Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 220(10)2023 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-37638887

RESUMO

In this new review, Rick Maizels and Bill Gause summarize how type 2 immune responses combat helminth parasites through novel mechanisms, coordinating multiple innate and adaptive cell and molecular players that can eliminate infection and repair-resultant tissue damage.


Assuntos
Helmintíase , Helmintos , Animais , Helmintíase/imunologia
2.
STAR Protoc ; 4(2): 102208, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37086410

RESUMO

Preserving structurally intact tissue through proper tissue fixation and cryopreservation minimizes tissue autolysis, desiccation, and enzymatic degradation. In this protocol, we describe the use of an optimized fresh freezing technique that cryopreserves the tissue and favors the retention of the natural protein structure of antigens. We also detail the use of cold, low-concentration paraformaldehyde (PFA) fixation to enhance tissue morphology and detection of fluorescent proteins, such as GFP and TdTomato, in tissues of genetically engineered mice. For complete details on the use and execution of this protocol, please refer to Chen et al. (2022)1 and El-Naccache et al. (2022).2.

3.
J Immunol ; 209(11): 2160-2171, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36426972

RESUMO

More than 2 billion people worldwide are infected with helminths. Thus, it is possible for individuals to experience concomitant infection with helminth and intracellular microbes. Although the helminth-induced type 2 response can suppress type 1 proinflammatory responses required for the immunity against intracellular pathogens in the context of a coinfection, conflicting evidence suggest that helminth infection can enhance antimicrobial immunity. Using a coinfection model with the intestinal helminth Heligmosomoides polygyrus followed by infection with Toxoplasma gondii in Mus Musculus, we showed that the complex and dynamic effect of helminth infection is highly suppressive during the innate phase (days 0-3) of T. gondii infection and less stringent during the acute phase (d10). Helminth coinfection had a strong suppressive effect on the neutrophil, monocytic, and early IFN-γ/IL-12 responses. The IFN-γ response was later restored by compensatory production from T cells despite decreased effector differentiation of T. gondii-specific CD8 T cells. In accordance with the attenuated IFN-γ response, parasite loads were elevated during the acute phase (d10) of T. gondii infection but were transiently controlled by the compensatory T cell response. Unexpectedly, 40% of helminth-coinfected mice exhibited a sustained weight loss phenotype during the postacute phase (d14-18) that was not associated with T. gondii outgrowth, indicating that coinfection led to decreased disease tolerance during T. gondii infection. Our work uncovers the dynamic nature of the helminth immunomodulatory effects on concomitant infections or immune responses and unveils a loss of disease tolerance phenotype triggered by coinfection with intestinal helminth.


Assuntos
Coinfecção , Nematospiroides dubius , Toxoplasma , Toxoplasmose , Animais , Camundongos , Tolerância Imunológica
4.
Proc Natl Acad Sci U S A ; 119(35): e2123267119, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35994660

RESUMO

The pregnant uterus is an immunologically rich organ, with dynamic changes in the inflammatory milieu and immune cell function underlying key stages of pregnancy. Recent studies have implicated dysregulated expression of the interleukin-1 (IL-1) family cytokine, IL-33, and its receptor, ST2, in poor pregnancy outcomes in women, including recurrent pregnancy loss, preeclampsia, and preterm labor. How IL-33 supports pregnancy progression in vivo is not well understood. Here, we demonstrate that maternal IL-33 signaling critically regulates uterine tissue remodeling and immune cell function during early pregnancy in mice. IL-33-deficient dams exhibit defects in implantation chamber formation and decidualization, and abnormal vascular remodeling during early pregnancy. These defects coincide with delays in early embryogenesis, increased resorptions, and impaired fetal and placental growth by late pregnancy. At a cellular level, myometrial fibroblasts, and decidual endothelial and stromal cells, are the main IL-33+ cell types in the uterus during decidualization and early placentation, whereas ST2 is expressed by uterine immune populations associated with type 2 immune responses, including ILC2s, Tregs, CD4+ T cells, M2- and cDC2-like myeloid cells, and mast cells. Early pregnancy defects in IL-33-deficient dams are associated with impaired type 2 cytokine responses by uterine lymphocytes and fewer Arginase-1+ macrophages in the uterine microenvironment. Collectively, our data highlight a regulatory network, involving crosstalk between IL-33-producing nonimmune cells and ST2+ immune cells at the maternal-fetal interface, that critically supports pregnancy progression in mice. This work has the potential to advance our understanding of how IL-33 signaling may support optimal pregnancy outcomes in women.


Assuntos
Interleucina-33 , Placenta , Placentação , Útero , Animais , Decídua/irrigação sanguínea , Decídua/citologia , Decídua/crescimento & desenvolvimento , Decídua/imunologia , Feminino , Feto/imunologia , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/deficiência , Interleucina-33/imunologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Placenta/imunologia , Placenta/metabolismo , Gravidez , Útero/irrigação sanguínea , Útero/crescimento & desenvolvimento , Útero/imunologia , Útero/metabolismo
5.
Cell Rep ; 40(5): 111150, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35926464

RESUMO

Intestinal nematode parasites can cross the epithelial barrier, causing tissue damage and release of danger-associated molecular patterns (DAMPs) that may promote host protective type 2 immunity. We investigate whether adenosine binding to the A2B adenosine receptor (A2BAR) on intestinal epithelial cells (IECs) plays an important role. Specific blockade of IEC A2BAR inhibits the host protective memory response to the enteric helminth, Heligmosomoides polygyrus bakeri (Hpb), including disruption of granuloma development at the host-parasite interface. Memory T cell development is blocked during the primary response, and transcriptional analyses reveal profound impairment of IEC activation. Extracellular ATP is visualized 24 h after inoculation and is shown in CD39-deficient mice to be critical for the adenosine production mediating the initiation of type 2 immunity. Our studies indicate a potent adenosine-mediated IEC pathway that, along with the tuft cell circuit, is critical for the activation of type 2 immunity.


Assuntos
Adenosina , Receptor A2B de Adenosina , Adenosina/metabolismo , Trifosfato de Adenosina , Animais , Células Epiteliais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor A2B de Adenosina/metabolismo
6.
Purinergic Signal ; 18(3): 345-358, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35838900

RESUMO

Extracellular adenosine is a biologically active signaling molecule that accumulates at sites of metabolic stress in sepsis. Extracellular adenosine has potent immunosuppressive effects by binding to and activating G protein-coupled A2A adenosine receptors (A2AARs) on the surface of neutrophils. A2AAR signaling reproduces many of the phenotypic changes in neutrophils that are characteristic of sepsis, including decreased degranulation, impaired chemotaxis, and diminished ability to ingest and kill bacteria. We hypothesized that A2AARs also suppress neutrophil aging, which precedes cell death, and N1 to N2 polarization. Using human neutrophils isolated from healthy subjects, we demonstrate that A2AAR stimulation slows neutrophil aging, suppresses cell death, and promotes the polarization of neutrophils from an N1 to N2 phenotype. Using genetic knockout and pharmacological blockade, we confirmed that A2AARs decrease neutrophil aging in murine sepsis induced by cecal ligation and puncture. A2AARs expression is increased in neutrophils from septic patients compared to healthy subject but A2AAR expression fails to correlate with aging or N1/N2 polarization. Our data reveals that A2AARs regulate neutrophil aging in healthy but not septic neutrophils.


Assuntos
Neutrófilos , Sepse , Adenosina , Envelhecimento , Animais , Humanos , Camundongos , Camundongos Knockout , Neutrófilos/metabolismo , Fenótipo , Receptor A2A de Adenosina/metabolismo
7.
Cell Rep ; 38(2): 110215, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35021079

RESUMO

Macrophages are known to mediate anti-helminth responses, but it remains uncertain which subsets are involved or how macrophages actually kill helminths. Here, we show rapid monocyte recruitment to the lung after infection with the nematode parasite Nippostrongylus brasiliensis. In this inflamed tissue microenvironment, these monocytes differentiate into an alveolar macrophage (AM)-like phenotype, expressing both SiglecF and CD11c, surround invading parasitic larvae, and preferentially kill parasites in vitro. Monocyte-derived AMs (Mo-AMs) express type 2-associated markers and show a distinct remodeling of the chromatin landscape relative to tissue-derived AMs (TD-AMs). In particular, they express high amounts of arginase-1 (Arg1), which we demonstrate mediates helminth killing through L-arginine depletion. These studies indicate that recruited monocytes are selectively programmed in the pulmonary environment to express AM markers and an anti-helminth phenotype.


