Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Int J Mol Sci ; 22(14)2021 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-34299148

RESUMO

During influenza A virus (IAV) infections, CD4+ T cell responses within infected lungs mainly involve T helper 1 (Th1) and regulatory T cells (Tregs). Th1-mediated responses favor the co-expression of T-box transcription factor 21 (T-bet) in Foxp3+ Tregs, enabling the efficient Treg control of Th1 responses in infected tissues. So far, the exact accumulation kinetics of T cell subsets in the lungs and lung-draining lymph nodes (dLN) of IAV-infected mice is incompletely understood, and the epigenetic signature of Tregs accumulating in infected lungs has not been investigated. Here, we report that the total T cell and the two-step Treg accumulation in IAV-infected lungs is transient, whereas the change in the ratio of CD4+ to CD8+ T cells is more durable. Within lungs, the frequency of Tregs co-expressing T-bet is steadily, yet transiently, increasing with a peak at Day 7 post-infection. Interestingly, T-bet+ Tregs accumulating in IAV-infected lungs displayed a strongly demethylated Tbx21 locus, similarly as in T-bet+ conventional T cells, and a fully demethylated Treg-specific demethylated region (TSDR) within the Foxp3 locus. In summary, our data suggest that T-bet+ but not T-bet- Tregs are epigenetically stabilized during IAV-induced infection in the lung.


Assuntos
Linfócitos T CD8-Positivos/virologia , Epigênese Genética , Fatores de Transcrição Forkhead/metabolismo , Pulmão/virologia , Infecções por Orthomyxoviridae/virologia , Proteínas com Domínio T/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Fatores de Transcrição Forkhead/genética , Vírus da Influenza A/fisiologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/patologia , Proteínas com Domínio T/genética
2.
Cell Mol Life Sci ; 74(15): 2839-2850, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28378044

RESUMO

Adaptive immunity critically contributes to control acute infection with enteropathogenic Yersinia pseudotuberculosis; however, the role of CD4+ T cell subsets in establishing infection and allowing pathogen persistence remains elusive. Here, we assessed the modulatory capacity of Y. pseudotuberculosis on CD4+ T cell differentiation. Using in vivo assays, we report that infection with Y. pseudotuberculosis resulted in enhanced priming of IL-17-producing T cells (Th17 cells), whereas induction of Foxp3+ regulatory T cells (Tregs) was severely disrupted in gut-draining mesenteric lymph nodes (mLNs), in line with altered frequencies of tolerogenic and proinflammatory dendritic cell (DC) subsets within mLNs. Additionally, by using a DC-free in vitro system, we could demonstrate that Y. pseudotuberculosis can directly modulate T cell receptor (TCR) downstream signaling within naïve CD4+ T cells and Tregs via injection of effector molecules through the type III secretion system, thereby affecting their functional properties. Importantly, modulation of naïve CD4+ T cells by Y. pseudotuberculosis resulted in an enhanced Th17 differentiation and decreased induction of Foxp3+ Tregs in vitro. These findings shed light to the adjustment of the Th17-Treg axis in response to acute Y. pseudotuberculosis infection and highlight the direct modulation of CD4+ T cell subsets by altering their TCR downstream signaling.


Assuntos
Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Reguladores/microbiologia , Células Th17/microbiologia , Infecções por Yersinia pseudotuberculosis/imunologia , Yersinia pseudotuberculosis/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Diferenciação Celular , Feminino , Fatores de Transcrição Forkhead/imunologia , Interações Hospedeiro-Patógeno , Intestinos/imunologia , Intestinos/microbiologia , Camundongos Endogâmicos BALB C , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Yersinia pseudotuberculosis/fisiologia
3.
PLoS Pathog ; 11(1): e1004613, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25615690

RESUMO

Enterovirus 71 (EV71) is the most virulent pathogen among enteroviruses that cause hand, foot and mouth disease in children but rarely in adults. The mechanisms that determine the age-dependent susceptibility remain largely unclear. Here, we found that the paucity of invariant natural killer T (iNKT) cells together with immaturity of the immune system was related to the susceptibility of neonatal mice to EV71 infection. iNKT cells were crucial antiviral effector cells to protect young mice from EV71 infection before their adaptive immune systems were fully mature. EV71 infection led to activation of iNKT cells depending on signaling through TLR3 but not other TLRs. Surprisingly, iNKT cell activation during EV71 infection required TLR3 signaling in macrophages, but not in dendritic cells (DCs). Mechanistically, interleukin (IL)-12 and endogenous CD1d-restricted antigens were both required for full activation of iNKT cells. Furthermore, CD1d-deficiency led to dramatically increased viral loads in central nervous system and more severe disease in EV71-infected mice. Altogether, our results suggest that iNKT cells may be involved in controlling EV71 infection in children when their adaptive immune systems are not fully developed, and also imply that iNKT cells might be an intervention target for treating EV71-infected patients.


Assuntos
Enterovirus Humano A/imunologia , Infecções por Enterovirus/imunologia , Imunidade Celular , Macrófagos/imunologia , Células T Matadoras Naturais/imunologia , Receptor 3 Toll-Like/fisiologia , Animais , Células Cultivadas , Infecções por Enterovirus/genética , Humanos , Imunidade Celular/genética , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Células T Matadoras Naturais/metabolismo , Transdução de Sinais/imunologia
4.
J Virol ; 89(21): 10860-7, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26292317

RESUMO

UNLABELLED: Coxsackievirus A16 (CVA16) is one of the major etiological agents of hand, foot, and mouth disease (HFMD) in children. The host defense mechanisms against CVA16 infection remain almost entirely unknown. Unlike previous observations with enterovirus 71 (EV71) infection, here we show that gamma interferon (IFN-γ) or invariant NK T cell deficiency does not affect disease development or the survival of CVA16-infected mice. In contrast, type I interferon receptor deficiency resulted in the development of more severe disease in mice, and the mice had a lower survival rate than wild-type mice. Similarly, a deficiency of Toll-like receptor 3 (TLR3) and TRIF, but not other pattern recognition receptors, led to the decreased survival of CVA16-infected mice. TLR3-TRIF signaling was indispensable for the induction of type I interferons during CVA16 infection in mice and protected young mice from disease caused by the infection. In particular, TRIF-mediated immunity was critical for preventing CVA16 replication in the neuronal system before disease occurred. IFN-ß treatment was also found to compensate for TRIF deficiency in mice and decreased the disease severity in and mortality of CVA16-infected mice. Altogether, type I interferons induced by TLR3-TRIF signaling mediate protective immunity against CVA16 infection. These findings may shed light on therapeutic strategies to combat HFMD caused by CVA16 infection. IMPORTANCE: Hand, foot, and mouth disease (HFMD) is a major threat to public health in the Asia-Pacific region. Both CVA16 and EV71 are major pathogens that are responsible for HFMD. The majority of research efforts have focused on the more virulent EV71, but little has been done with CVA16. Thus far, host immune responses to CVA16 infection have not yet been elucidated. The present study discovered an initial molecular mechanism underlying host protective immunity against CVA16 infection, providing the first explanation for why CVA16 and EV71 cause different clinical outcomes upon infection of humans. Therefore, different therapeutic strategies should be developed to treat HFMD cases caused by these two viruses.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Infecções por Coxsackievirus/prevenção & controle , Interferon Tipo I/imunologia , Transdução de Sinais/imunologia , Receptor 3 Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Animais , Infecções por Coxsackievirus/tratamento farmacológico , Primers do DNA/genética , Células Dendríticas/imunologia , Citometria de Fluxo , Interferon Tipo I/metabolismo , Interferon beta/genética , Interferon beta/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 3 Toll-Like/deficiência
5.
Cell Rep ; 43(5): 114153, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38687643

RESUMO

Gut-draining mesenteric and celiac lymph nodes (mLNs and celLNs) critically contribute to peripheral tolerance toward food and microbial antigens by supporting the de novo induction of regulatory T cells (Tregs). These tolerogenic properties of mLNs and celLNs are stably imprinted within stromal cells (SCs) by microbial signals and vitamin A (VA), respectively. Here, we report that a single, transient gastrointestinal infection in the neonatal, but not adult, period durably abrogates the efficient Treg-inducing capacity of celLNs by altering the subset composition and gene expression profile of celLNSCs. These cells carry information about the early-life pathogen encounter until adulthood and durably instruct migratory dendritic cells entering the celLN with reduced tolerogenic properties. Mechanistically, transiently reduced VA levels cause long-lasting celLN functional impairment, which can be rescued by early-life treatment with VA. Together, our data highlight the therapeutic potential of VA to prevent sequelae post gastrointestinal infections in infants.


Assuntos
Linfonodos , Linfócitos T Reguladores , Vitamina A , Animais , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/efeitos dos fármacos , Vitamina A/farmacologia , Vitamina A/uso terapêutico , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Camundongos , Animais Recém-Nascidos , Tolerância Imunológica/efeitos dos fármacos , Células Dendríticas/imunologia , Camundongos Endogâmicos C57BL , Feminino
6.
Front Immunol ; 13: 991671, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36119090

RESUMO

The first wave of Foxp3+ regulatory T cells (Tregs) generated in neonates is critical for the life-long prevention of autoimmunity. Although it is widely accepted that neonates are highly susceptible to infections, the impact of neonatal infections on this first wave of Tregs is completely unknown. Here, we challenged newborn Treg fate-mapping mice (Foxp3eGFPCreERT2xROSA26STOP-eYFP) with the Toll-like receptor (TLR) agonists LPS and poly I:C to mimic inflammatory perturbations upon neonatal bacterial or viral infections, respectively, and subsequently administrated tamoxifen during the first 8 days of life to selectively label the first wave of Tregs. Neonatally-tagged Tregs preferentially accumulated in non-lymphoid tissues (NLTs) when compared to secondary lymphoid organs (SLOs) irrespective of the treatment. One week post challenge, no differences in the frequency and phenotypes of neonatally-tagged Tregs were observed between challenged mice and untreated controls. However, upon aging, a decreased frequency of neonatally-tagged Tregs in both NLTs and SLOs was detected in challenged mice when compared to untreated controls. This decrease became significant 12 weeks post challenge, with no signs of altered Foxp3 stability. Remarkably, this late decrease in the frequency of neonatally-tagged Tregs only occurred when newborns were challenged, as treating 8-days-old mice with TLR agonists did not result in long-lasting alterations of the first wave of Tregs. Combined single-cell T cell receptor (TCR)-seq and RNA-seq revealed that neonatal inflammatory perturbations drastically diminished TCR diversity and long-lastingly altered the transcriptome of neonatally-tagged Tregs, exemplified by lower expression of Tigit, Foxp3, and Il2ra. Together, our data demonstrate that a single, transient encounter with a pathogen in early life can have long-lasting consequences for the first wave of Tregs, which might affect immunological tolerance, prevention of autoimmunity, and other non-canonical functions of tissue-resident Tregs in adulthood.


Assuntos
Linfócitos T Reguladores , Transcriptoma , Animais , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Lipopolissacarídeos/metabolismo , Camundongos , Poli I/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Tamoxifeno/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
7.
Eur J Microbiol Immunol (Bp) ; 10(2): 98-106, 2020 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-32644940

RESUMO

Listeria monocytogenes (Lm) is a food-borne pathogen with a high chance of infecting neonates, pregnant women, elderly and immunocompromised individuals. Lm infection in neonates can cause neonatal meningitis and sepsis with a high risk of severe neurological and developmental sequelae and high mortality rates. However, whether an acute neonatal Lm infection causes long-term effects on the immune system persisting until adulthood has not been fully elucidated. Here, we established a neonatal Lm infection model and monitored the composition of major immune cell subsets at defined time points post infection (p.i.) in secondary lymphoid organs and the intestine. Twelve weeks p.i., the CD8+ T cell population was decreased in colon and mesenteric lymph nodes (mLNs) with an opposing increase in the spleen. In the colon, we observed an accumulation of CD4+ and CD8+ effector/memory T cells with an increase of T-bet+ T helper 1 (Th1) cells. In addition, 12 weeks p.i. an altered composition of innate lymphoid cell (ILC) and dendritic cell (DC) subsets was still observed in colon and mLNs, respectively. Together, these findings highlight organ-specific long-term consequences of an acute neonatal Lm infection on both the adaptive and innate immune system.

8.
Nat Commun ; 10(1): 746, 2019 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-30765691

RESUMO

Excessive or uncontrolled release of proinflammatory cytokines caused by severe viral infections often results in host tissue injury or even death. Phospholipase C (PLC)s degrade phosphatidylinositol-4, 5-bisphosphate (PI(4,5)P2) lipids and regulate multiple cellular events. Here, we report that PLCß2 inhibits the virus-induced expression of pro-inflammatory cytokines by interacting with and inhibiting transforming growth factor-ß-activated kinase 1 (TAK1) activation. Mechanistically, PI(4,5)P2 lipids directly interact with TAK1 at W241 and N245, and promote its activation. Impairing of PI(4,5)P2's binding affinity or mutation of PIP2-binding sites on TAK1 abolish its activation and the subsequent production of pro-inflammatory cytokines. Moreover, PLCß2-deficient mice exhibit increased expression of proinflammatory cytokines and a higher frequency of death in response to virus infection, while the PLCß2 activator, m-3M3FBS, protects mice from severe Coxsackie virus A 16 (CVA16) infection. Thus, our findings suggest that PLCß2 negatively regulates virus-induced pro-inflammatory responses by inhibiting phosphoinositide-mediated activation of TAK1.


Assuntos
Infecções por Coxsackievirus/metabolismo , Citocinas/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipase C beta/metabolismo , Animais , Células Cultivadas , Chlorocebus aethiops , Infecções por Coxsackievirus/genética , Infecções por Coxsackievirus/virologia , Citocinas/genética , Enterovirus/fisiologia , Ativação Enzimática , Regulação da Expressão Gênica , Células HEK293 , Humanos , MAP Quinase Quinase Quinases/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfolipase C beta/genética , Ligação Proteica , Células Vero
9.
J Mol Med (Berl) ; 96(12): 1387-1394, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30357435

RESUMO

Foxp3+ regulatory T cells (Tregs) co-expressing the Th17-lineage specification factor RORγt represent a unique Treg subpopulation that has been reported to be induced upon response to gut microbiota within the intestinal immune system. Hence, RORγt+ Tregs are considered to solely consist of peripherally induced Foxp3+ Tregs (pTregs), and the possibility that also thymus-derived Treg (tTregs) can upregulate RORγt expression and contribute to the pool of RORγt+ Tregs was largely ignored. Here, we expand our knowledge on the origin of RORγt+ Tregs by demonstrating that also tTregs can attain RORγt expression. In transgenic Foxp3∆CNS1-Cre reporter mice, a substantial fraction of CNS1-independent Tregs, predominantly consisting of tTregs, was found to co-express RORγt. In addition, genuine tTregs isolated from thymi of Foxp3hCD2RAGGFP reporter mice initiated RORγt expression both in vitro and in vivo, particularly under inflammatory conditions. In conclusion, our data demonstrate that tTregs can upregulate RORγt expression under inflammatory conditions and that hence RORγt+ Tregs can be regarded as a heterogeneous population consisting of both pTregs and tTregs. KEY MESSAGES: RORγt cannot be considered as a marker for pTregs. CNS1-independent tTregs within the colon display RORγt expression. RORγt can be induced in genuine tTregs, particularly under inflammatory conditions. RORγt+ Tregs are a heterogeneous population consisting of both pTregs and tTregs.


Assuntos
Fatores de Transcrição Forkhead/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Colite/imunologia , Colite/terapia , Inflamação/imunologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T Reguladores/transplante , Regulação para Cima
10.
Sci Rep ; 5: 12941, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26251280

RESUMO

Transgenic expression of I-E molecules prevents diabetes in NOD mice. So far, the precise role of these non-classical MHC II molecules remains elusive. Here, we showed that transgenic expression of I-E(k) alpha 16 molecule in NOD mice selectively reduced Th17 cells in the thymus and pancreatic draining lymph nodes. The reduction in Th17 cells was associated with both attenuated IL-6 production and decreased activation of macrophages. Mechanistically, transgenic expression of the I-E molecule diminished expression of intracellular classical MHC II molecule and led to impaired TLR4-mediated signaling. In contrast to classical MHC II molecule, this non-classical MHC II molecule negatively regulates the inflammatory responses of macrophages. Altogether, our study reveals a novel regulatory role of I-E molecules in modulating inflammatory immune responses.


Assuntos
Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Células Th17/imunologia , Animais , Inflamação/imunologia , Interleucina-6/imunologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos NOD , Pâncreas/imunologia , Transdução de Sinais/imunologia , Timo/imunologia , Receptor 4 Toll-Like/imunologia , Transativadores
11.
Int J Biochem Cell Biol ; 54: 89-97, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25035167

RESUMO

Myeloid-derived suppressor cells (MDSCs) play an important role in maintaining immune tolerance in response to tumors and inflammatory diseases. Several liver MDSCs have been described in hepatitis in humans and mouse models. Although all the murine MDSCs are CD11b(+)Gr-1(+), their true phenotype and mechanism of suppression remain elusive. This study revealed that SSC(high)CD11b(high)Ly-6C(high)Ly-6G(low) monocytic cells but not the other liver-infiltrating, CD11b(+)Gr-1(+) subsets could suppress CD4 T cell responses. Their suppressive activity was remarkably effective even at a ratio of 1:50 when co-cultured with CD4 T cells. Mechanistically, the suppression was dependent on nitric oxide production by inducible nitric oxide synthase (iNOS). Furthermore, the suppressive function by these liver MDSCs was found to require direct contact with activated CD4 T cells. Adoptive transfer experiments demonstrate that these liver MDSCs can dramatically ameliorate concanavalin A (Con A)-induced fulminant hepatitis in mice. Finally, MDSC-mediated suppression in vivo was dependent on iNOS expression. Altogether, SSC(high)CD11b(high)Ly-6C(high)Ly-6G(low) cells represent authentic MDSCs in the inflammatory liver and may function to minimize collateral damage caused by an overzealous CD4 T cell response following hepatitis infection.


Assuntos
Acetiltransferases/metabolismo , Antígenos Ly/metabolismo , Antígeno CD11b/metabolismo , Linfócitos T CD4-Positivos/imunologia , Hepatite/imunologia , Células Mieloides/imunologia , Óxido Nítrico Sintase Tipo II/fisiologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Proliferação de Células , Elongases de Ácidos Graxos , Feminino , Citometria de Fluxo , Hepatite/metabolismo , Hepatite/patologia , Tolerância Imunológica , Terapia de Imunossupressão , Falência Hepática Aguda/imunologia , Falência Hepática Aguda/metabolismo , Falência Hepática Aguda/patologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Células Mieloides/metabolismo , Células Mieloides/patologia , Óxido Nítrico/metabolismo
12.
Microbes Infect ; 16(8): 672-7, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24769417

RESUMO

Most patients infected with avian H7N9 influenza virus develop severe disease, including respiratory failure, acute respiratory distress syndrome (ARDS), and multi-organ failure. The pathogenesis of H7N9 infection is not fully understood. This study revealed that H7N9-infected patients who had fatal outcomes or critical illness all had pre-existing chronic diseases. The patients did not have obvious systemic inflammation compared to the healthy controls. However, their fatal outcomes and critically severe illness were significantly associated with high serum levels of tumor necrosis factor (TNF)-α. Interestingly, the degree of liver damage in these patients significantly correlated with their serum levels of Th2 cytokines, interleukin (IL)-4, and IL-9. Taken together, our results suggest that Th2-type inflammation in H7N9-infected patients with pre-existing chronic diseases likely contributes to the pathogenesis of H7N9 infection and is linked to poor clinical outcomes.


Assuntos
Subtipo H7N9 do Vírus da Influenza A/imunologia , Influenza Humana/complicações , Influenza Humana/virologia , Falência Hepática/patologia , Fígado/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Feminino , Humanos , Interleucina-4/sangue , Interleucina-9/sangue , Masculino , Pessoa de Meia-Idade , Fator de Necrose Tumoral alfa/sangue
13.
Virus Res ; 177(1): 62-5, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23886670

RESUMO

The immunopathogenesis of severe hand, foot and mouth disease (HFMD) remains elusive. This study revealed that enterovirus 71 (EV71) epitope-specific CD4+ T cell responses of HFMD patients were skewed toward a Th2 cytokine profile. Patients that demonstrated higher levels of IL-4 expression in their CD4 T cells following antigen stimulation in vitro tended to have a more prolonged period of high fevers and a longer duration of illness. Thus, an increase of EV71 epitope-specific Th2 type response may portend the poor prognosis for some HFMD patients.


Assuntos
Enterovirus Humano A/imunologia , Doença de Mão, Pé e Boca/diagnóstico , Doença de Mão, Pé e Boca/imunologia , Células Th2/imunologia , Estudos de Casos e Controles , Pré-Escolar , Feminino , Doença de Mão, Pé e Boca/virologia , Humanos , Imunidade Celular , Lactente , Interleucina-4/imunologia , Masculino , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA