells. Our findings show that galectin-9 secreted by IEC apically exposed to TLR9 ligand in the presence of scGOS/lcFOS is involved in Th1 and Treg polarization and may be a promising target to prevent or treat allergic disease.


Assuntos
Células Epiteliais/imunologia , Galectinas/imunologia , Imunomodulação/imunologia , Mucosa Intestinal/imunologia , Oligossacarídeos/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Bifidobacterium/genética , Bifidobacterium/fisiologia , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , DNA Bacteriano/imunologia , DNA Bacteriano/farmacologia , Células Epiteliais/metabolismo , Citometria de Fluxo , Galectinas/metabolismo , Células HT29 , Humanos , Imunomodulação/efeitos dos fármacos , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Oligossacarídeos/farmacologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Receptor Toll-Like 9/imunologia , Receptor Toll-Like 9/metabolismo
17.
Eur J Pharmacol ; 697(1-3): 59-64, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23051675

RESUMO

We investigated the effects of minocycline, a microglia suppressant, on olfactory bulbectomized (OBX) rats, a model of cognitive and behavioral impairments arising from neurodegenerative processes. Previously, we demonstrated that the major OBX-induced behavioral and cognitive impairments develop between day 3 and 7 following bulbectomy. Here we show that the onset of these cognitive changes parallel in time with signs of microglia activation (increased mRNA levels of IL-1ß and CD68) in hippocampus. Next, rats were treated with minocycline (50mg/kg, i.p.) once daily for 4 weeks. OBX surgery was done at day 3 of drug treatment. Animals were tested in a battery of behavioral assays: open field, passive avoidance (fear learning and memory-acquired prior to OBX) and T-maze (spatial memory, conducted post bulbectomy). Minocycline normalized OBX-induced hyperactivity in the open field. Minocycline failed to prevent fear memory loss, but protected the OBX rats against hippocampal-dependent spatial memory deficit. Our findings suggest that treatment with minocycline may be effective in the early phase of a neurodegenerative disease.


Assuntos
Comportamento Animal/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Medo/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Memória/efeitos dos fármacos , Minociclina/farmacologia , Nootrópicos/farmacologia , Bulbo Olfatório/cirurgia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/metabolismo , Aprendizagem da Esquiva/efeitos dos fármacos , Cognição/efeitos dos fármacos , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/genética , Transtornos Cognitivos/psicologia , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Injeções Intraperitoneais , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Transtornos da Memória/etiologia , Transtornos da Memória/genética , Transtornos da Memória/psicologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Minociclina/administração & dosagem , Testes Neuropsicológicos , Nootrópicos/administração & dosagem , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Regulação para Cima
18.
Br J Nutr ; 107(1): 96-105, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21733338

RESUMO

Dietary non-digestible carbohydrates reduce the development of cows' milk allergy in mice. In the present study, the contribution of CD25+ regulatory T-cells (Treg) was investigated using in vivo Treg depletion and adoptive transfer studies. Mice were orally sensitised with casein and fed a diet containing 2 % short-chain galacto-, long-chain fructo- and acidic oligosaccharides (GFA) or a control diet. Donor splenocytes of mice sensitised with casein and fed the GFA or control diet were adoptively transferred to naive recipient mice, which were casein- or sham-sensitised and fed the control diet. In addition, in vivo or ex vivo CD25+ Treg depletion was performed using anti-CD25 (PC61). The acute allergic skin response upon intradermal casein challenge and casein-specific Ig were determined. Furthermore, T-helper (TH) 1 and TH2 cell numbers were analysed in the mesenteric lymph nodes. The oligosaccharide diet strongly reduced the development of the acute allergic skin response, which was abrogated by the in vivo anti-CD25 treatment. The diet enhanced the percentage of TH1 cells and tended to reduce the percentage of TH2 cells in casein-sensitised mice. Recipient mice were protected against the development of an acute allergic skin response when transferred with splenocytes from casein-sensitised GFA-fed donor mice before sensitisation. Ex vivo depletion of CD25+ Treg abrogated this transfer of tolerance. Splenocytes from sham-sensitised GFA-fed donor mice did not suppress the allergic response in recipient mice. In conclusion, CD25+ Treg contribute to the suppression of the allergic effector response in casein-sensitised mice induced by dietary intervention with non-digestible carbohydrates.


Assuntos
Caseínas/efeitos adversos , Carboidratos da Dieta/uso terapêutico , Imunomodulação , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Hipersensibilidade a Leite/prevenção & controle , Oligossacarídeos/uso terapêutico , Linfócitos T Reguladores/imunologia , Imunidade Adaptativa , Animais , Transplante de Células , Feminino , Frutose/química , Galactose/química , Linfonodos/imunologia , Linfonodos/patologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C3H , Hipersensibilidade a Leite/imunologia , Hipersensibilidade a Leite/metabolismo , Oligossacarídeos/química , Pele/imunologia , Organismos Livres de Patógenos Específicos , Baço/imunologia , Baço/patologia , Linfócitos T Reguladores/metabolismo
19.
Pediatr Allergy Immunol ; 22(8): 820-6, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21933283

RESUMO

BACKGROUND: Hypoallergenic formulas are considered a good option for infants at risk for cow's milk allergy. The aim of this animal study was to investigate whether whey hydrolyzates (WH) have the capacity to induce oral tolerance to whey. METHODS: Whey, partial or extensive WH was given via gavages to naïve mice prior to oral whey sensitization using cholera toxin as an adjuvant. The acute allergic skin response, mouse mast cell protease-1 (mMCP-1), whey-specific IgE, IgG(1) and effector Th2-cells, Th1-cells, and Foxp3(+) regulatory T-cells were determined in the mesenteric lymph nodes (MLN). MLN cells from tolerized mice were adoptively transferred to naïve recipient mice prior to whey sensitization. RESULTS: In contrast to the extensive WH, pre-treatment of naïve mice with whey or partial WH reduced the acute allergic skin response and mast cell degranulation after whey challenge. However, only treatment with whey prevented the generation of serum-specific IgE/IgG(1) . In partial WH tolerized mice, Foxp3(+) regulatory T-cell numbers in the MLN were increased compared to whey-sensitized mice. Both whey and partial WH treatment showed a tendency toward a decreased number of effector Th2-cells. Transfer of MLN cells from tolerized mice protected recipient mice from developing an acute allergic skin response. CONCLUSION: These results show that partial WH with limited sensitizing properties reduced the effector response upon whey challenge. This effect is transferable using MLN cells and was associated with enhanced Foxp3(+) regulatory T-cell numbers in the MLN. Partial WH retained the capacity to induce active immune suppression in mice which may be relevant for allergy prevention.


Assuntos
Alérgenos/administração & dosagem , Dessensibilização Imunológica , Tolerância Imunológica , Hipersensibilidade a Leite/imunologia , Proteínas do Leite/administração & dosagem , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo , Administração Oral , Alérgenos/efeitos adversos , Alérgenos/química , Animais , Células Cultivadas , Quimases/metabolismo , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Hidrólise , Imunoglobulina E/sangue , Linfonodos/patologia , Mesentério/patologia , Camundongos , Camundongos Endogâmicos C3H , Hipersensibilidade a Leite/tratamento farmacológico , Proteínas do Leite/efeitos adversos , Proteínas do Leite/química , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Equilíbrio Th1-Th2 , Proteínas do Soro do Leite
20.
Eur J Pharmacol ; 668 Suppl 1: S124-32, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21816141

RESUMO

The intestinal mucosa is constantly exposed to the luminal content, which includes micro-organisms and dietary components. Prebiotic non-digestible oligosaccharides may be supplemented to the diet to exert modulation of immune responses in the intestine. Short chain galacto- and long chain fructo-oligosaccharides (scGOS/lcFOS), functionally mimicking oligosaccharides present in human milk, have been reported to reduce the development of allergy through modulation of the intestinal microbiota and immune system. Nonetheless, the underlying working mechanisms of scGOS/lcFOS are unclear. Intestinal epithelial cells lining the mucosa are known to express carbohydrate (glycan)-binding receptors that may be involved in modulation of the mucosal immune response. This review aims to provide an overview of glycan-binding receptors, in particular galectins, which are expressed by intestinal epithelial cells and immune cells. In addition, their involvement in health and disease will be addressed, especially in food allergy and inflammatory bowel disease, diseases originating from the gastro-intestinal tract. Insight in the recognition of glycans in the intestinal tract may open new avenues for the treatment of intestinal inflammatory diseases by either nutritional concepts or pharmacological intervention.


Assuntos
Dietoterapia/métodos , Dieta , Sistema Imunitário/imunologia , Imunomodulação/imunologia , Intestinos/imunologia , Polissacarídeos/imunologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...