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










Base de dados
Intervalo de ano de publicação
1.
Blood ; 138(12): 1034-1039, 2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34232994

RESUMO

Hemophagocytic lymphohistiocytosis (HLH) is an inflammatory disorder in which numerous cytokines are elevated, though interferon-γ (IFN-γ) is central to disease pathogenesis and a key therapeutic target. Experimental and early clinical reports have shown that ruxolitinib, a small molecule inhibitor of Janus kinases (JAKs), which are essential for cytokine signaling, may be therapeutic in HLH. In contrast, we found that intermittently administered ruxolitinib at various dose levels failed to prevent HLH development or treat established murine HLH. High doses of ruxolitinib blocked IFN-γ signaling only transiently after administration, consistent with human pharmacokinetics, and only continuously administered drug could prevent HLH development or treat established HLH. Continuously administered ruxolitinib was therapeutic in only a narrow dose range and intermittently dosed ruxolitinib worsened survival and decreased bone marrow cellularity of animals concurrently treated with anti-IFN-γ antibody, indicating a narrow therapeutic window and potential toxicity. Because JAK2 is essential for hematopoietic cytokine signaling, we also tested a JAK1-selective inhibitor and observed therapeutic benefit without apparent toxicity, though it did not improve survival when combined with anti-IFN-γ. We conclude that continuous blockade of IFN-γ signaling is necessary for optimal control of HLH and that JAK2 inhibition may be toxic in this disorder.


Assuntos
Interferon gama/imunologia , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/imunologia , Linfo-Histiocitose Hemofagocítica , Nitrilas/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Janus Quinase 2/genética , Linfo-Histiocitose Hemofagocítica/tratamento farmacológico , Linfo-Histiocitose Hemofagocítica/imunologia , Camundongos , Transdução de Sinais/imunologia
2.
Blood ; 137(17): 2337-2346, 2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33512385

RESUMO

Hemophagocytic lymphohistiocytosis (HLH) is a fatal disorder of immune hyperactivation that has been described as a cytokine storm. Sepsis due to known or suspected infection has also been viewed as a cytokine storm. Although clinical similarities between these syndromes suggest similar immunopathology and may create diagnostic uncertainty, distinguishing them is critical as treatments are widely divergent. We examined T-cell profiles from children with either HLH or sepsis and found that HLH is characterized by acute T-cell activation, in clear contrast to sepsis. Activated T cells in patients with HLH were characterized as CD38high/HLA-DR+ effector cells, with activation of CD8+ T cells being most pronounced. Activated T cells were type 1 polarized, proliferative, and displayed evidence of recent and persistent activation. Circulating activated T cells appeared to be broadly characteristic of HLH, as they were seen in children with and without genetic lesions or identifiable infections and resolved with conventional treatment of HLH. Furthermore, we observed even greater activation and type 1 polarization in tissue-infiltrating T cells, described here for the first time in a series of patients with HLH. Finally, we observed that a threshold of >7% CD38high/HLA-DR+ cells among CD8+ T cells had strong positive and negative predictive value for distinguishing HLH from early sepsis or healthy controls. We conclude that the cytokine storm of HLH is marked by distinctive T-cell activation whereas early sepsis is not, and that these 2 syndromes can be readily distinguished by T-cell phenotypes.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Linfócitos T CD8-Positivos/imunologia , Síndrome da Liberação de Citocina/diagnóstico , Antígenos HLA-DR/metabolismo , Ativação Linfocitária/imunologia , Linfo-Histiocitose Hemofagocítica/diagnóstico , Glicoproteínas de Membrana/metabolismo , Sepse/diagnóstico , Adolescente , Adulto , Estudos de Casos e Controles , Criança , Pré-Escolar , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/patologia , Diagnóstico Diferencial , Feminino , Humanos , Lactente , Linfo-Histiocitose Hemofagocítica/imunologia , Linfo-Histiocitose Hemofagocítica/patologia , Masculino , Sepse/imunologia , Sepse/patologia , Adulto Jovem
3.
Front Immunol ; 8: 1561, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29201027

RESUMO

Activation, recruitment, and effector function of T lymphocytes are essential for control of mycobacterial infection. These processes are tightly regulated in T cells by the availability of l-arginine within the microenvironment. In turn, mycobacterial infection dampens T cell responsiveness through arginase induction in myeloid cells, promoting sequestration of l-arginine within the local milieu. Here, we show T cells can replenish intracellular l-arginine through metabolism of l-citrulline to mediate inflammatory function, allowing anti-mycobacterial T cells to overcome arginase-mediated suppression. Furthermore, T cell l-citrulline metabolism is necessary for accumulation of CD4+ T cells at the site of infection, suggesting this metabolic pathway is involved during anti-mycobacterial T cell immunity in vivo. Together, these findings establish a contribution for l-arginine synthesis by T cells during mycobacterial infection, and implicate l-citrulline as a potential immuno-nutrient to modulate host immunity.

4.
Proc Natl Acad Sci U S A ; 114(24): E4782-E4791, 2017 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-28533414

RESUMO

Antigen-activated lymphocytes undergo extraordinarily rapid cell division in the course of immune responses. We hypothesized that this unique aspect of lymphocyte biology leads to unusual genomic stress in recently antigen-activated lymphocytes and that targeted manipulation of DNA damage-response (DDR) signaling pathways would allow for selective therapeutic targeting of pathological T cells in disease contexts. Consistent with these hypotheses, we found that activated mouse and human T cells display a pronounced DDR in vitro and in vivo. Upon screening a variety of small-molecule compounds, we found that potentiation of p53 (via inhibition of MDM2) or impairment of cell cycle checkpoints (via inhibition of CHK1/2 or WEE1) led to the selective elimination of activated, pathological T cells in vivo. The combination of these strategies [which we termed "p53 potentiation with checkpoint abrogation" (PPCA)] displayed therapeutic benefits in preclinical disease models of hemophagocytic lymphohistiocytosis and multiple sclerosis, which are driven by foreign antigens or self-antigens, respectively. PPCA therapy targeted pathological T cells but did not compromise naive, regulatory, or quiescent memory T-cell pools, and had a modest nonimmune toxicity profile. Thus, PPCA is a therapeutic modality for selective, antigen-specific immune modulation with significant translational potential for diverse immune-mediated diseases.


Assuntos
Dano ao DNA/imunologia , Doenças do Sistema Imunitário/terapia , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/imunologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/terapia , Etoposídeo/administração & dosagem , Humanos , Doenças do Sistema Imunitário/imunologia , Ativação Linfocitária , Linfo-Histiocitose Hemofagocítica/imunologia , Linfo-Histiocitose Hemofagocítica/terapia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Esclerose Múltipla/imunologia , Esclerose Múltipla/terapia , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-mdm2/imunologia , Transdução de Sinais/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia , Proteína Supressora de Tumor p53/imunologia
5.
JCI Insight ; 2(5): e91288, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28289719

RESUMO

Preterm birth (PTB) is a leading worldwide cause of morbidity and mortality in infants. Maternal inflammation induced by microbial infection is a critical predisposing factor for PTB. However, biological processes associated with competency of pathogens, including viruses, to induce PTB or sensitize for secondary bacterial infection-driven PTB are unknown. We show that pathogen/pathogen-associated molecular pattern-driven activation of type I IFN/IFN receptor (IFNAR) was sufficient to prime for systemic and uterine proinflammatory chemokine and cytokine production and induction of PTB. Similarly, treatment with recombinant type I IFNs recapitulated such effects by exacerbating proinflammatory cytokine production and reducing the dose of secondary inflammatory challenge required for induction of PTB. Inflammatory challenge-driven induction of PTB was eliminated by defects in type I IFN, TLR, or IL-6 responsiveness, whereas the sequence of type I IFN sensing by IFNAR on hematopoietic cells was essential for regulation of proinflammatory cytokine production. Importantly, we also show that type I IFN priming effects are conserved from mice to nonhuman primates and humans, and expression of both type I IFNs and proinflammatory cytokines is upregulated in human PTB. Thus, activation of the type I IFN/IFNAR axis in pregnancy primes for inflammation-driven PTB and provides an actionable biomarker and therapeutic target for mitigating PTB risk.


Assuntos
Inflamação/fisiopatologia , Interferon Tipo I/fisiologia , Nascimento Prematuro , Animais , Citocinas/fisiologia , Suscetibilidade a Doenças , Feminino , Humanos , Recém-Nascido , Interferon Tipo I/metabolismo , Camundongos , Gravidez , Transdução de Sinais
6.
Immunology ; 148(2): 216-26, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26931177

RESUMO

SHARPIN is an essential component of the linear ubiquitin chain assembly complex (LUBAC) complex that controls signalling pathways of various receptors, including the tumour necrosis factor receptor (TNFR), Toll-like receptor (TLR) and antigen receptor, in part by synthesis of linear, non-degrading ubiquitin chains. Consistent with SHARPIN's function in different receptor pathways, the phenotype of SHARPIN-deficient mice is complex, including the development of inflammatory systemic and skin diseases, the latter of which depend on TNFR signal transduction. Given the established function of SHARPIN in primary and malignant B cells, we hypothesized that SHARPIN might also regulate T-cell receptor (TCR) signalling and thereby control T-cell biology. Here, we focus primarily on the role of SHARPIN in T cells, specifically regulatory T (Treg) cells. We found that SHARPIN-deficient (Sharpin(cpdm/cpdm) ) mice have significantly reduced numbers of FOXP3(+) Treg cells in lymphoid organs and the peripheral blood. Competitive reconstitution of irradiated mice with mixed bone marrow from wild-type and SHARPIN-deficient mice revealed an overall reduced thymus population with SHARPIN-deficient cells with almost complete loss of thymic Treg development. Consistent with this cell-intrinsic function of SHARPIN in Treg development, TCR stimulation of SHARPIN-deficient thymocytes revealed reduced activation of nuclear factor-κB and c-Jun N-terminal kinase, establishing a function of SHARPIN in TCR signalling, which may explain the defective Treg development. In turn, in vitro generation and suppressive activity of mature SHARPIN-deficient Treg cells were comparable to wild-type cells, suggesting that maturation, but not function, of SHARPIN-deficient Treg cells is impaired. Taken together, these findings show that SHARPIN controls TCR signalling and is required for efficient generation of Treg cells in vivo, whereas the inhibitory function of mature Treg cells appears to be independent of SHARPIN.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Linfócitos T Reguladores/fisiologia , Timócitos/fisiologia , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Células Cultivadas , Fatores de Transcrição Forkhead/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/genética , Transdução de Sinais/genética
7.
J Clin Invest ; 125(4): 1713-25, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25751061

RESUMO

Mammalian pregnancy requires protection against immunological rejection of the developing fetus bearing discordant paternal antigens. Immune evasion in this developmental context entails silenced expression of chemoattractant proteins (chemokines), thereby preventing harmful immune cells from penetrating the maternal-fetal interface. Here, we demonstrate that fetal wastage triggered by prenatal Listeria monocytogenes infection is driven by placental recruitment of CXCL9-producing inflammatory neutrophils and macrophages that promote infiltration of fetal-specific T cells into the decidua. Maternal CD8+ T cells with fetal specificity upregulated expression of the chemokine receptor CXCR3 and, together with neutrophils and macrophages, were essential for L. monocytogenes-induced fetal resorption. Conversely, decidual accumulation of maternal T cells with fetal specificity and fetal wastage were extinguished by CXCR3 blockade or in CXCR3-deficient mice. Remarkably, protection against fetal wastage and in utero L. monocytogenes invasion was maintained even when CXCR3 neutralization was initiated after infection, and this protective effect extended to fetal resorption triggered by partial ablation of immune-suppressive maternal Tregs, which expand during pregnancy to sustain fetal tolerance. Together, our results indicate that functionally overriding chemokine silencing at the maternal-fetal interface promotes the pathogenesis of prenatal infection and suggest that therapeutically reinforcing this pathway represents a universal approach for mitigating immune-mediated pregnancy complications.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Morte Fetal/prevenção & controle , Listeriose/imunologia , Complicações Infecciosas na Gravidez/imunologia , Receptores CXCR3/fisiologia , Subpopulações de Linfócitos T/imunologia , Transferência Adotiva , Ampicilina/uso terapêutico , Animais , Antibacterianos/uso terapêutico , Quimiocina CXCL9/biossíntese , Quimiocina CXCL9/genética , Quimiocina CXCL9/fisiologia , Quimiocinas/metabolismo , Cruzamentos Genéticos , Decídua/imunologia , Feminino , Morte Fetal/etiologia , Reabsorção do Feto/imunologia , Reabsorção do Feto/prevenção & controle , Listeriose/tratamento farmacológico , Macrófagos/imunologia , Masculino , Troca Materno-Fetal , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neutrófilos/imunologia , Ovalbumina/genética , Ovalbumina/imunologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Gravidez , Complicações Infecciosas na Gravidez/tratamento farmacológico , Receptores CXCR3/antagonistas & inibidores , Receptores CXCR3/biossíntese , Receptores CXCR3/deficiência , Receptores CXCR3/genética , Baço/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T , Linfócitos T Reguladores/imunologia , Regulação para Cima , Virulência
8.
Cell Mol Immunol ; 11(6): 595-605, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25242275

RESUMO

Listeria monocytogenes (Lm) is an intracellular bacterium with unique predisposition for systemic maternal infection during pregnancy and morbid consequences for the developing fetus. Given the high mortality associated with prenatal Lm infection, strategies for augmenting protective immunity during the exceedingly vulnerable period of pregnancy are urgently needed. Herein, protection conferred by attenuated Lm administered before pregnancy against subsequent virulent Lm prenatal infection was evaluated. We show that protection against secondary Lm infection in non-pregnant mice is sharply moderated during allogeneic pregnancy because significantly more bacteria are recovered from maternal tissues, despite the numerical and functional preservation of pathogen-specific CD8(+) T cells. More importantly, preconceptual priming does not protect against in utero invasion or fetal wastage because mice inoculated with attenuated Lm prior to pregnancy and naive pregnant controls each showed near complete fetal resorption and pathogen recovery from individual concepti after Lm infection during pregnancy. Remarkably, the lack of protection against prenatal Lm infection with preconceptual priming in allogeneic pregnancy is restored during syngeneic pregnancy. Thus, maternal-fetal antigen discordance dictates the ineffectiveness of preconceptual vaccination against fetal complications after prenatal Lm infection, despite the numerical and functional preservation of pathogen-specific CD8(+) T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Listeria monocytogenes/fisiologia , Listeriose/imunologia , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Vacinas Atenuadas , Animais , Suscetibilidade a Doenças , Feminino , Histocompatibilidade , Humanos , Isoantígenos/imunologia , Listeriose/complicações , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Cuidado Pré-Concepcional , Gravidez/imunologia , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/imunologia , Vacinação , Vacinas Atenuadas/administração & dosagem
9.
Proc Natl Acad Sci U S A ; 111(29): 10672-7, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-25002484

RESUMO

The costimulatory B7-1 (CD80)/B7-2 (CD86) molecules, along with T-cell receptor stimulation, together facilitate T-cell activation. This explains why in vivo B7 costimulation neutralization efficiently silences a variety of human autoimmune disorders. Paradoxically, however, B7 blockade also potently moderates accumulation of immune-suppressive regulatory T cells (Tregs) essential for protection against multiorgan systemic autoimmunity. Here we show that B7 deprivation in mice overrides the necessity for Tregs in averting systemic autoimmunity and inflammation in extraintestinal tissues, whereas peripherally induced Tregs retained in the absence of B7 selectively mitigate intestinal inflammation caused by Th17 effector CD4(+) T cells. The need for additional immune suppression in the intestine reflects commensal microbe-driven T-cell activation through the accessory costimulation molecules ICOSL and OX40L. Eradication of commensal enteric bacteria mitigates intestinal inflammation and IL-17 production triggered by Treg depletion in B7-deficient mice, whereas re-establishing intestinal colonization with Candida albicans primes expansion of Th17 cells with commensal specificity. Thus, neutralizing B7 costimulation uncovers an essential role for Tregs in selectively averting intestinal inflammation by Th17 CD4(+) T cells with commensal microbe specificity.


Assuntos
Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Linfócitos T CD4-Positivos/imunologia , Ligante Coestimulador de Linfócitos T Induzíveis/metabolismo , Inflamação/imunologia , Interleucina-17/biossíntese , Intestinos/patologia , Ligante OX40/metabolismo , Animais , Linfócitos T CD4-Positivos/citologia , Antígeno CTLA-4/metabolismo , Candida albicans/fisiologia , Diferenciação Celular/imunologia , Proliferação de Células , Humanos , Inflamação/microbiologia , Inflamação/patologia , Intestinos/imunologia , Intestinos/microbiologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Linfócitos T Reguladores/imunologia , Células Th17/imunologia
10.
J Immunol ; 192(11): 4949-56, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24837152

RESUMO

The immunological alterations required for successful pregnancy in eutherian placental mammals have remained a scientific enigma since the discovery of MHC haplotype diversity and unique immune signatures among individuals. Within the past 10 years, accumulating data suggest that immune-suppressive regulatory T cells (Tregs) confer essential protective benefits in sustaining tolerance to the semiallogeneic fetus during pregnancy, along with their more established roles in maintaining tolerance to self and "extended self" commensal Ags that averts autoimmunity. Reciprocally, many human pregnancy complications stemming from inadequacies in fetal tolerance have been associated with defects in maternal Tregs. Thus, further elucidating the immunological shifts during pregnancy not only have direct translational implications for improving perinatal health, they have enormous potential for unveiling new clues about how Tregs work in other biological contexts. In this article, epidemiological data in human pregnancy and complementary animal studies implicating a pivotal protective role for maternal Tregs are summarized.


Assuntos
Antígenos/imunologia , Tolerância Imunológica , Complicações na Gravidez/imunologia , Linfócitos T Reguladores/imunologia , Animais , Feminino , Humanos , Gravidez , Complicações na Gravidez/patologia , Linfócitos T Reguladores/patologia
11.
J Immunol ; 192(7): 2970-4, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24591368

RESUMO

Pregnancy stimulates induced Foxp3 expression among maternal CD4(+) T cells with fetal specificity. Although sustained maternal regulatory CD4(+) T cell (Treg) expansion is essential for maintaining fetal tolerance during pregnancy, the necessity for Foxp3(+) cells with fetal specificity remains undefined. In this study, we demonstrate that mitigating Treg differentiation among maternal CD4(+) T cells with a single surrogate fetal specificity elicits Ag-specific fetal loss. Using recombinant Listeria monocytogenes to prime stably differentiated Th1 CD4(+) T cells with fetal I-A(b):2W1S55-68 specificity refractory to pregnancy-induced Foxp3 expression, we show that Ag delivery by cytoplasmic L. monocytogenes causes selective loss of 2W1S(+) offspring through CD4 cell- and IFN-γ-dependent pathways. In contrast, CD4(+) T cells primed by L. monocytogenes restricted from the cell cytoplasm are markedly more plastic for induced Foxp3 expression, with normal pregnancy outcomes. Thus, committed Th1 polarization blocks pregnancy induced Treg differentiation among maternal CD4(+) T cells with fetal specificity and triggers Ag-specific fetal loss.


Assuntos
Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Feto/imunologia , Fatores de Transcrição Forkhead/imunologia , Células Th1/imunologia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/transplante , Diferenciação Celular/imunologia , Feminino , Feto/metabolismo , Citometria de Fluxo , Fatores de Transcrição Forkhead/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Listeria monocytogenes/imunologia , Listeria monocytogenes/fisiologia , Listeriose/imunologia , Listeriose/microbiologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Gravidez , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/metabolismo
12.
Chimerism ; 5(1): 16-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24553046

RESUMO

Long-term maintenance of immune components with defined specificity, without antigen is the hallmark feature of immunological memory. However, there are fundamental differences in how memory CD8(+) compared with CD4(+) T cells are maintained. After complete antigen elimination, CD8(+) T cells can persist as a self-renewing numerically stable cell population, and therefore satisfy the most stringent definition of "memory." Comparatively, CD4(+) T cell maintenance is considerably less stable, often requiring low-level antigen persistence or antigenic reminders. Recent studies show these basic memory features, classically ascribed to effector CD8(+) and CD4(+) T cells, extend to immune suppressive Foxp3(+) regulatory CD4(+) T cells (Tregs). In particular, gestational expansion and postpartum retention of maternal Tregs with fetal specificity may explain the protective benefits of primary pregnancy on complications in subsequent pregnancy. Herein, the possibility of ongoing antigenic reminders from fetal cell microchimerism in postpartum maintenance of maternal Tregs with fetal specificity is considered.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Quimera/imunologia , Memória Imunológica , Troca Materno-Fetal , Linfócitos T Reguladores/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Embrião de Mamíferos , Feminino , Humanos , Gravidez , Linfócitos T Reguladores/imunologia
13.
Nature ; 504(7478): 158-62, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24196717

RESUMO

Newborn infants are highly susceptible to infection. This defect in host defence has generally been ascribed to the immaturity of neonatal immune cells; however, the degree of hyporesponsiveness is highly variable and depends on the stimulation conditions. These discordant responses illustrate the need for a more unified explanation for why immunity is compromised in neonates. Here we show that physiologically enriched CD71(+) erythroid cells in neonatal mice and human cord blood have distinctive immunosuppressive properties. The production of innate immune protective cytokines by adult cells is diminished after transfer to neonatal mice or after co-culture with neonatal splenocytes. Neonatal CD71(+) cells express the enzyme arginase-2, and arginase activity is essential for the immunosuppressive properties of these cells because molecular inhibition of this enzyme or supplementation with L-arginine overrides immunosuppression. In addition, the ablation of CD71(+) cells in neonatal mice, or the decline in number of these cells as postnatal development progresses parallels the loss of suppression, and restored resistance to the perinatal pathogens Listeria monocytogenes and Escherichia coli. However, CD71(+) cell-mediated susceptibility to infection is counterbalanced by CD71(+) cell-mediated protection against aberrant immune cell activation in the intestine, where colonization with commensal microorganisms occurs swiftly after parturition. Conversely, circumventing such colonization by using antimicrobials or gnotobiotic germ-free mice overrides these protective benefits. Thus, CD71(+) cells quench the excessive inflammation induced by abrupt colonization with commensal microorganisms after parturition. This finding challenges the idea that the susceptibility of neonates to infection reflects immune-cell-intrinsic defects and instead highlights processes that are developmentally more essential and inadvertently mitigate innate immune protection. We anticipate that these results will spark renewed investigation into the need for immunosuppression in neonates, as well as improved strategies for augmenting host defence in this vulnerable population.


Assuntos
Antígenos CD/metabolismo , Células Eritroides/imunologia , Infecções por Escherichia coli/imunologia , Tolerância Imunológica/imunologia , Listeriose/imunologia , Receptores da Transferrina/metabolismo , Animais , Animais Recém-Nascidos , Arginase/genética , Arginase/metabolismo , Suscetibilidade a Doenças/imunologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células Eritroides/enzimologia , Escherichia coli/imunologia , Feminino , Sangue Fetal/citologia , Humanos , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/genética , Listeria monocytogenes/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo
15.
Nat Methods ; 10(8): 795-803, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23749299

RESUMO

Investigation of immune-cell differentiation and function is limited by shortcomings of suitable and scalable experimental systems. Here we show that retroviral delivery of an estrogen-regulated form of Hoxb8 into mouse bone marrow cells can be used along with Flt3 ligand to conditionally immortalize early hematopoietic progenitor cells (Hoxb8-FL cells). Hoxb8-FL cells have lost self-renewal capacity and potential to differentiate into megakaryocytes and erythrocytes but retain the potential to differentiate into myeloid and lymphoid cells. They differentiate in vitro and in vivo into macrophages, granulocytes, dendritic cells, B lymphocytes and T lymphocytes that are phenotypically and functionally indistinguishable from their primary counterparts. Quantitative in vitro assays indicate that myeloid and B-cell potential of Hoxb8-FL cells is comparable to that of primary lymphoid-primed multipotent progenitors, whereas T-cell potential is diminished. The simplicity of this system and the unlimited proliferative capacity of Hoxb8-FL cells will enable studies of immune-cell differentiation and function.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Linfócitos/citologia , Células Mieloides/citologia , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Feminino , Citometria de Fluxo , Linfócitos/ultraestrutura , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/ultraestrutura , Análise de Componente Principal , Proteômica
16.
Mol Immunol ; 51(2): 234-44, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22487722

RESUMO

The heterodimeric IL-12 cytokine family is characterized by the sharing of three α (p19, p28, p35) and two ß (p40 and Ebi3) subunits, and includes IL-12 (p35/p40), IL-23 (p19/p40), IL-27 (p28/Ebi3) and IL-35 (p35/Ebi3). In this study, the dimerization interfaces of IL-12 family members were characterized, with emphasis on IL-35. Ebi3 and p35 subunits from human and mouse paired effectively with each other, indicating there is no species barrier to IL-35 dimerization and suggesting a conserved dimerization interface. Specific p35 residues that contribute to formation of the IL-12 interface were assessed for their contribution to the IL-35 interface, and candidate Ebi3 residues were screened for their contribution to both IL-27 and IL-35 interfaces. Several residues were identified as critical to the IL-12 or IL-27 interfaces. Conversely, no single mutation was identified that completely disrupts p35/Ebi3 pairing. Linear alanine scanning mutagenesis on both p35 and Ebi3 subunits was performed, focusing on residues that are conserved between the mouse and human proteins. Additionally, a structure-based alanine-scanning approach in which mutations were clustered based on proximitiy was performed on the p35 subunit. Both approaches suggest that IL-35 has distinct criteria for subunit pairing and is remarkabley less sensitive to structural perturbation than IL-12 and IL-27. Additionally, studies using a panel of anti-p35 and anti-Ebi3 antibodies indicate differential availability of epitopes within IL-12 family members that share these subunits, suggesting that IL-35 has distinct structural features, relative to IL-12 and IL-27. These results may be useful in future directed therapeutic targeting of IL-12 family members.


Assuntos
Interleucina-12/química , Interleucina-12/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Sequência de Aminoácidos , Animais , Sequência Conservada , Dimerização , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Humanos , Immunoblotting , Imunoprecipitação , Subunidade p35 da Interleucina-12/química , Subunidade p35 da Interleucina-12/metabolismo , Interleucinas/química , Interleucinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Antígenos de Histocompatibilidade Menor , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Receptores de Citocinas/química , Receptores de Citocinas/metabolismo , Alinhamento de Sequência , Especificidade da Espécie
17.
Nat Immunol ; 13(3): 290-9, 2012 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-22306691

RESUMO

Interleukin 35 (IL-35) belongs to the IL-12 family of heterodimeric cytokines but has a distinct functional profile. IL-35 suppresses T cell proliferation and converts naive T cells into IL-35-producing induced regulatory T cells (iTr35 cells). Here we found that IL-35 signaled through a unique heterodimer of receptor chains IL-12Rß2 and gp130 or homodimers of each chain. Conventional T cells were sensitive to IL-35-mediated suppression in the absence of one receptor chain but not both receptor chains, whereas signaling through both chains was required for IL-35 expression and conversion into iTr35 cells. Signaling through the IL-35 receptor required the transcription factors STAT1 and STAT4, which formed a unique heterodimer that bound to distinct sites in the promoters of the genes encoding the IL-12 subunits p35 and Ebi3. This unconventional mode of signaling, distinct from that of other members of the IL-12 family, may broaden the spectrum and specificity of IL-35-mediated suppression.


Assuntos
Receptores de Interleucina-1/imunologia , Receptores de Interleucina/imunologia , Transdução de Sinais , Animais , Receptor gp130 de Citocina/imunologia , Interleucinas/imunologia , Camundongos , Camundongos Knockout , Modelos Moleculares , Multimerização Proteica , Estrutura Quaternária de Proteína , Receptores de Interleucina/química , Receptores de Interleucina/deficiência , Receptores de Interleucina/metabolismo , Receptores de Interleucina-1/química , Receptores de Interleucina-1/deficiência , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-12/imunologia , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT4/imunologia
18.
J Immunol ; 186(12): 6661-6, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21576509

RESUMO

Human regulatory T cells (T(reg)) are essential for the maintenance of immune tolerance. However, the mechanisms they use to mediate suppression remain controversial. Although IL-35 has been shown to play an important role in T(reg)-mediated suppression in mice, recent studies have questioned its relevance in human T(reg). In this study, we show that human T(reg) express and require IL-35 for maximal suppressive capacity. Substantial upregulation of EBI3 and IL12A, but not IL10 and TGFB, was observed in activated human T(reg) compared with conventional T cells (T(conv)). Contact-independent T(reg)-mediated suppression was IL-35 dependent and did not require IL-10 or TGF-ß. Lastly, human T(reg)-mediated suppression led to the conversion of the suppressed T(conv) into iTr35 cells, an IL-35-induced T(reg) population, in an IL-35-dependent manner. Thus, IL-35 contributes to human T(reg)-mediated suppression, and its conversion of suppressed target T(conv) into IL-35-induced T(reg) may contribute to infectious tolerance.


Assuntos
Tolerância Imunológica/imunologia , Interleucinas/imunologia , Linfócitos T Reguladores/imunologia , Humanos , Interleucina-10 , Subunidade p35 da Interleucina-12 , Antígenos de Histocompatibilidade Menor , Fator de Crescimento Transformador beta
19.
Nat Immunol ; 11(12): 1093-101, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20953201

RESUMO

Regulatory T cells (T(reg) cells) have a critical role in the maintenance of immunological self-tolerance. Here we show that treatment of naive human or mouse T cells with IL-35 induced a regulatory population, which we call 'iT(R)35 cells', that mediated suppression via IL-35 but not via the inhibitory cytokines IL-10 or transforming growth factor-ß (TGF-ß). We found that iT(R)35 cells did not express or require the transcription factor Foxp3, and were strongly suppressive and stable in vivo. T(reg) cells induced the generation of iT(R)35 cells in an IL-35- and IL-10-dependent manner in vitro and induced their generation in vivo under inflammatory conditions in intestines infected with Trichuris muris and within the tumor microenvironment (B16 melanoma and MC38 colorectal adenocarcinoma), where they contributed to the regulatory milieu. Thus, iT(R)35 cells constitute a key mediator of infectious tolerance and contribute to T(reg) cell-mediated tumor progression. Furthermore, iT(R)35 cells generated ex vivo might have therapeutic utility.


Assuntos
Tolerância Imunológica/imunologia , Interleucinas/imunologia , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/imunologia , Separação Celular , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Immunoblotting , Imunoprecipitação , Interleucinas/metabolismo , Camundongos , Microscopia Confocal , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/citologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo
20.
J Immunol ; 182(10): 6121-8, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19414764

RESUMO

Regulatory T cells (T(reg)) are believed to suppress conventional T cell (T(conv)) proliferation in vitro in a contact-dependent, cytokine-independent manner, based in part on experiments in which T(reg) and T(conv) are separated by a permeable membrane. We show that the production of IL-35, a novel inhibitory cytokine expressed by natural T(reg), increases substantially following contact with T(conv). Surprisingly, T(reg) were able to mediate potent suppression of T(conv) across a permeable membrane when placed in direct contact with T(conv) in the upper chamber of a Transwell plate. Suppression was IL-35 and IL-10 dependent, and T(conv) activation was required for maximal potentiation of T(reg) suppression. These data suggest that it is the induction of suppression, rather than the function of T(reg) that is obligatorily contact dependent.


Assuntos
Comunicação Celular/imunologia , Interleucina-10/imunologia , Interleucinas/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T/imunologia , Animais , Western Blotting , Proliferação de Células , Citometria de Fluxo , Imunoprecipitação , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...