Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
Sci Rep ; 11(1): 20428, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34650101

RESUMEN

Glycosylation is the most common post-translational modification and has myriad of biological functions. However, glycan analysis has always been a challenge. Here, we would like to present new techniques for glycan fingerprinting based on enzymatic fluorescent labeling and gel electrophoresis. The method is illustrated on SARS2 spike (S) glycoproteins. SARS2, a novel coronavirus and the causative agent of the COVID-19 pandemic, has had significant social and economic impacts since the end of 2019. To obtain the N-glycan fingerprint of an S protein, glycans released from the protein are first labeled through enzymatic incorporation of fluorophore-conjugated sialic acid or fucose, then separated by SDS-PAGE, and finally visualized with a fluorescent imager. To identify the labeled glycans of a fingerprint, glycan standards and glycan ladders are enzymatically generated and run alongside the samples as references. By comparing the mobility of a labeled glycan to that of a glycan standard, the identity of glycans maybe determined. O-glycans can also be fingerprinted. Due to the lack of an enzyme for broad O-glycan release, O-glycans on the S protein can be labeled with fluorescent sialic acid and digested with trypsin to obtain labeled glycan peptides that are then separated by gel electrophoresis. Glycan fingerprinting could serve as a quick method for globally assessing the glycosylation of a specific glycoprotein.


Asunto(s)
COVID-19/virología , Polisacáridos/análisis , SARS-CoV-2/química , Glicoproteína de la Espiga del Coronavirus/química , Carbocianinas/química , Electroforesis en Gel de Poliacrilamida , Colorantes Fluorescentes/química , Fucosa/análogos & derivados , Glicosilación , Humanos , Ácido N-Acetilneuramínico/análogos & derivados , Imagen Óptica
3.
Cell Host Microbe ; 29(10): 1531-1544.e9, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34536347

RESUMEN

The minimal genetic requirements for microbes to survive within multiorganism communities, including host-pathogen interactions, remain poorly understood. Here, we combined targeted gene mutagenesis with phenotype-guided genetic reassembly to identify a cooperative network of SPI-2 T3SS effector genes that are sufficient for Salmonella Typhimurium (STm) to cause disease in a natural host organism. Five SPI-2 effector genes support pathogen survival within the host cell cytoplasm by coordinating bacterial replication with Salmonella-containing vacuole (SCV) division. Unexpectedly, this minimal genetic repertoire does not support STm systemic infection of mice. In vivo screening revealed a second effector-gene network, encoded by the spv operon, that expands the life cycle of STm from growth in cells to deep-tissue colonization in a murine model of typhoid fever. Comparison between Salmonella infection models suggests how cooperation between effector genes drives tissue tropism in a pathogen group.


Asunto(s)
Proteínas Bacterianas/genética , Redes Reguladoras de Genes , Infecciones por Salmonella/microbiología , Salmonella typhimurium/genética , Animales , Proteínas Bacterianas/metabolismo , Citoplasma/microbiología , Femenino , Islas Genómicas , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Endogámicos C57BL , Viabilidad Microbiana , Operón , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/patogenicidad , Salmonella typhimurium/fisiología , Tropismo , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo , Virulencia
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.
Immunity ; 43(5): 896-908, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26572061

RESUMEN

Deletion of self-antigen-specific T cells during thymic development provides protection from autoimmunity. However, it is unclear how efficiently this occurs for tissue-restricted self antigens, or how immune tolerance is maintained for self-antigen-specific T cells that routinely escape deletion. Here we show that endogenous CD4+ T cells with specificity for a set of tissue-restricted self antigens were not deleted at all. For pancreatic self antigen, this resulted in an absence of steady-state tolerance, while for the lung and intestine, tolerance was maintained by the enhanced presence of thymically-derived antigen-specific Foxp3+ regulatory T (Treg) cells. Unlike deletional tolerance, Treg cell-mediated tolerance was broken by successive antigen challenges. These findings reveal that for some tissue-restricted self antigens, tolerance relies entirely on nondeletional mechanisms that are less durable than T cell deletion. This might explain why autoimmunity is often tissue-specific, and it offers a rationale for cancer vaccine strategies targeting tissue-restricted tumor antigens.


Asunto(s)
Autoantígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Tolerancia Inmunológica/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoinmunidad/inmunología , Vacunas contra el Cáncer/inmunología , Factores de Transcripción Forkhead/inmunología , Ratones , Ratones Endogámicos C57BL
7.
Chimerism ; 6(1-2): 8-20, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-26517600

RESUMEN

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.