Assuntos
Pulmão/imunologia , Macrófagos Alveolares/imunologia , Infecções por Strongylida/imunologia , Animais , Arginase/metabolismo , Diferenciação Celular , Citocinas , Feminino , Pulmão/parasitologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nippostrongylus , Infecções por Strongylida/parasitologia
8.
FASEB J ; 35(11): e21935, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34591327

RESUMO

Inosine monophosphate (IMP) is the intracellular precursor for both adenosine monophosphate and guanosine monophosphate and thus plays a central role in intracellular purine metabolism. IMP can also serve as an extracellular signaling molecule, and can regulate diverse processes such as taste sensation, neutrophil function, and ischemia-reperfusion injury. How IMP regulates inflammation induced by bacterial products or bacteria is unknown. In this study, we demonstrate that IMP suppressed tumor necrosis factor (TNF)-α production and augmented IL-10 production in endotoxemic mice. IMP exerted its effects through metabolism to inosine, as IMP only suppressed TNF-α following its CD73-mediated degradation to inosine in lipopolysaccharide-activated macrophages. Studies with gene targeted mice and pharmacological antagonism indicated that A2A , A2B, and A3 adenosine receptors are not required for the inosine suppression of TNF-α production. The inosine suppression of TNF-α production did not require its metabolism to hypoxanthine through purine nucleoside phosphorylase or its uptake into cells through concentrative nucleoside transporters indicating a role for alternative metabolic/uptake pathways. Inosine augmented IL-ß production by macrophages in which inflammasome was activated by lipopolysaccharide and ATP. In contrast to its effects in endotoxemia, IMP failed to affect the inflammatory response to abdominal sepsis and pneumonia. We conclude that extracellular IMP and inosine differentially regulate the inflammatory response.


Assuntos
Endotoxemia/metabolismo , Inosina Monofosfato/metabolismo , Inosina/metabolismo , Pneumonia Pneumocócica/metabolismo , Streptococcus pneumoniae , Antagonistas do Receptor A2 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/farmacologia , Animais , Modelos Animais de Doenças , Interleucina-10/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica/microbiologia , Quinazolinas/farmacologia , Receptor A2A de Adenosina/metabolismo , Receptor A2B de Adenosina/metabolismo , Receptor A3 de Adenosina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Triazóis/farmacologia , Fator de Necrose Tumoral alfa/biossíntese
9.
Trends Immunol ; 42(2): 151-164, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33386241

RESUMO

Type 2 immune responses are typically associated with protection against helminth infections and also with harmful inflammation in response to allergens. Recent advances have revealed that type 2 immunity also contributes to sterile inflammation, cancer, and microbial infections. However, the early events that initiate type 2 immune responses remain poorly defined. New insights reveal major contributions from danger-associated molecular patterns (DAMPs) in the initiation of type 2 immune responses. In this review, we examine the molecules released by the host and pathogens and the role they play in mediating the initiation of mammalian innate type 2 immune responses under a variety of conditions.


Assuntos
Helmintíase , Imunidade Inata , Alarminas , Alérgenos , Animais , Humanos , Inflamação
10.
Trends Mol Med ; 27(4): 345-364, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33495068

RESUMO

Helminths are an emerging source of therapeutics for dysregulated inflammatory diseases. Excretory/secretory (ES) molecules, released during infection, are responsible for many of these immunomodulatory effects and are likely to have evolved as a means for parasite survival in the host. While the mechanisms of action of these molecules have not been fully defined, evidence demonstrates that they target various pathways in the immune response, ranging from initiation to effector cell modulation. These molecules are applied in controlling specific effector mechanisms of type 1 and type 2 immune responses. Recently, studies have further focused on their therapeutic potential in specific disease models. Here we review recent findings on ES molecule modulation of immune functions, specifically highlighting their clinical implications for future use in inflammatory disease therapeutics.


Assuntos
Antígenos de Helmintos/imunologia , Helmintos/imunologia , Imunomodulação , Inflamação/terapia , Animais , Helmintos/metabolismo , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunidade , Linfócitos T Reguladores
11.
Cancer Res ; 81(3): 698-712, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33239426

RESUMO

Despite the promising clinical benefit of targeted and immune checkpoint blocking therapeutics, current strategies have limited success in breast cancer, indicating that additional inhibitory pathways are required to complement existing therapeutics. TAM receptors (Tyro-3, Axl, and Mertk) are often correlated with poor prognosis because of their capacities to sustain an immunosuppressive environment. Here, we ablate Axl on tumor cells using CRISPR/Cas9 gene editing, and by targeting Mertk in the tumor microenvironment (TME), we observed distinct functions of TAM as oncogenic kinases, as well as inhibitory immune receptors. Depletion of Axl suppressed cell intrinsic oncogenic properties, decreased tumor growth, reduced the incidence of lung metastasis and increased overall survival of mice when injected into mammary fat pad of syngeneic mice, and demonstrated synergy when combined with anti-PD-1 therapy. Blockade of Mertk function on macrophages decreased efferocytosis, altered the cytokine milieu, and resulted in suppressed macrophage gene expression patterns. Mertk-knockout mice or treatment with anti-Mertk-neutralizing mAb also altered the cellular immune profile, resulting in a more inflamed tumor environment with enhanced T-cell infiltration into tumors and T-cell-mediated cytotoxicity. The antitumor activity from Mertk inhibition was abrogated by depletion of cytotoxic CD8α T cells by using anti-CD8α mAb or by transplantation of tumor cells into B6.CB17-Prkdc SCID mice. Our data indicate that targeting Axl expressed on tumor cells and Mertk in the TME is predicted to have a combinatorial benefit to enhance current immunotherapies and that Axl and Mertk have distinct functional activities that impair host antitumor response. SIGNIFICANCE: This study demonstrates how TAM receptors act both as oncogenic tyrosine kinases and as receptors that mediate immune evasion in cancer progression.


Assuntos
Evasão da Resposta Imune/imunologia , Neoplasias Mamárias Experimentais/imunologia , Proteínas Proto-Oncogênicas/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Transdução de Sinais/imunologia , c-Mer Tirosina Quinase/imunologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Evasão da Resposta Imune/genética , Imunoterapia/métodos , Estimativa de Kaplan-Meier , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/terapia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/genética , c-Mer Tirosina Quinase/genética , c-Mer Tirosina Quinase/metabolismo , Receptor Tirosina Quinase Axl
12.
Nat Rev Immunol ; 20(10): 603-614, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32367051

RESUMO

Type 2 immune responses operate under varying conditions in distinct tissue environments and are crucial for protection against helminth infections and for the maintenance of tissue homeostasis. Here we explore how different layers of heterogeneity influence type 2 immunity. Distinct insults, such as allergens or infections, can induce type 2 immune responses through diverse mechanisms, and this can have heterogeneous consequences, ranging from acute or chronic inflammation to deficits in immune regulation and tissue repair. Technological advances have provided new insights into the molecular heterogeneity of different developmental lineages of type 2 immune cells. Genetic and environmental heterogeneity also contributes to the varying magnitude and quality of the type 2 immune response during infection, which is an important determinant of the balance between pathology and disease resolution. Hence, understanding the mechanisms underlying the heterogeneity of type 2 immune responses between individuals and between different tissues will be crucial for treating diseases in which type 2 immunity is an important component.


Assuntos
Imunidade Inata/imunologia , Animais , Humanos
13.
Cell Host Microbe ; 27(2): 165-168, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-32053785

RESUMO

Recent studies show that neutrophils mediate both tissue damage and host protection in response to multicellular parasites. In this issue of Cell Host & Microbe, Bouchery et al. demonstrate the importance of neutrophil extracellular traps in helminth damage after primary infections.


Assuntos
Armadilhas Extracelulares , Ancylostomatoidea , Animais , Desoxirribonucleases , Neutrófilos
14.
Nat Mater ; 18(3): 289-297, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30664693

RESUMO

Initiation of the innate sterile inflammatory response that can develop in response to microparticle exposure is little understood. Here, we report that a potent type 2 immune response associated with the accumulation of neutrophils, eosinophils and alternatively activated (M2) macrophages was observed in response to sterile microparticles similar in size to wear debris associated with prosthetic implants. Although elevations in interleukin-33 (IL-33) and type 2 cytokines occurred independently of caspase-1 inflammasome signalling, the response was dependent on Bruton's tyrosine kinase (BTK). IL-33 was produced by macrophages and BTK-dependent expression of IL-33 by macrophages was sufficient to initiate the type 2 response. Analysis of inflammation in patient periprosthetic tissue also revealed type 2 responses under aseptic conditions in patients undergoing revision surgery. These findings indicate that microparticle-induced sterile inflammation is initiated by macrophages activated to produce IL-33. They further suggest that both BTK and IL-33 may provide therapeutic targets for wear debris-induced periprosthetic inflammation.


Assuntos
Tirosina Quinase da Agamaglobulinemia/metabolismo , Interleucina-33/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Falha de Prótese , Artroplastia/efeitos adversos , Caspase 1/metabolismo , Humanos , Imunidade Inata/efeitos dos fármacos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Interleucina-33/biossíntese , Macrófagos/imunologia , Transdução de Sinais/efeitos dos fármacos
15.
Cell Rep ; 25(10): 2775-2783.e3, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30517865

RESUMO

Emphysema results in destruction of alveolar walls and enlargement of lung airspaces and has been shown to develop during helminth infections through IL-4R-independent mechanisms. We examined whether interleukin 17A (IL-17A) may instead modulate development of emphysematous pathology in mice infected with the helminth parasite Nippostrongylus brasiliensis. We found that transient elevations in IL-17A shortly after helminth infection triggered subsequent emphysema that destroyed alveolar structures. Furthermore, lung B cells, activated through IL-4R signaling, inhibited early onset of emphysematous pathology. IL-10 and other regulatory cytokines typically associated with B regulatory cell function did not play a major role in this response. Instead, at early stages of the response, B cells produced high levels of the tissue-protective protein, Resistin-like molecule α (RELMα), which then downregulated IL-17A expression. These studies show that transient elevations in IL-17A trigger emphysema and reveal a helminth-induced immune regulatory mechanism that controls IL-17A and the severity of emphysema.


Assuntos
Linfócitos B/metabolismo , Enfisema/imunologia , Enfisema/parasitologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-17/metabolismo , Nippostrongylus/fisiologia , Infecções por Strongylida/parasitologia , Lesão Pulmonar Aguda/complicações , Lesão Pulmonar Aguda/imunologia , Animais , Anticorpos/farmacologia , Regulação para Baixo , Imunidade/efeitos dos fármacos , Pulmão/imunologia , Pulmão/parasitologia , Pulmão/patologia , Camundongos Endogâmicos BALB C , Fenótipo , Receptores de Interleucina-4/metabolismo , Transdução de Sinais
16.
Front Immunol ; 9: 2135, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30386324

RESUMO

Helminths are ubiquitous and have chronically infected vertebrates throughout their evolution. As such helminths have likely exerted considerable selection pressure on our immune systems. The large size of multicellular helminths and their limited replicative capacity in the host necessarily elicits different host protective mechanisms than the immune response evoked by microbial pathogens such as bacteria, viruses and intracellular parasites. The cellular damage resulting from helminth migration through tissues is a major trigger of the type 2 and regulatory immune responses, which activates wound repair mechanisms that increases tissue tolerance to injury and resistance mechanisms that enhance resistance to further colonization with larval stages. While these wound healing and anti-inflammatory responses may be beneficial to the helminth infected host, they may also compromise the host's ability to mount protective immune responses to microbial pathogens. In this review we will first describe helminth-induced tolerance mechanisms that develop in specific organs including the lung and the intestine, and how adaptive immunity may contribute to these responses through differential activation of T cells in the secondary lymphoid organs. We will then integrate studies that have examined how the immune response is modulated in these specific tissues during coinfection of helminths with viruses, protozoa, and bacteria.


Assuntos
Infecções Bacterianas/imunologia , Helmintíase/imunologia , Interações Hospedeiro-Parasita/imunologia , Ativação Linfocitária , Infecções por Protozoários/imunologia , Linfócitos T/imunologia , Viroses/imunologia , Animais , Infecções Bacterianas/patologia , Suscetibilidade a Doenças , Helmintíase/patologia , Humanos , Infecções por Protozoários/patologia , Linfócitos T/patologia , Viroses/patologia
17.
Cell Host Microbe ; 22(5): 717, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-29120747
19.
Cell Host Microbe ; 22(4): 429-431, 2017 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-29024637

RESUMO

The determinants of helminth resistance are not well understood. In this issue of Cell Host & Microbe, Entwistle et al. (2017) provide intriguing evidence that a phospholipase A2 (Pla2gb1) produced by epithelial cells can impair larval development in helminths, providing a novel mechanism contributing to intestinal nematode resistance.


Assuntos
Fosfolipases A2 do Grupo IB
20.
J Immunol ; 198(2): 634-639, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27956529

RESUMO

Concurrent helminth infection potently inhibits T cell immunity; however, whether helminthes prevent T cell priming or skew clonal recruitment and effector differentiation is not known. Using coinfection with two natural mouse pathogens, Heligmosomoides polygyrus and Toxoplasma gondii, to investigate the negative impact of helminthes on the CD8 T cell response, we demonstrate helminth-induced suppression of IL-12-dependent differentiation of killer-like receptor G1+ effector CD8 T cells and IFN-γ production. Nevertheless, reversal of helminth suppression of the innate IL-12 response of CD8α+ dendritic cells, which occurred in STAT6-deficient mice, was not sufficient to normalize CD8 T cell differentiation. Instead, a combined deficiency in IL-4 and IL-10 was required to reverse the negative effects of helminth coinfection on the CD8 T cell response. Monoclonal T. gondii-specific CD8 T cells adoptively transferred into coinfected mice recapitulated the spectrum of helminth-induced effects on the polyclonal CD8 T response, indicating the lack of requirement for clonal skewing.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Coinfecção/imunologia , Helmintíase/imunologia , Ativação Linfocitária/imunologia , Células Th2/imunologia , Transferência Adotiva , Animais , Diferenciação Celular/imunologia , Modelos Animais de Doenças , Interleucina-10/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nematospiroides dubius/imunologia , Toxoplasma/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA