Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood Adv ; 8(10): 2384-2397, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38489234

RESUMO

ABSTRACT: Sézary syndrome (SS) is an aggressive leukemic expansion of skin-derived malignant CD4+ T cells. Drug monotherapy often results in disease relapse because of the heterogenous nature of malignant CD4+ T cells, but how therapies can be optimally combined remains unclear because of limitations in understanding the disease pathogenesis. We identified immunologic transitions that interlink mycosis fungoides with SS using single-cell transcriptome analysis in parallel with high-throughput T-cell receptor sequencing. Nascent peripheral CD4+ T cells acquired a distinct profile of transcription factors and trafficking receptors that gave rise to antigenically mature Sézary cells. The emergence of malignant CD4+ T cells coincided with the accumulation of dysfunctional monocytes with impaired fragment crystallizable γ-dependent phagocytosis, decreased responsiveness to cytokine stimulation, and limited repertoire of intercellular interactions with Sézary cells. Type I interferon supplementation when combined with a monoclonal antibody targeting the chemokine receptor type 4 (CCR4), unleashed monocyte induced phagocytosis and eradication of Sézary cells in vitro. In turn, coadministration of interferon-α with the US Food and Drug Administration-approved anti-CCR4 antibody, mogamulizumab, in patients with SS induced marked depletion of peripheral malignant CD4+ T cells. Importantly, residual CD4+ T cells after Sézary cell ablation lacked any immunologic shifts. These findings collectively unveil an auxiliary role for augmenting monocytic activity during mogamulizumab therapy in the treatment of SS and underscore the importance of targeted combination therapy in this disease.


Assuntos
Interferon Tipo I , Monócitos , Receptores CCR4 , Síndrome de Sézary , Humanos , Síndrome de Sézary/tratamento farmacológico , Síndrome de Sézary/imunologia , Monócitos/metabolismo , Monócitos/imunologia , Interferon Tipo I/metabolismo , Receptores CCR4/antagonistas & inibidores , Receptores CCR4/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais Humanizados/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia
2.
J Invest Dermatol ; 2023 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-38036289

RESUMO

Cutaneous T-cell lymphomas are a heterogeneous group of neoplasms originating in the skin, with mycosis fungoides (MF) and Sézary syndrome (SS) representing the most common variants. The cellular origin of cutaneous lymphomas has remained controversial owing to their immense phenotypic heterogeneity that obfuscates lineage reconstruction on the basis of classical surface biomarkers. To overcome this heterogeneity and reconstruct the differentiation trajectory of malignant cells in MF and SS, TCR sequencing was performed in parallel with targeted transcriptomics at the single-cell resolution among cutaneous samples in MF and SS. Unsupervised lineage reconstruction showed that Sézary cells exist as a population of CD4+ T cells distinct from those in patch, plaque, and tumor MF. Further investigation of malignant cell heterogeneity in SS showed that Sézary cells phenotypically comprised at least 3 subsets on the basis of differential proliferation potentials and expression of exhaustion markers. A T helper 1-polarized cell type, intermediate cell type, and exhausted T helper 2-polarized cell type were identified, with T helper 1- and T helper 2-polarized cells displaying divergent proliferation potentials. Collectively, these findings provide evidence to clarify the relationship between MF and SS and reveal cell subsets in SS that suggest a possible mechanism for therapeutic resistance.

3.
Cell Rep ; 42(11): 113323, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37889750

RESUMO

Intestinal colonization by antigenically foreign microbes necessitates expanded peripheral immune tolerance. Here we show commensal microbiota prime expansion of CD4 T cells unified by the Kruppel-like factor 2 (KLF2) transcriptional regulator and an essential role for KLF2+ CD4 cells in averting microbiota-driven intestinal inflammation. CD4 cells with commensal specificity in secondary lymphoid organs and intestinal tissues are enriched for KLF2 expression, and distinct from FOXP3+ regulatory T cells or other differentiation lineages. Mice with conditional KLF2 deficiency in T cells develop spontaneous rectal prolapse and intestinal inflammation, phenotypes overturned by eliminating microbiota or reconstituting with donor KLF2+ cells. Activated KLF2+ cells selectively produce IL-10, and eliminating IL-10 overrides their suppressive function in vitro and protection against intestinal inflammation in vivo. Together with reduced KLF2+ CD4 cell accumulation in Crohn's disease, a necessity for the KLF2+ subpopulation of T regulatory type 1 (Tr1) cells in sustaining commensal tolerance is demonstrated.


Assuntos
Linfócitos T CD4-Positivos , Microbiota , Camundongos , Animais , Interleucina-10/metabolismo , Linfócitos T Reguladores , Fatores de Transcrição/metabolismo , Inflamação/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo
4.
J Invest Dermatol ; 142(3 Pt A): 512-515, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35184799

RESUMO

Immune-modulatory therapies are widely appreciated to rejuvenate host antitumor immunity and improve mortality in solid-organ cancers. Targeting the exhausted markers such as PD-1, CTLA4, TIM3, LAG3 are particularly attractive owing to the activation of the immune response. However, their role in cutaneous T-cell lymphomas is less defined owing to the expression of those exhausted markers on both nonmalignant and malignant lymphocytes. In a new article of the Journal of Investigative Dermatology, Han et al. (2021) showed that microRNAs in malignant T cells could regulate the expression of PD-1, CTLA4, TIM3, and LAG3 and simultaneously mediate evasion from immune surveillance. These findings get us one step closer in our further investigation of whether those molecules could be targeted therapeutically.


Assuntos
Linfoma Cutâneo de Células T , MicroRNAs , Biomarcadores , Antígeno CTLA-4 , Receptor Celular 2 do Vírus da Hepatite A/genética , Receptor Celular 2 do Vírus da Hepatite A/imunologia , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Humanos , MicroRNAs/genética , Receptor de Morte Celular Programada 1/genética , Navios , Linfócitos T/imunologia
5.
Br J Dermatol ; 187(2): 265-267, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35191024

RESUMO

Six out of 12 Sézary patients shared one clonotype (TRAV13-1*01-TRAJ49*01-TRBV20-1*01-TRBJ2-3*01). TRBV20-1*01 (also known as Vb2) that binds toxic shock syndrome toxin-1 was utilized by Sézary cells among half of the cohort, which would be expected for a common unifying origin.


Assuntos
Síndrome de Sézary , Neoplasias Cutâneas , Humanos , Linfócitos , Fenótipo , Receptores de Antígenos de Linfócitos T/genética
6.
Cancers (Basel) ; 13(23)2021 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-34885092

RESUMO

Cancer progression in mycosis fungoides, the most common form of cutaneous T-cell lymphoma, occurs in a predictable, sequential pattern that starts from patches and that evolves to plaques and later to tumors. Therefore, unlocking the relationship between the microarchitecture of mycosis fungoides and the clinical counterparts of that microstructure represents important steps for the design of targeted therapies. Using multispectral fluorescent imaging, we show that the progression of mycosis fungoides from plaque to tumor parallels the cutaneous expansion of the malignant CD4+ T cells that express TOX. The density of exhausted BTLA+ CD4+ T cells around malignant CD4+TOX+ cells was higher in tumors than it was in plaques, suggesting that undesired safeguards are in place within the tumor microenvironment that prevent immune activation and subsequent cancer eradication. Overriding the CD47 checkpoint with an intralesional SIRPαFc fusion decoy receptor induced the resolution of mycosis fungoides in patients that paralleled an amplified expansion of NK and CD8+ T cells in addition to a reduction of the exhausted BTLA+ CD4+ T cells that were engaged in promiscuous intercellular interactions. These therapeutic benefits of the CD47 blockade were further unleashed by adjuvant interferon-α, which stimulates cytotoxic cells, underscoring the importance of an inflamed microenvironment in facilitating the response to immunotherapy. Collectively, these findings support CD47 as a therapeutic target in treating mycosis fungoides and demonstrate a synergistic role of interferon-α in exploiting these clinical benefits.

7.
Cell Host Microbe ; 25(3): 404-417.e6, 2019 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-30870622

RESUMO

Mucosal barriers are densely colonized by pathobiont microbes such as Candida albicans, capable of invasive disseminated infection. However, systemic infections occur infrequently in healthy individuals, suggesting that pathobiont commensalism may elicit host benefits. We show that intestinal colonization with C. albicans drives systemic expansion of fungal-specific Th17 CD4+ T cells and IL-17 responsiveness by circulating neutrophils, which synergistically protect against C. albicans invasive infection. Protection conferred by commensal C. albicans requires persistent fungal colonization and extends to other extracellular invasive pathogens such as Staphylococcus aureus. However, commensal C. albicans does not protect against intracellular influenza virus infection and exacerbates allergic airway inflammation susceptibility, indicating that positively calibrating systemic Th17 responses is not uniformly beneficial. Thus, systemic Th17 inflammation driven by CD4+ T cells responsive to tonic stimulation by commensal C. albicans improves host defense against extracellular pathogens, but with potentially harmful immunological consequences.


Assuntos
Candida albicans/imunologia , Candidíase Invasiva/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Células Th17/imunologia , Animais , Proteção Cruzada , Modelos Animais de Doenças , Interleucina-17/metabolismo , Camundongos , Infecções por Orthomyxoviridae/prevenção & controle , Infecções Estafilocócicas/prevenção & controle
8.
PLoS Pathog ; 13(11): e1006684, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29145516

RESUMO

Pregnant women, and their fetal offspring, are uniquely susceptible to Zika virus and other microbial pathogens capable of congenital fetal infection. Unavoidable exposure to Zika virus in endemic areas underscores the need for identifying at-risk individuals, and protecting expecting mothers and their fetal offspring against prenatal infection. Here we show that primary Zika virus asymptomatic infection in mice confers protection against re-infection, and that these protective benefits are maintained during pregnancy. Zika virus recovery was sharply reduced in maternal tissues and amongst fetal concepti after prenatal challenge in mothers with resolved subclinical infection prior to pregnancy compared with mice undergoing primary prenatal infection. These benefits coincide with expanded accumulation of viral-specific antibodies in maternal serum and fetal tissues that protect against infection by the identical or heterologous Zika virus genotype strains. Thus, preconceptual infection primes Zika virus-specific antibodies that confer cross-genotype protection against re-infection during pregnancy.


Assuntos
Infecções Assintomáticas/epidemiologia , Complicações Infecciosas na Gravidez/virologia , Infecção por Zika virus/virologia , Animais , Anticorpos Antivirais , Coinfecção/etiologia , Modelos Animais de Doenças , Feminino , Humanos , Recém-Nascido , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Gravidez , Complicações Infecciosas na Gravidez/epidemiologia , Infecção por Zika virus/prevenção & controle
9.
Cell Host Microbe ; 22(6): 809-816.e4, 2017 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-29174402

RESUMO

Commensal intestinal microbes are collectively beneficial in preventing local tissue injury and augmenting systemic antimicrobial immunity. However, given the near-exclusive focus on bacterial species in establishing these protective benefits, the contributions of other types of commensal microbes remain poorly defined. Here, we show that commensal fungi can functionally replace intestinal bacteria by conferring protection against injury to mucosal tissues and positively calibrating the responsiveness of circulating immune cells. Susceptibility to colitis and influenza A virus infection occurring upon commensal bacteria eradication is efficiently overturned by mono-colonization with either Candida albicans or Saccharomyces cerevisiae. The protective benefits of commensal fungi are mediated by mannans, a highly conserved component of fungal cell walls, since intestinal stimulation with this moiety alone overrides disease susceptibility in mice depleted of commensal bacteria. Thus, commensal enteric fungi safeguard local and systemic immunity by providing tonic microbial stimulation that can functionally replace intestinal bacteria.


Assuntos
Resistência à Doença , Fungos/crescimento & desenvolvimento , Fungos/imunologia , Microbioma Gastrointestinal , Imunidade Celular , Imunidade Inata , Simbiose , Animais , Colite/prevenção & controle , Camundongos , Infecções por Orthomyxoviridae/prevenção & controle
10.
Cell Rep ; 18(9): 2088-2095, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28249155

RESUMO

Genetic S6K1 inactivation can induce apoptosis in PTEN-deficient cells. We analyzed the therapeutic potential of S6K1 inhibitors in PTEN-deficient T cell leukemia and glioblastoma. Results revealed that the S6K1 inhibitor LY-2779964 was relatively ineffective as a single agent, while S6K1-targeting AD80 induced cytotoxicity selectively in PTEN-deficient cells. In vivo, AD80 rescued 50% of mice transplanted with PTEN-deficient leukemia cells. Cells surviving LY-2779964 treatment exhibited inhibitor-induced S6K1 phosphorylation due to increased mTOR-S6K1 co-association, which primed the rapid recovery of S6K1 signaling. In contrast, AD80 avoided S6K1 phosphorylation and mTOR co-association, resulting in durable suppression of S6K1-induced signaling and protein synthesis. Kinome analysis revealed that AD80 coordinately inhibits S6K1 together with the TAM family tyrosine kinase AXL. TAM suppression by BMS-777607 or genetic knockdown potentiated cytotoxic responses to LY-2779964 in PTEN-deficient glioblastoma cells. These results reveal that combination targeting of S6K1 and TAMs is a potential strategy for treatment of PTEN-deficient malignancy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , PTEN Fosfo-Hidrolase/deficiência , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Aminopiridinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes/métodos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Leucemia de Células T/tratamento farmacológico , Leucemia de Células T/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Piridonas/farmacologia , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
11.
Cell Rep ; 17(7): 1783-1794, 2016 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-27829150

RESUMO

Self-reactive CD4 T cells are incompletely deleted during thymic development, and their peripheral seeding highlights the need for additional safeguards to avert autoimmunity. Here, we show an essential role for the coinhibitory molecule programmed death-1 (PD-1) in silencing the activation of high-affinity autoreactive CD4 T cells. Each wave of self-reactive CD4 T cells that escapes thymic deletion autonomously upregulates PD-1 to maintain self-tolerance. By tracking the progeny derived from individual autoreactive CD4 T cell clones, we demonstrate that self-reactive cells with the greatest autoimmune threat and highest self-antigen affinity express the most PD-1. Reciprocally, PD-1 deprivation unleashes high-affinity self-reactive CD4 T cells in target tissues to exacerbate neuronal inflammation and autoimmune diabetes. Reliance on PD-1 to actively maintain self-tolerance may explain why exploiting this pathway by cancerous cells and invasive microbes efficiently subverts protective immunity, and why autoimmune side effects can develop after PD-1-neutralizing checkpoint therapies.


Assuntos
Autoimunidade , Linfócitos T CD4-Positivos/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Animais , Autoantígenos/imunologia , Proliferação de Células , Células Clonais , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/metabolismo , Tolerância a Antígenos Próprios/imunologia
12.
Immunity ; 44(5): 1085-7, 2016 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-27192574

RESUMO

Pregnancy uniquely allows genetically discordant tissues of the mother and child to intimately coexist in harmony. In this issue of Immunity, Ou and colleagues show that hepatitis B virus exploits these naturally occurring immune tolerance pathways to establish persistent postnatal infection in offspring.


Assuntos
Tolerância Imunológica , Mães , Feminino , Humanos , Gravidez
13.
Chimerism ; 6(1-2): 8-20, 2015 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-26517600

RESUMO

Compulsory exposure to genetically foreign maternal tissue imprints in offspring sustained tolerance to noninherited maternal antigens (NIMA). Immunological tolerance to NIMA was first described by Dr. Ray D. Owen for women genetically negative for erythrocyte rhesus (Rh) antigen with reduced sensitization from developmental Rh exposure by their mothers. Extending this analysis to HLA haplotypes has uncovered the exciting potential for therapeutically exploiting NIMA-specific tolerance naturally engrained in mammalian reproduction for improved clinical outcomes after allogeneic transplantation. Herein, we summarize emerging scientific concepts stemming from tolerance to NIMA that includes postnatal maintenance of microchimeric maternal origin cells in offspring, expanded accumulation of immune suppressive regulatory T cells with NIMA-specificity, along with teleological benefits and immunological consequences of NIMA-specific tolerance conserved across mammalian species.


Assuntos
Antígenos HLA/imunologia , Tolerância Imunológica , Memória Imunológica , Sistema do Grupo Sanguíneo Rh-Hr/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos HLA/história , História do Século XX , História do Século XXI , Humanos , Sistema do Grupo Sanguíneo Rh-Hr/história
14.
Aging Cell ; 14(6): 1122-6, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26248606

RESUMO

Aging confers increased susceptibility to common pathogens including influenza A virus. Despite shared vulnerability to infection with advancing age in humans and rodents, the relatively long time required for immune senescence to take hold practically restricts the use of naturally aged mice to investigate aging-induced immunological shifts. Here, we show accelerated aging Lmna(Dhe) mice with spontaneous mutation in the nuclear scaffolding protein, lamin A, replicate infection susceptibility, and substantial immune cell shifts that occur with advancing age. Naturally aged (≥ 20 month) and 2- to 3-month-old Lmna(Dhe) mice share near identically increased influenza A susceptibility compared with age-matched Lmna(WT) control mice. Increased mortality and higher viral burden after influenza infection in Lmna(Dhe) mice parallel reduced accumulation of lung alveolar macrophage cells, systemic expansion of immune suppressive Foxp3⁺ regulatory T cells, and skewed immune dominance among viral-specific CD8⁺T cells similar to the immunological phenotype of naturally aged mice. Thus, aging-induced infection susceptibility and immune senescence are replicated in accelerated aging Lmna(Dhe) mice.


Assuntos
Envelhecimento/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Pulmão/imunologia , Infecções por Orthomyxoviridae/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Suscetibilidade a Doenças/imunologia , Lamina Tipo A/genética , Pulmão/virologia , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Progéria/genética , Progéria/imunologia , Linfócitos T Reguladores/imunologia
15.
Cell ; 162(3): 505-15, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-26213383

RESUMO

Exposure to maternal tissue during in utero development imprints tolerance to immunologically foreign non-inherited maternal antigens (NIMA) that persists into adulthood. The biological advantage of this tolerance, conserved across mammalian species, remains unclear. Here, we show maternal cells that establish microchimerism in female offspring during development promote systemic accumulation of immune suppressive regulatory T cells (Tregs) with NIMA specificity. NIMA-specific Tregs expand during pregnancies sired by males expressing alloantigens with overlapping NIMA specificity, thereby averting fetal wastage triggered by prenatal infection and non-infectious disruptions of fetal tolerance. Therefore, exposure to NIMA selectively enhances reproductive success in second-generation females carrying embryos with overlapping paternally inherited antigens. These findings demonstrate that genetic fitness, canonically thought to be restricted to Mendelian inheritance, is enhanced in female placental mammals through vertically transferred maternal cells that promote conservation of NIMA and enforce cross-generational reproductive benefits.


Assuntos
Feto/imunologia , Aptidão Genética , Tolerância Imunológica , Mamíferos/fisiologia , Gravidez/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos/imunologia , Quimerismo , Feminino , Humanos , Masculino , Mamíferos/imunologia , Camundongos , Placenta/imunologia
16.
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
17.
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
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...