Asunto(s)
Antígenos HLA/inmunología , Tolerancia Inmunológica , Memoria Inmunológica , Sistema del Grupo Sanguíneo Rh-Hr/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos HLA/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Sistema del Grupo Sanguíneo Rh-Hr/historia
8.
Aging Cell ; 14(6): 1122-6, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26248606

RESUMEN

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.


Asunto(s)
Envejecimiento/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Lamina Tipo A/genética , Pulmón/virología , Macrófagos Alveolares/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Progeria/genética , Progeria/inmunología , Linfocitos T Reguladores/inmunología
9.
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
10.
J Clin Invest ; 125(4): 1713-25, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25751061

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Muerte Fetal/prevención & control , Listeriosis/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Receptores CXCR3/fisiología , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Ampicilina/uso terapéutico , Animales , Antibacterianos/uso terapéutico , Quimiocina CXCL9/biosíntesis , Quimiocina CXCL9/genética , Quimiocina CXCL9/fisiología , Quimiocinas/metabolismo , Cruzamientos Genéticos , Decidua/inmunología , Femenino , Muerte Fetal/etiología , Reabsorción del Feto/inmunología , Reabsorción del Feto/prevención & control , Listeriosis/tratamiento farmacológico , Macrófagos/inmunología , Masculino , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Neutrófilos/inmunología , Ovalbúmina/genética , Ovalbúmina/inmunología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Embarazo , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Receptores CXCR3/antagonistas & inhibidores , Receptores CXCR3/biosíntesis , Receptores CXCR3/deficiencia , Receptores CXCR3/genética , Bazo/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T Reguladores/inmunología , Regulación hacia Arriba , Virulencia
11.
Cell Mol Immunol ; 11(6): 595-605, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25242275

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Efectos Tardíos de la Exposición Prenatal/prevención & control , Vacunas Atenuadas , Animales , Susceptibilidad a Enfermedades , Femenino , Histocompatibilidad , Humanos , Isoantígenos/inmunología , Listeriosis/complicaciones , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Atención Preconceptiva , Embarazo/inmunología , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/inmunología , Vacunación , Vacunas Atenuadas/administración & dosificación
12.
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
13.
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
14.
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
15.
Chimerism ; 5(1): 16-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24553046

RESUMEN

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


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Quimera/inmunología , Memoria Inmunológica , Intercambio Materno-Fetal , Linfocitos T Reguladores/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Embrión de Mamíferos , Femenino , Humanos , Embarazo , Linfocitos T Reguladores/inmunología
16.
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
17.
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
18.
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
19.
J Leukoc Biol ; 94(2): 367-76, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23744647

RESUMEN

Although T cell activation has been classically described to require distinct, positive stimulation signals that include B7-1 (CD80) and B7-2 (CD86) costimulation, overriding suppression signals that avert immune-mediated host injury are equally important. How these opposing stimulation and suppression signals work together remains incompletely defined. Our recent studies demonstrate that CD8 Teff activation in response to cognate peptide stimulation is actively suppressed by the Foxp3(+) subset of CD4 cells, called Tregs. Here, we show that the elimination of Treg suppression does not bypass the requirement for positive B7-1/B7-2 costimulation. The expansion, IFN-γ cytokine production, cytolytic, and protective features of antigen-specific CD8 T cells stimulated with purified cognate peptide in Treg-ablated mice were each neutralized effectively by CTLA-4-Ig that blocks B7-1/B7-2. In turn, given the efficiency whereby CTLA-4-Ig overrides the effects of Treg ablation, the role of Foxp3(+) cell-intrinsic CTLA-4 in mitigating CD8 Teff activation was also investigated. With the use of mixed chimera mice that contain CTLA-4-deficient Tregs exclusively after the ablation of WT Foxp3(+) cells, a critical role for Treg CTLA-4 in suppressing the expansion, cytokine production, cytotoxicity, and protective features of peptide-stimulated CD8 T cells is revealed. Thus, the activation of protective CD8 T cells requires positive B7-1/B7-2 costimulation even when suppression by Tregs and in particular, Treg-intrinsic CTLA-4 is circumvented.


Asunto(s)
Antígeno B7-1/antagonistas & inhibidores , Antígeno B7-2/antagonistas & inhibidores , Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Abatacept , Traslado Adoptivo , Animales , Antígeno B7-1/deficiencia , Antígeno B7-1/fisiología , Antígeno B7-2/deficiencia , Antígeno B7-2/fisiología , Citotoxicidad Inmunológica , Factores de Transcripción Forkhead/análisis , Inmunoconjugados/farmacología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Quimera por Radiación
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA