Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Cell ; 162(3): 505-15, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26213383

RESUMEN

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.


Asunto(s)
Feto/inmunología , Aptitud Genética , Tolerancia Inmunológica , Mamíferos/fisiología , Embarazo/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos/inmunología , Quimerismo , Femenino , Humanos , Masculino , Mamíferos/inmunología , Ratones , Placenta/inmunología
2.
Cell ; 153(4): 785-96, 2013 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-23663778

RESUMEN

A naive CD4(+) T cell population specific for a microbial peptide:major histocompatibility complex II ligand (p:MHCII) typically consists of about 100 cells, each with a different T cell receptor (TCR). Following infection, this population produces a consistent ratio of effector cells that activate microbicidal functions of macrophages or help B cells make antibodies. We studied the mechanism that underlies this division of labor by tracking the progeny of single naive T cells. Different naive cells produced distinct ratios of macrophage and B cell helpers but yielded the characteristic ratio when averaged together. The effector cell pattern produced by a given naive cell correlated with the TCR-p:MHCII dwell time or the amount of p:MHCII. Thus, the consistent production of effector cell subsets by a polyclonal population of naive cells results from averaging the diverse behaviors of individual clones, which are instructed in part by the strength of TCR signaling.


Asunto(s)
Infecciones Bacterianas/inmunología , Linfocitos T CD4-Positivos/citología , Diferenciación Celular , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Ratones , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología
3.
Immunity ; 38(6): 1261-70, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23791647

RESUMEN

Thymically derived Foxp3⁺ regulatory T (Treg) cells have a propensity to recognize self-peptide:MHC complexes, but their ability to respond to epitope-defined foreign antigens during infectious challenge has not been demonstrated. Here we show that pulmonary infection with Mycobacterium tuberculosis (Mtb), but not Listeria monocytogenes (Lm), induced robust lymph node expansion of a highly activated population of pathogen-specific Treg cells from the pre-existing pool of thymically derived Treg cells. These antigen-specific Treg cells peaked in numbers 3 weeks after infection but subsequently underwent selective elimination driven, in part, by interleukin-12-induced intrinsic expression of the Th1-cell-promoting transcription factor T-bet. Thus, the initial Mtb-induced inflammatory response promotes pathogen-specific Treg cell proliferation, but these cells are actively culled later, probably to prevent suppression during later stages of infection. These findings have important implications for the prevention and treatment of tuberculosis and other chronic diseases in which antigen-specific Treg cells restrict immunity.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Interleucina-12/inmunología , Mycobacterium tuberculosis/inmunología , Proteínas de Dominio T Box/metabolismo , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Proliferación Celular , Células Cultivadas , Supresión Clonal , Epítopos de Linfocito T/inmunología , Factores de Transcripción Forkhead/genética , Evasión Inmune , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos/inmunología , Proteínas de Dominio T Box/genética , Linfocitos T Reguladores/microbiología , Timo/patología
4.
Glycobiology ; 31(11): 1435-1443, 2021 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-34280262

RESUMEN

Glycosaminoglycans (GAGs), such as hyaluronan (HA) and heparan sulfate (HS), are a large group of polysaccharides found in the extracellular matrix and on the cell surface. The turnover of these molecules is controlled by de novo synthesis and catabolism through specific endoglycosidases, which are the keys to our understanding of the homeostasis of GAGs and could hold opportunities for therapeutic intervention. Herein, we describe assays for endoglycosidases using nonreducing end fluorophore-labeled GAGs, in which GAGs were labeled via incorporation of GlcNAz by specific synthases and cycloaddition of alkyne fluorophores and then digested with corresponding endoglycosidases. Assays of various HA-specific hyaluronidases (HYALs), including PH-20 or SPAM1, and HS-specific heparanase (HPSE) are presented. We demonstrated the distinctive pH profiles, substrate specificities and specific activities of these enzymes and provided evidence that both HYAL3 and HYAL4 are authentic hyaluronidases. In addition, while all HYALs are active on high-molecular-weight HA, they are active on low-molecular-weight HA. Subsequently, we defined a new way of measuring the activities of HYALs. Our results indicate that the activities of HYALs must be under strict pH regulation. Our quantitative methods of measuring the activity GAG endoglycosidases could bring the opportunity of designing novel therapeutics by targeting these important enzymes.


Asunto(s)
Glucuronidasa/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/metabolismo , Imagen Óptica , Electroforesis en Gel de Poliacrilamida , Concentración de Iones de Hidrógeno , Pasteurella multocida/enzimología , Proteínas Recombinantes/metabolismo , Streptococcus agalactiae/enzimología , Especificidad por Sustrato
5.
Glycobiology ; 30(12): 970-980, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-32248235

RESUMEN

Like sialylation, fucose usually locates at the nonreducing ends of various glycans on glycoproteins and constitutes important glycan epitopes. Detecting the substrate glycans of fucosyltransferases is important for understanding how these glycan epitopes are regulated in response to different growth conditions and external stimuli. Here we report the detection of these glycans on glycoproteins as well as in their free forms via enzymatic incorporation of fluorophore-conjugated fucose using FUT2, FUT6, FUT7, FUT8 and FUT9. Specifically, we describe the detection of the substrate glycans of these enzymes on fetal bovine fetuin, recombinant H1N1 viral neuraminidase and therapeutic antibodies. The detected glycans include complex and high-mannose N-glycans. By establishing a series of precursors for the synthesis of Lewis X and sialyl Lewis X structures, we not only provide convenient electrophoresis methods for studying glycosylation but also demonstrate the substrate specificities and some kinetic features of these enzymes. Our results support the notion that fucosyltransferases are key targets for regulating the synthesis of Lewis X and sialyl Lewis X structures.


Asunto(s)
Colorantes Fluorescentes/química , Fucosa/química , Fucosiltransferasas/química , Polisacáridos/análisis , Animales , Bovinos , Electroforesis , Fetuínas/química , Fetuínas/metabolismo , Colorantes Fluorescentes/metabolismo , Fucosa/metabolismo , Fucosiltransferasas/metabolismo , Polisacáridos/metabolismo , Especificidad por Sustrato
6.
Nature ; 496(7443): 106-9, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23535599

RESUMEN

Protein N-myristoylation is a 14-carbon fatty-acid modification that is conserved across eukaryotic species and occurs on nearly 1% of the cellular proteome. The ability of the myristoyl group to facilitate dynamic protein-protein and protein-membrane interactions (known as the myristoyl switch) makes it an essential feature of many signal transduction systems. Thus pathogenic strategies that facilitate protein demyristoylation would markedly alter the signalling landscape of infected host cells. Here we describe an irreversible mechanism of protein demyristoylation catalysed by invasion plasmid antigen J (IpaJ), a previously uncharacterized Shigella flexneri type III effector protein with cysteine protease activity. A yeast genetic screen for IpaJ substrates identified ADP-ribosylation factor (ARF)1p and ARF2p, small molecular mass GTPases that regulate cargo transport through the Golgi apparatus. Mass spectrometry showed that IpaJ cleaved the peptide bond between N-myristoylated glycine-2 and asparagine-3 of human ARF1, thereby providing a new mechanism for host secretory inhibition by a bacterial pathogen. We further demonstrate that IpaJ cleaves an array of N-myristoylated proteins involved in cellular growth, signal transduction, autophagasome maturation and organelle function. Taken together, these findings show a previously unrecognized pathogenic mechanism for the site-specific elimination of N-myristoyl protein modification.


Asunto(s)
Antígenos Bacterianos/metabolismo , Ácido Mirístico/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis , Shigella flexneri/metabolismo , Factores de Virulencia/metabolismo , Factor 1 de Ribosilacion-ADP/química , Factor 1 de Ribosilacion-ADP/metabolismo , Factores de Ribosilacion-ADP/metabolismo , Secuencia de Aminoácidos , Animales , Asparagina/metabolismo , Autofagia , Biocatálisis , Proteasas de Cisteína/metabolismo , Disentería Bacilar , Femenino , Glicina/metabolismo , Aparato de Golgi/metabolismo , Aparato de Golgi/patología , Células HEK293 , Células HeLa , Humanos , Listeria monocytogenes/fisiología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Fagosomas/metabolismo , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Shigella flexneri/enzimología , Transducción de Señal , Especificidad por Sustrato , Virulencia
7.
Nature ; 504(7478): 158-62, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24196717

RESUMEN

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.


Asunto(s)
Antígenos CD/metabolismo , Células Eritroides/inmunología , Infecciones por Escherichia coli/inmunología , Tolerancia Inmunológica/inmunología , Listeriosis/inmunología , Receptores de Transferrina/metabolismo , Animales , Animales Recién Nacidos , Arginasa/genética , Arginasa/metabolismo , Susceptibilidad a Enfermedades/inmunología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Eritroides/enzimología , Escherichia coli/inmunología , Femenino , Sangre Fetal/citología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/genética , Listeria monocytogenes/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo
8.
Nature ; 490(7418): 102-6, 2012 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-23023128

RESUMEN

Pregnancy is an intricately orchestrated process where immune effector cells with fetal specificity are selectively silenced. This requires the sustained expansion of immune-suppressive maternal FOXP3(+) regulatory T cells (T(reg) cells), because even transient partial ablation triggers fetal-specific effector T-cell activation and pregnancy loss. In turn, many idiopathic pregnancy complications proposed to originate from disrupted fetal tolerance are associated with blunted maternal T(reg) expansion. Importantly, however, the antigen specificity and cellular origin of maternal T(reg) cells that accumulate during gestation remain incompletely defined. Here we show that pregnancy selectively stimulates the accumulation of maternal FOXP3(+) CD4 cells with fetal specificity using tetramer-based enrichment that allows the identification of rare endogenous T cells. Interestingly, after delivery, fetal-specific T(reg) cells persist at elevated levels, maintain tolerance to pre-existing fetal antigen, and rapidly re-accumulate during subsequent pregnancy. The accelerated expansion of T(reg) cells during secondary pregnancy was driven almost exclusively by proliferation of fetal-specific FOXP3(+) cells retained from prior pregnancy, whereas induced FOXP3 expression and proliferation of pre-existing FOXP3(+) cells each contribute to T(reg) expansion during primary pregnancy. Furthermore, fetal resorption in secondary compared with primary pregnancy becomes more resilient to partial maternal FOXP3(+) cell ablation. Thus, pregnancy imprints FOXP3(+) CD4 cells that sustain protective regulatory memory to fetal antigen. We anticipate that these findings will spark further investigation on maternal regulatory T-cell specificity that unlocks new strategies for improving pregnancy outcomes and novel approaches for therapeutically exploiting T(reg) cell memory.


Asunto(s)
Antígenos/inmunología , Anergia Clonal/inmunología , Proteínas Fetales/inmunología , Memoria Inmunológica/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Femenino , Feto/inmunología , Factores de Transcripción Forkhead/metabolismo , Memoria Inmunológica/genética , Interferón gamma/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Periodo Posparto/inmunología , Embarazo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
9.
Proc Natl Acad Sci U S A ; 111(29): 10672-7, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-25002484

RESUMEN

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.


Asunto(s)
Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Linfocitos T CD4-Positivos/inmunología , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Inflamación/inmunología , Interleucina-17/biosíntesis , Intestinos/patología , Ligando OX40/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Antígeno CTLA-4/metabolismo , Candida albicans/fisiología , Diferenciación Celular/inmunología , Proliferación Celular , Humanos , Inflamación/microbiología , Inflamación/patología , Intestinos/inmunología , Intestinos/microbiología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Fenotipo , Linfocitos T Reguladores/inmunología , Células Th17/inmunología
10.
J Immunol ; 192(7): 2970-4, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24591368

RESUMEN

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.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Feto/inmunología , Factores de Transcripción Forkhead/inmunología , Células TH1/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Diferenciación Celular/inmunología , Femenino , Feto/metabolismo , Citometría de Flujo , Factores de Transcripción Forkhead/metabolismo , Interacciones Huésped-Patógeno/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Listeria monocytogenes/inmunología , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Listeriosis/microbiología , Ratones , Ratones Congénicos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Embarazo , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células TH1/metabolismo
11.
J Immunol ; 192(11): 4949-56, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24837152

RESUMEN

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.


Asunto(s)
Antígenos/inmunología , Tolerancia Inmunológica , Complicaciones del Embarazo/inmunología , Linfocitos T Reguladores/inmunología , Animales , Femenino , Humanos , Embarazo , Complicaciones del Embarazo/patología , Linfocitos T Reguladores/patología
12.
PLoS Pathog ; 8(8): e1002873, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22916020

RESUMEN

Although the intracellular bacterium Listeria monocytogenes has an established predilection for disseminated infection during pregnancy that often results in spontaneous abortion or stillbirth, the specific host-pathogen interaction that dictates these disastrous complications remain incompletely defined. Herein, we demonstrate systemic maternal Listeria infection during pregnancy fractures fetal tolerance and triggers fetal wastage in a dose-dependent fashion. Listeria was recovered from the majority of concepti after high-dose infection illustrating the potential for in utero invasion. Interestingly with reduced inocula, fetal wastage occurred without direct placental or fetal invasion, and instead paralleled reductions in maternal Foxp3(+) regulatory T cell suppressive potency with reciprocal expansion and activation of maternal fetal-specific effector T cells. Using mutants lacking virulence determinants required for in utero invasion, we establish Listeria cytoplasmic entry is essential for disrupting fetal tolerance that triggers maternal T cell-mediated fetal resorption. Thus, infection-induced reductions in maternal Foxp3(+) regulatory T cell suppression with ensuing disruptions in fetal tolerance play critical roles in pathogenesis of immune-mediated fetal wastage.


Asunto(s)
Reabsorción del Feto/inmunología , Factores de Transcripción Forkhead , Tolerancia Inmunológica , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Linfocitos T Reguladores/inmunología , Animales , Citoplasma/inmunología , Citoplasma/microbiología , Femenino , Reabsorción del Feto/genética , Reabsorción del Feto/microbiología , Reabsorción del Feto/patología , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Listeriosis/genética , Listeriosis/patología , Ratones , Ratones Endogámicos BALB C , Embarazo , Complicaciones Infecciosas del Embarazo/patología , Linfocitos T Reguladores/patología
13.
J Immunol ; 189(2): 876-84, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22711893

RESUMEN

Although best characterized for sustaining T cell exhaustion during persistent viral infection, programmed death ligand-1 (PDL-1) also stimulates the expansion of protective T cells after infection with intracellular bacterial pathogens. Therefore, establishing the molecular signals that control whether PDL-1 stimulates immune suppression or activation is important as immune modulation therapies based on manipulating PDL-1 are being developed. In this study, the requirement for PDL-1 blockade initiated before infection with the intracellular bacterium Listeria monocytogenes in reducing pathogen-specific T cell expansion is demonstrated. In turn, the role of proinflammatory cytokines triggered early after L. monocytogenes infection in controlling PDL-1-mediated T cell stimulation was investigated using mice with targeted defects in specific cytokines or cytokine receptors. These experiments illustrate an essential role for IL-12 or type I IFNs in PDL-1-mediated expansion of pathogen-specific CD8(+) T cells. Unexpectedly, direct stimulation by neither IL-12 nor type I IFNs on pathogen-specific CD8(+) cells was essential for PDL-1-mediated expansion. Instead, the absence of early innate IFN-γ production in mice with combined defects in both IL-12 and type I IFNR negated the impacts of PDL-1 blockade. In turn, IFN-γ ablation using neutralizing Abs or in mice with targeted defects in IFN-γR each eliminated the PDL-1-mediated stimulatory impacts on pathogen-specific T cell expansion. Thus, innate IFN-γ is essential for PDL-1-mediated T cell stimulation.


Asunto(s)
Antígeno B7-H1/fisiología , Inmunidad Innata , Interferón gamma/fisiología , Listeriosis/inmunología , Activación de Linfocitos/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/microbiología , Animales , Anticuerpos Neutralizantes/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Linfocitos T CD8-positivos/patología , Contraindicaciones , Inmunidad Innata/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-12/deficiencia , Interleucina-12/fisiología , Listeriosis/genética , Listeriosis/patología , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Interferón/deficiencia , Receptores de Interferón/metabolismo , Receptores de Interferón/fisiología , Subgrupos de Linfocitos T/patología , Receptor de Interferón gamma
14.
Proc Natl Acad Sci U S A ; 108(33): 13694-9, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21813761

RESUMEN

Immunity against new infections declines in the last quartile of life, as do numbers of naive T cells. Peripheral maintenance of naive T cells over the lifespan is necessary because their production drastically declines by puberty, a result of thymic involution. We report that this maintenance is not random in advanced aging. As numbers and diversity of naive CD8(+) T cells declined with aging, surviving cells underwent faster rates of homeostatic proliferation, were selected for high T-cell receptor:pMHC avidity, and preferentially acquired "memory-like" phenotype. These high-avidity precursors preferentially responded to infection and exhibited strong antimicrobial function. Thus, T-cell receptor avidity for self-pMHC provides a proofreading mechanism to maintain some of the fittest T cells in the otherwise crumbling naive repertoire, providing a degree of compensation for numerical and diversity defects in old T cells.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD8-positivos/citología , Antígenos HLA/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Senescencia Celular/inmunología , Homeostasis/inmunología , Inmunidad/inmunología , Memoria Inmunológica , Recuento de Linfocitos , Ratones
15.
Reproduction ; 146(6): R191-203, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23929902

RESUMEN

Pregnancy in placental mammals offers exceptional comprehensive benefits of in utero protection, nutrition, and metabolic waste elimination for the developing fetus. However, these benefits also require durable strategies to mitigate maternal rejection of fetal tissues expressing foreign paternal antigens. Since the initial postulate of expanded maternal immune tolerance by Sir Peter Medawar 60 years ago, an amazingly elaborate assortment of molecular and cellular modifications acting both locally at the maternal-placental interface and systemically have been shown to silence potentially detrimental maternal immune responses. In turn, simultaneously maintaining host defense against the infinite array of potential pathogens during pregnancy is equally important. Fortunately, resistance against most infections is preserved seamlessly throughout gestation. On the other hand, recent studies on pathogens with unique predisposition for prenatal infections have uncovered distinctive holes in host defense associated with the reproductive process. Using these infections to probe the response during pregnancy, the immune suppressive regulatory subset of maternal CD4 T cells has been increasingly shown to dictate the inter-workings between prenatal infection susceptibility and pathogenesis of ensuing pregnancy complications. Herein, the recent literature suggesting a necessity for maternal regulatory T cells (Tregs) in pregnancy-induced immunological shifts that sustain fetal tolerance is reviewed. Additional discussion is focused on how expansion of maternal Treg suppression may become exploited by pathogens that cause prenatal infections and the perilous potential of infection-induced immune activation that may mitigate fetal tolerance and inadvertently inject hostility into the protective in utero environment.


Asunto(s)
Complicaciones Infecciosas del Embarazo/inmunología , Embarazo/inmunología , Linfocitos T Reguladores/fisiología , Femenino , Enfermedades Fetales/inmunología , Feto/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Inmunidad Innata/fisiología
16.
J Immunol ; 187(5): 2569-77, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21810602

RESUMEN

T cell activation is controlled by incompletely defined opposing stimulation and suppression signals that together sustain the balance between optimal host defense against infection and peripheral tolerance. In this article, we explore the impacts of Foxp3(+) regulatory T cell (Treg) suppression in priming Ag-specific T cell activation under conditions of noninfection and infection. We find the transient ablation of Foxp3(+) Tregs unleashes the robust expansion and activation of peptide-stimulated CD8(+) T cells that provide protection against Listeria monocytogenes infection in an Ag-specific fashion. By contrast, Treg ablation had nonsignificant impacts on the CD8(+) T cell response primed by infection with recombinant L. monocytogenes. Similarly, nonrecombinant L. monocytogenes administered with peptide stimulated the expansion and activation of CD8(+) T cells that paralleled the response primed by Treg ablation. Interestingly, these adjuvant properties of L. monocytogenes did not require CD8(+) T cell stimulation by IL-12 produced in response to infection, but instead were associated with sharp reductions in Foxp3(+) Treg suppressive potency. Therefore, Foxp3(+) Tregs impose critical barriers that, when overcome naturally during infection or artificially with ablation, allow the priming of protective Ag-specific CD8(+) T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Comunicación Celular/inmunología , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Ratones , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo
17.
Immunology ; 136(1): 1-10, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22211994

RESUMEN

The immune system is intricately regulated allowing potent effectors to expand and become rapidly mobilized after infection, while simultaneously silencing potentially detrimental responses that averts immune-mediated damage to host tissues. This relies in large part on the delicate interplay between immune suppressive regulatory CD4(+) T (Treg) cells and immune effectors that without active suppression by Treg cells cause systemic and organ-specific autoimmunity. Although these beneficial roles have been classically described as counterbalanced by impaired host defence against infection, newfound protective roles for Treg cells against specific viral pathogens (e.g. herpes simplex virus 2, lymphocytic choriomeningitis virus, West Nile virus) have been uncovered using transgenic mice that allow in vivo Treg-cell ablation based on Foxp3 expression. In turn, Foxp3(+) Treg cells also provide protection against some parasitic (Plasmodium sp., Toxoplasma gondii) and fungal (Candida albicans) pathogens. By contrast, for bacterial and mycobacterial infections (e.g. Listeria monocytogenes, Salmonella enterica, Mycobacterium tuberculosis), experimental manipulation of Foxp3(+) cells continues to indicate detrimental roles for Treg cells in host defence. This variance is probably related to functional plasticity in Treg cell suppression that shifts discordantly following infection with different types of pathogens. Furthermore, the efficiency whereby Treg cells silence immune activation coupled with the plasticity in Foxp3(+) cell activity suggest that overriding Treg-mediated suppression represents a prerequisite 'signal zero' that together with other stimulation signals [T-cell receptor (signal 1), co-stimulation (signal 2), inflammatory cytokines (signal 3)] are essential for T-cell activation in vivo. Herein, the importance of Foxp3(+) Treg cells in host defence against infection, and the significance of infection-induced shifts in Treg-cell suppression are summarized.


Asunto(s)
Infecciones Bacterianas/inmunología , Linfocitos T Reguladores/inmunología , Virosis/inmunología , Animales , Factores de Transcripción Forkhead/inmunología , Humanos , Tolerancia Inmunológica
18.
PLoS Pathog ; 6(8): e1001043, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20714351

RESUMEN

The pathogenesis of persistent infection is dictated by the balance between opposing immune activation and suppression signals. Herein, virulent Salmonella was used to explore the role and potential importance of Foxp3-expressing regulatory T cells in dictating the natural progression of persistent bacterial infection. Two distinct phases of persistent Salmonella infection are identified. In the first 3-4 weeks after infection, progressively increasing bacterial burden was associated with delayed effector T cell activation. Reciprocally, at later time points after infection, reductions in bacterial burden were associated with robust effector T cell activation. Using Foxp3(GFP) reporter mice for ex vivo isolation of regulatory T cells, we demonstrate that the dichotomy in infection tempo between early and late time points is directly paralleled by drastic changes in Foxp3(+) Treg suppressive potency. In complementary experiments using Foxp3(DTR) mice, the significance of these shifts in Treg suppressive potency on infection outcome was verified by enumerating the relative impacts of regulatory T cell ablation on bacterial burden and effector T cell activation at early and late time points during persistent Salmonella infection. Moreover, Treg expression of CTLA-4 directly paralleled changes in suppressive potency, and the relative effects of Treg ablation could be largely recapitulated by CTLA-4 in vivo blockade. Together, these results demonstrate that dynamic regulation of Treg suppressive potency dictates the course of persistent bacterial infection.


Asunto(s)
Tolerancia Inmunológica/inmunología , Infecciones por Salmonella/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD/inmunología , Antígeno CTLA-4 , Femenino , Factores de Transcripción Forkhead/inmunología , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
19.
J Immunol ; 184(2): 869-76, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20026741

RESUMEN

T cell activation required for host defense against infection is an intricately regulated and precisely controlled process. Although in vitro studies indicate that three distinct stimulatory signals are required for T cell activation, the precise contribution of each signal in regulating T cell proliferation and differentiation after in vivo infection is unknown. In this study, altered peptide ligands (APLs) derived from the protective Salmonella-specific FliC Ag and CD4+ T cells specific for the immune-dominant FliC(431-439) peptide within this Ag were used to determine how changes in TCR stimulation impact CD4+ T cell proliferation, differentiation, and protective potency. To explore the prevalence and potential use of altered TCR stimulation by bacterial pathogens, naturally occurring APLs containing single amino acid substitutions in putative TCR contact residues within the FliC(431-439) peptide were identified and used for stimulation under both noninfection and infection conditions. On the basis of this analysis, naturally-occurring APLs that prime proliferation of FliC-specific CD4+ T cells either more potently or less potently compared with the wild-type FliC(431-439) peptide were identified. Remarkably, despite these differences in proliferation, all of the APLs primed reduced IFN-gamma production by FliC(431-439)-specific CD4+ T cells after stimulation in vivo. Moreover, after expression of the parental FliC(431-439) peptide or each APL in recombinant Listeria monocytogenes, only CD4+ T cells stimulated with the wild-type FliC(431-439) peptide conferred significant protection against challenge with virulent Salmonella. These results reveal important and unanticipated roles for TCR stimulation in controlling pathogen-specific CD4+ T cell proliferation, differentiation, and protective potency.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Interferón gamma/biosíntesis , Péptidos/inmunología , Salmonella/inmunología , Animales , Antígenos Bacterianos , Linfocitos T CD4-Positivos/microbiología , Diferenciación Celular/inmunología , Ligandos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Infecciones por Salmonella/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T
20.
Commun Biol ; 5(1): 501, 2022 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-35614314

RESUMEN

Glycan synthesis and degradation are not template but enzyme only driven processes. Substrate specificities of glyco-enzymes determine the structures of specific natural glycans. Using endoglycosidases as examples, we describe methods to study these enzymes. Endoglycosidase S/S2 specifically deglycosylates the conserved N-glycans of human immunoglobulin G. Endo-ß-Galactosidase hydrolyzes internal ß-galactosyl linkage in polylactosaminoglycan structures. To assay these enzymes, eleven fluorophore-labeled N-glycans and one polylactosamine ladder are synthesized. Digestion of these glycans result in mobility shift in gel electrophoresis. Results on Endo S/S2 assays reveal that they are most active on the agalactosylated biantennary N-glycans with decreased activity on galactosylated and sialylated glycans and little or no activity on branched and bisected glycans. Assays on Endo-ß-Gal reveal that the enzyme is active from pH 3.5 to 9.0 and the ß3-linked GlcNAc adjacent to the cleavage site is minimal for the enzyme recognition with the optimal recognition motif spanning at least four lactosamine repeats. Our methods will provide an opportunity to understand how specific glycans are synthesized and degraded.


Asunto(s)
Glicósido Hidrolasas , Polisacáridos , Glicósido Hidrolasas/metabolismo , Humanos , Inmunoglobulina G , Polisacáridos/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA