Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunohorizons ; 7(11): 771-787, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-38015461

RESUMEN

CD8+ T cell dysfunction contributes to severe respiratory viral infection outcomes in older adults. CD8+ T cells are the primary cell type responsible for viral clearance. With increasing age, CD8+ T cell function declines in conjunction with an accumulation of cytotoxic tissue-resident memory (TRM) CD8+ T cells. We sought to elucidate the role of PD-1 signaling on aged CD8+ T cell function and accumulation of CD8+ TRM cells during acute viral respiratory tract infection, given the importance of PD-1 regulating CD8+ T cells during acute and chronic infections. PD-1 blockade or genetic ablation in aged mice yielded improved CD8+ T cell granzyme B production comparable to that in young mice during human metapneumovirus and influenza viral infections. Syngeneic transplant and adoptive transfer strategies revealed that improved granzyme B production in aged Pdcd1-/- CD8+ T cells was primarily cell intrinsic because aged wild-type CD8+ T cells did not have increased granzyme B production when transplanted into a young host. PD-1 signaling promoted accumulation of cytotoxic CD8+ TRM cells in aged mice. PD-1 blockade of aged mice during rechallenge infection resulted in improved clinical outcomes that paralleled reduced accumulation of CD8+ TRM cells. These findings suggest that PD-1 signaling impaired CD8+ T cell granzyme B production and contributed to CD8+ TRM cell accumulation in the aged lung. These findings have implications for future research investigating PD-1 checkpoint inhibitors as a potential therapeutic option for elderly patients with severe respiratory viral infections.


Asunto(s)
Infecciones del Sistema Respiratorio , Virosis , Animales , Humanos , Ratones , Linfocitos T CD8-positivos , Granzimas , Inhibidores de Puntos de Control Inmunológico , Receptor de Muerte Celular Programada 1
2.
Cell Rep ; 42(11): 113323, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37889750

RESUMEN

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


Asunto(s)
Linfocitos T CD4-Positivos , Microbiota , Ratones , Animales , Interleucina-10/metabolismo , Linfocitos T Reguladores , Factores de Transcripción/metabolismo , Inflamación/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo
3.
Science ; 381(6664): 1324-1330, 2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37733857

RESUMEN

Pregnancy confers partner-specific protection against complications in future pregnancy that parallel persistence of fetal microchimeric cells (FMcs) in mothers after parturition. We show that preexisting FMcs become displaced by new FMcs during pregnancy and that FMc tonic stimulation is essential for expansion of protective fetal-specific forkhead box P3 (FOXP3)-positive regulatory T cells (Treg cells). Maternal microchimeric cells and accumulation of Treg cells with noninherited maternal antigen (NIMA) specificity are similarly overturned in daughters after pregnancy, highlighting a fixed microchimeric cell niche. Whereas NIMA-specific tolerance is functionally erased by pregnancy, partner-specific resiliency against pregnancy complications persists in mothers despite paternity changes in intervening pregnancy. Persistent fetal tolerance reflects FOXP3 expression plasticity, which allows mothers to more durably remember their babies, whereas daughters forget their mothers with new pregnancy-imprinted immunological memories.


Asunto(s)
Quimerismo , Feto , Tolerancia Inmunológica , Memoria Inmunológica , Intercambio Materno-Fetal , Embarazo , Animales , Femenino , Ratones , Embarazo/inmunología , Antígenos/inmunología , Plasticidad de la Célula , Feto/citología , Feto/inmunología , Factores de Transcripción Forkhead/inmunología , Intercambio Materno-Fetal/inmunología , Ratones Endogámicos C57BL , Linfocitos T Reguladores/inmunología
4.
Immun Ageing ; 20(1): 40, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37528458

RESUMEN

BACKGROUND: Lower respiratory infections are a leading cause of severe morbidity and mortality among older adults. Despite ubiquitous exposure to common respiratory pathogens throughout life and near universal seropositivity, antibodies fail to effectively protect the elderly. Therefore, we hypothesized that severe respiratory illness in the elderly is due to deficient CD8+ T cell responses. RESULTS: Here, we establish an aged mouse model of human metapneumovirus infection (HMPV) wherein aged C57BL/6 mice exhibit worsened weight loss, clinical disease, lung pathology and delayed viral clearance compared to young adult mice. Aged mice generate fewer lung-infiltrating HMPV epitope-specific CD8+ T cells. Those that do expand demonstrate higher expression of PD-1 and other inhibitory receptors and are functionally impaired. Transplant of aged T cells into young mice and vice versa, as well as adoptive transfer of young versus aged CD8+ T cells into Rag1-/- recipients, recapitulates the HMPV aged phenotype, suggesting a cell-intrinsic age-associated defect. HMPV-specific aged CD8+ T cells exhibit a terminally exhausted TCF1/7- TOX+ EOMES+ phenotype. We confirmed similar terminal exhaustion of aged CD8+ T cells during influenza viral infection. CONCLUSIONS: This study identifies terminal CD8+ T cell exhaustion as a mechanism of severe disease from respiratory viral infections in the elderly.

5.
Nature ; 606(7915): 769-775, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35676476

RESUMEN

Adaptive immune components are thought to exert non-overlapping roles in antimicrobial host defence, with antibodies targeting pathogens in the extracellular environment and T cells eliminating infection inside cells1,2. Reliance on antibodies for vertically transferred immunity from mothers to babies may explain neonatal susceptibility to intracellular infections3,4. Here we show that pregnancy-induced post-translational antibody modification enables protection against the prototypical intracellular pathogen Listeria monocytogenes. Infection susceptibility was reversed in neonatal mice born to preconceptually primed mothers possessing L. monocytogenes-specific IgG or after passive transfer of antibodies from primed pregnant, but not virgin, mice. Although maternal B cells were essential for producing IgGs that mediate vertically transferred protection, they were dispensable for antibody acquisition of protective function, which instead required sialic acid acetyl esterase5 to deacetylate terminal sialic acid residues on IgG variable-region N-linked glycans. Deacetylated L. monocytogenes-specific IgG protected neonates through the sialic acid receptor CD226,7, which suppressed IL-10 production by B cells leading to antibody-mediated protection. Consideration of the maternal-fetal dyad as a joined immunological unit reveals protective roles for antibodies against intracellular infection and fine-tuned adaptations to enhance host defence during pregnancy and early life.


Asunto(s)
Inmunidad Materno-Adquirida , Inmunoglobulina G , Espacio Intracelular , Listeria monocytogenes , Madres , Embarazo , Acetilesterasa , Animales , Animales Recién Nacidos , Linfocitos B , Femenino , Inmunidad Materno-Adquirida/inmunología , Inmunoglobulina G/inmunología , Interleucina-10/biosíntesis , Espacio Intracelular/inmunología , Espacio Intracelular/microbiología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/prevención & control , Ratones , Ácido N-Acetilneuramínico/metabolismo , Embarazo/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico , Linfocitos T
6.
Sci Rep ; 11(1): 20769, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34675220

RESUMEN

Human metapneumovirus (HMPV) is a leading cause of acute lower respiratory tract illness in children and adults. Repeated infections are common and can be severe in young, elderly, and immunocompromised persons due to short-lived protective humoral immunity. In turn, few protective T cell epitopes have been identified in humans. Thus, we infected transgenic mice expressing the common human HLA MHC-I allele B*07:02 (HLA-B7) with HMPV and screened a robust library of overlapping and computationally predicted HLA-B7 binding peptides. Six HLA-B7-restricted CD8+ T cell epitopes were identified using ELISPOT screening in the F, M, and N proteins, with M195-203 (M195) eliciting the strongest responses. MHC-tetramer flow cytometric staining confirmed HLA-B7 epitope-specific CD8+ T cells migrated to lungs and spleen of HMPV-immune mice. Immunization with pooled HLA-B7-restricted peptides reduced viral titer and protected mice from virulent infection. Finally, we confirmed that CD8+ T cells from HLA-B7 positive humans also recognize the identified epitopes. These results enable identification of HMPV-specific CD8+ T cells in humans and help to inform future HMPV vaccine design.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Antígeno HLA-B7/inmunología , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/inmunología , Animales , Células Cultivadas , Epítopos de Linfocito T/uso terapéutico , Humanos , Interferón gamma/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Paramyxoviridae/prevención & control , Péptidos/inmunología , Péptidos/uso terapéutico , Vacunas Virales/inmunología , Vacunas Virales/uso terapéutico
7.
Transpl Infect Dis ; 23(4): e13655, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34057792

RESUMEN

Tacrolimus is widely used to prevent graft rejection after allogeneic transplantation by suppressing T cells in a non-antigen-specific fashion. Global T-cell suppression makes transplant recipients more susceptible to infection, especially infection by opportunistic intracellular pathogens. Infection followed by secondary challenge with the opportunistic intracellular bacterial pathogen, Listeria monocytogenes, was used to probe when tacrolimus most significantly impacts antimicrobial host defense. Tacrolimus-treated mice showed no difference in innate susceptibility following primary infection, whereas susceptibility to secondary challenge was significantly increased. Modifying the timing of tacrolimus initiation with respect to primary infection compared with secondary challenge showed significantly reduced susceptibility in tacrolimus-treated mice where tacrolimus was discontinued prior to secondary challenge. Thus, tacrolimus overrides protection against secondary infection primed by primary infection (and presumably live attenuated vaccines), with the most critical window for tacrolimus-induced infection susceptibility being exposure immediately prior to secondary challenge. These results have important implications for strategies designed to boost antimicrobial T-cell-mediated immunity in transplant recipients.


Asunto(s)
Listeria monocytogenes , Listeriosis , Animales , Humanos , Inmunidad Celular , Ratones , Ratones Endogámicos C57BL , Tacrolimus/farmacología
8.
mBio ; 12(1)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33622714

RESUMEN

Maternal sepsis is a leading cause of morbidity and mortality during pregnancy. Escherichia coli is a primary cause of bacteremia in women and occurs more frequently during pregnancy. Several key outstanding questions remain regarding how to identify women at highest infection risk and how to boost immunity against E. coli infection during pregnancy. Here, we show that pregnancy-induced susceptibility to E. coli systemic infection extends to rodents as a model of human infection. Mice infected during pregnancy contain >100-fold-more recoverable bacteria in target tissues than nonpregnant controls. Infection leads to near complete fetal wastage that parallels placental plus congenital fetal invasion. Susceptibility in maternal tissues positively correlates with the number of concepti, suggesting important contributions by expanded placental-fetal target tissue. Remarkably, these pregnancy-induced susceptibility phenotypes are also efficiently overturned in mice with resolved sublethal infection prior to pregnancy. Preconceptual infection primes the accumulation of E. coli-specific IgG and IgM antibodies, and adoptive transfer of serum containing these antibodies to naive recipient mice protects against fetal wastage. Together, these results suggest that the lack of E. coli immunity may help discriminate individuals at risk during pregnancy, and that overriding susceptibility to E. coli prenatal infection by preconceptual priming is a potential strategy for boosting immunity in this physiological window of vulnerability.IMPORTANCE Pregnancy makes women especially vulnerable to infection. The most common cause of bloodstream infection during pregnancy is by a bacterium called Escherichia coli This bacterium is a very common cause of bloodstream infection, not just during pregnancy but in all individuals, from newborn babies to the elderly, probably because it is always present in our intestine and can intermittently invade through this mucosal barrier. We first show that pregnancy in animals also makes them more susceptible to E. coli bloodstream infection. This is important because many of the dominant factors likely to control differences in human infection susceptibility can be property controlled for only in animals. Despite this vulnerability induced by pregnancy, we also show that animals with resolved E. coli infection are protected against reinfection during pregnancy, including having resistance to most infection-induced pregnancy complications. Protection against reinfection is mediated by antibodies that can be measured in the blood. This information may help to explain why most women do not develop E. coli infection during pregnancy, enabling new approaches for identifying those at especially high risk of infection and strategies for preventing infection during pregnancy.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Sepsis/inmunología , Sepsis/microbiología , Traslado Adoptivo , Animales , Anticuerpos Antibacterianos/administración & dosificación , Infecciones por Escherichia coli/etiología , Infecciones por Escherichia coli/prevención & control , Femenino , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Placenta , Embarazo , Complicaciones Infecciosas del Embarazo/microbiología , Factores de Riesgo , Sepsis/mortalidad , Sepsis/prevención & control
9.
J Immunol ; 205(2): 447-453, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32522837

RESUMEN

Vaccines against Zika virus (ZIKV) infection that target CD8+ T cells are of considerable interest because Abs may enhance infection susceptibility. However, whether CD8+ T cells are protective or promote susceptibility to clinical infection symptoms remains uncertain. To more precisely investigate ZIKV-specific CD8+ T cells in isolation, we engineered a Listeria monocytogenes-based vector to express a single MHC class I-restricted immune dominant peptide, E294-302, from ZIKV envelope protein. We show accumulation of activated ZIKV-specific CD8+ T cells primed by recombinant L. monocytogenes is associated with reductions in circulating virus levels after ZIKV challenge in type I IFN receptor-deficient mice and wildtype mice administered neutralizing Abs against type I IFN receptor. Interestingly, susceptibility to ZIKV clinical infection including weight loss and mortality each persists and is neither significantly improved nor worsened compared with isogenic L. monocytogenes-primed control mice. These data demonstrating persistent ZIKV clinical susceptibility despite reduced viral burden in mice with expanded virus-specific CD8+ T cells highlights the need for targeting other adaptive immune components in developing vaccines against ZIKV infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Receptor de Interferón alfa y beta/metabolismo , Infección por el Virus Zika/inmunología , Virus Zika/fisiología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Células Cultivadas , Resistencia a la Enfermedad , Susceptibilidad a Enfermedades , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/inmunología , Receptor de Interferón alfa y beta/genética , Proteínas del Envoltorio Viral/inmunología , Carga Viral
10.
Water Res ; 114: 338-350, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28279879

RESUMEN

Intermittent drinking water supply is common in low- and middle-income countries throughout the world and can cause water quality to degrade in the distribution system. In this study, we characterized water quality in one study zone with continuous supply and three zones with intermittent supply in the drinking water distribution network in Arraiján, Panama. Low or zero pressures occurred in all zones, and negative pressures occurred in the continuous zone and two of the intermittent zones. Despite hydraulic conditions that created risks for backflow and contaminant intrusion, only four of 423 (0.9%) grab samples collected at random times were positive for total coliform bacteria and only one was positive for E. coli. Only nine of 496 (1.8%) samples had turbidity >1.0 NTU and all samples had ≥0.2 mg/L free chlorine residual. In contrast, water quality was often degraded during the first-flush period (when supply first returned after an outage). Still, routine and first-flush water quality under intermittent supply was much better in Arraiján than that reported in a previous study conducted in India. Better water quality in Arraiján could be due to better water quality leaving the treatment plant, shorter supply outages, higher supply pressures, a more consistent and higher chlorine residual, and fewer contaminant sources near pipes. The results illustrate that intermittent supply and its effects on water quality can vary greatly between and within distribution networks. The study also demonstrated that monitoring techniques designed specifically for intermittent supply, such as continuous pressure monitoring and sampling the first flush, can detect water quality threats and degradation that would not likely be detected with conventional monitoring.


Asunto(s)
Escherichia coli , Calidad del Agua , Cloro , Agua Potable/microbiología , Microbiología del Agua , Abastecimiento de Agua
11.
J Immunol ; 197(1): 233-43, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27259857

RESUMEN

Viruses are frequent causes of lower respiratory infection (LRI). Programmed cell death-1 (PD-1) signaling contributes to pulmonary CD8(+) T cell (TCD8) functional impairment during acute viral LRI, but the role of TCD8 impairment in viral clearance and immunopathology is unclear. We now find that human metapneumovirus infection induces virus-specific lung TCD8 that fail to produce effector cytokines or degranulate late postinfection, with minimally increased function even in the absence of PD-1 signaling. Impaired lung TCD8 upregulated multiple inhibitory receptors, including PD-1, lymphocyte activation gene 3 (LAG-3), T cell Ig mucin 3, and 2B4. Moreover, coexpression of these receptors continued to increase even after viral clearance, with most virus-specific lung TCD8 expressing three or more inhibitory receptors on day 14 postinfection. Viral infection also increased expression of inhibitory ligands by both airway epithelial cells and APCs, further establishing an inhibitory environment. In vitro Ab blockade revealed that multiple inhibitory receptors contribute to TCD8 impairment induced by either human metapneumovirus or influenza virus infection. In vivo blockade of T cell Ig mucin 3 signaling failed to enhance TCD8 function or reduce viral titers. However, blockade of LAG-3 in PD-1-deficient mice restored TCD8 effector functions but increased lung pathology, indicating that LAG-3 mediates lung TCD8 impairment in vivo and contributes to protection from immunopathology during viral clearance. These results demonstrate that an orchestrated network of pathways modifies lung TCD8 functionality during viral LRI, with PD-1 and LAG-3 serving prominent roles. Lung TCD8 impairment may prevent immunopathology but also contributes to recurrent lung infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Pulmón/inmunología , Metapneumovirus/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Paramyxoviridae/inmunología , Infecciones del Sistema Respiratorio/inmunología , Animales , Antígenos CD/metabolismo , Linfocitos T CD8-positivos/virología , Células Cultivadas , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mucina 3/metabolismo , Receptor de Muerte Celular Programada 1/genética , Infecciones del Sistema Respiratorio/virología , Transducción de Señal , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Proteína del Gen 3 de Activación de Linfocitos
12.
J Immunol ; 195(9): 4319-30, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26401005

RESUMEN

Acute viral infections typically generate functional effector CD8(+) T cells (TCD8) that aid in pathogen clearance. However, during acute viral lower respiratory infection, lung TCD8 are functionally impaired and do not optimally control viral replication. T cells also become unresponsive to Ag during chronic infections and cancer via signaling by inhibitory receptors such as programmed cell death-1 (PD-1). PD-1 also contributes to TCD8 impairment during viral lower respiratory infection, but how it regulates TCD8 impairment and the connection between this state and T cell exhaustion during chronic infections are unknown. In this study, we show that PD-1 operates in a cell-intrinsic manner to impair lung TCD8. In light of this, we compared global gene expression profiles of impaired epitope-specific lung TCD8 to functional spleen TCD8 in the same human metapneumovirus-infected mice. These two populations differentially regulate hundreds of genes, including the upregulation of numerous inhibitory receptors by lung TCD8. We then compared the gene expression of TCD8 during human metapneumovirus infection to those in acute or chronic lymphocytic choriomeningitis virus infection. We find that the immunophenotype of lung TCD8 more closely resembles T cell exhaustion late into chronic infection than do functional effector T cells arising early in acute infection. Finally, we demonstrate that trafficking to the infected lung alone is insufficient for TCD8 impairment or inhibitory receptor upregulation, but that viral Ag-induced TCR signaling is also required. Our results indicate that viral Ag in infected lungs rapidly induces an exhaustion-like state in lung TCD8 characterized by progressive functional impairment and upregulation of numerous inhibitory receptors.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/inmunología , Infecciones del Sistema Respiratorio/inmunología , Enfermedad Aguda , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Análisis por Conglomerados , Perfilación de la Expresión Génica/métodos , Interacciones Huésped-Patógeno/inmunología , Humanos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Metapneumovirus/fisiología , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Fenotipo , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/virología , Bazo/inmunología , Bazo/metabolismo , Bazo/virología , Transcriptoma/genética , Transcriptoma/inmunología
13.
J Virol ; 89(8): 4405-20, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25653440

RESUMEN

UNLABELLED: Type I IFN signaling, which is initiated through activation of the alpha interferon receptor (IFNAR), regulates the expression of proteins that are crucial contributors to immune responses. Paramyxoviruses, including human metapneumovirus (HMPV), have evolved mechanisms to inhibit IFNAR signaling, but the specific contribution of IFNAR signaling to the control of HMPV replication, pathogenesis, and adaptive immunity is unknown. We used IFNAR-deficient (IFNAR(-/-)) mice to assess the effect of IFNAR signaling on HMPV replication and the CD8(+) T cell response. HMPV-infected IFNAR(-/-) mice had a higher peak of early viral replication but cleared the virus with kinetics similar to those of wild-type (WT) mice. However, IFNAR(-/-) mice infected with HMPV displayed less airway dysfunction and lung inflammation. CD8(+) T cells of IFNAR(-/-) mice after HMPV infection expressed levels of the inhibitory receptor programmed death 1 (PD-1) similar to those of WT mice. However, despite lower expression of inhibitory programmed death ligand 1 (PD-L1), HMPV-specific CD8(+) T cells of IFNAR(-/-) mice were more functionally impaired than those of WT mice and upregulated the inhibitory receptor Tim-3. Analysis of the antigen-presenting cell subsets in the lungs revealed that the expansion of PD-L1(low) dendritic cells (DCs), but not PD-L1(high) alveolar macrophages, was dependent on IFNAR signaling. Collectively, our results indicate a role for IFNAR signaling in the early control of HMPV replication, disease progression, and the development of an optimal adaptive immune response. Moreover, our findings suggest an IFNAR-independent mechanism of lung CD8(+) T cell impairment. IMPORTANCE: Human metapneumovirus (HMPV) is a leading cause of acute respiratory illness. CD8(+) T cells are critical for clearing viral infection, yet recent evidence shows that HMPV and other respiratory viruses induce CD8(+) T cell impairment via PD-1-PD-L1 signaling. We sought to understand the role of type I interferon (IFN) in the innate and adaptive immune responses to HMPV by using a mouse model lacking IFN signaling. Although HMPV titers were higher in the absence of type I IFN, virus was nonetheless cleared and mice were less ill, indicating that type I IFN is not required to resolve HMPV infection but contributes to pathogenesis. Further, despite lower levels of the inhibitory ligand PD-L1 in mice lacking type I IFN, CD8(+) T cells were more impaired in these mice than in WT mice. Our data suggest that specific antigen-presenting cell subsets and the inhibitory receptor Tim-3 may contribute to CD8(+) T cell impairment.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Interferón Tipo I/metabolismo , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/inmunología , Transducción de Señal/inmunología , Replicación Viral/fisiología , Análisis de Varianza , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citometría de Flujo , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Interferón Tipo I/genética , Metapneumovirus/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oximetría , Infecciones por Paramyxoviridae/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Virales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
J Immunol ; 193(10): 5108-17, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25339663

RESUMEN

Reinfections with respiratory viruses are common and cause significant clinical illness, yet precise mechanisms governing this susceptibility are ill defined. Lung Ag-specific CD8(+) T cells (T(CD8)) are impaired during acute viral lower respiratory infection by the inhibitory receptor programmed death-1 (PD-1). To determine whether PD-1 contributes to recurrent infection, we first established a model of reinfection by challenging B cell-deficient mice with human metapneumovirus (HMPV) several weeks after primary infection, and found that HMPV replicated to high titers in the lungs. A robust secondary effector lung TCD8 response was generated during reinfection, but these cells were more impaired and more highly expressed the inhibitory receptors PD-1, LAG-3, and 2B4 than primary T(CD8). In vitro blockade demonstrated that PD-1 was the dominant inhibitory receptor early after reinfection. In vivo therapeutic PD-1 blockade during HMPV reinfection restored lung T(CD8) effector functions (i.e., degranulation and cytokine production) and enhanced viral clearance. PD-1 also limited the protective efficacy of HMPV epitope-specific peptide vaccination and impaired lung T(CD8) during heterotypic influenza virus challenge infection. Our results indicate that PD-1 signaling may contribute to respiratory virus reinfection and evasion of vaccine-elicited immune responses. These results have important implications for the design of effective vaccines against respiratory viruses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Paramyxoviridae/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Infecciones del Sistema Respiratorio/inmunología , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Degranulación de la Célula/inmunología , Regulación de la Expresión Génica , Humanos , Evasión Inmune , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Recuento de Linfocitos , Metapneumovirus/inmunología , Ratones , Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/virología , Receptor de Muerte Celular Programada 1/genética , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología , Transducción de Señal , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Carga Viral , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Replicación Viral , Proteína del Gen 3 de Activación de Linfocitos
15.
J Virol ; 88(11): 6368-79, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24672031

RESUMEN

UNLABELLED: Human metapneumovirus (HMPV) is a leading cause of respiratory disease in infants, children, and the elderly worldwide, yet no licensed vaccines exist. Live-attenuated vaccines present safety challenges, and protein subunit vaccines induce primarily antibody responses. Virus-like particles (VLPs) are an attractive alternative vaccine approach because of reduced safety concerns compared with live vaccines. We generated HMPV VLPs by expressing viral proteins in suspension-adapted human embryonic kidney epithelial (293-F) cells and found that the viral matrix (M) and fusion (F) proteins were sufficient to form VLPs. We previously reported that the VLPs resemble virus morphology and incorporate fusion-competent F protein (R. G. Cox, S. B. Livesay, M. Johnson, M. D. Ohi, and J. V. Williams, J. Virol. 86:12148-12160, 2012), which we hypothesized would elicit F-specific antibody and T cell responses. In this study, we tested whether VLP immunization could induce protective immunity to HMPV by using a mouse model. C57BL/6 mice were injected twice intraperitoneally with VLPs alone or with adjuvant and subsequently challenged with HMPV. Mice were euthanized 5 days postinfection, and virus titers, levels of neutralizing antibodies, and numbers of CD3(+) T cells were quantified. Mice immunized with VLPs mounted an F-specific antibody response and generated CD8(+) T cells recognizing an F protein-derived epitope. VLP immunization induced a neutralizing-antibody response that was enhanced by the addition of either TiterMax Gold or α-galactosylceramide adjuvant, though adjuvant reduced cellular immune responses. Two doses of VLPs conferred complete protection from HMPV replication in the lungs of mice and were not associated with a Th2-skewed cytokine response. These results suggest that nonreplicating VLPs are a promising vaccine candidate for HMPV. IMPORTANCE: Human metapneumovirus (HMPV) is a leading cause of acute respiratory infection in infants, children, and the elderly worldwide, yet no licensed vaccines exist. Live-attenuated vaccines present safety challenges, and protein subunit vaccines induce primarily antibody responses. Virus-like particles (VLPs) are an attractive alternative vaccine approach. We generated HMPV VLPs by expressing the viral matrix (M) and fusion (F) proteins in mammalian cells. We found that mice immunized with VLPs mounted an F-specific antibody response and generated CD8(+) T cells recognizing an F protein-derived epitope. VLP immunization induced a neutralizing-antibody response that was enhanced by the addition of either TiterMax Gold or α-galactosylceramide adjuvant. Two doses of VLPs conferred complete protection against HMPV replication in the lungs of mice and were not associated with a Th2-skewed cytokine response. These results suggest that nonreplicating VLPs are a promising vaccine candidate for HMPV.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular/inmunología , Metapneumovirus/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Análisis de Varianza , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Ensayo de Immunospot Ligado a Enzimas , Citometría de Flujo , Galactosilceramidas , Células HEK293 , Humanos , Inmunohistoquímica , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Poloxaleno , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vacunas de Partículas Similares a Virus/ultraestructura
16.
J Clin Invest ; 123(5): 1976-87, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23543059

RESUMEN

CD8+ T cells (TCD8) confer protective immunity against many infectious diseases, suggesting that microbial TCD8 determinants are promising vaccine targets. Nevertheless, current T cell antigen identification approaches do not discern which epitopes drive protective immunity during active infection - information that is critical for the rational design of TCD8-targeted vaccines. We employed a proteomics-based approach for large-scale discovery of naturally processed determinants derived from a complex pathogen, vaccinia virus (VACV), that are presented by the most frequent representatives of four major HLA class I supertypes. Immunologic characterization revealed that many previously unidentified VACV determinants were recognized by smallpox-vaccinated human peripheral blood cells in a variegated manner. Many such determinants were recognized by HLA class I-transgenic mouse immune TCD8 too and elicited protective TCD8 immunity against lethal intranasal VACV infection. Notably, efficient processing and stable presentation of immune determinants as well as the availability of naive TCD8 precursors were sufficient to drive a multifunctional, protective TCD8 response. Our approach uses fundamental insights into T cell epitope processing and presentation to define targets of protective TCD8 immunity within human pathogens that have complex proteomes, suggesting that this approach has general applicability in vaccine sciences.


Asunto(s)
Antígenos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T/citología , Virus Vaccinia/metabolismo , Animales , Presentación de Antígeno/inmunología , Epítopos/inmunología , Epítopos de Linfocito T/inmunología , Células HeLa , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Epítopos Inmunodominantes/inmunología , Espectrometría de Masas , Ratones , Ratones Transgénicos , Péptidos/inmunología , Fenotipo
17.
Exp Parasitol ; 132(2): 304-7, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22841523

RESUMEN

Human reticulocytes are one of the fundamental components needed to study the in vitro invasion processes of the human malaria parasite Plasmodium vivax. Additionally examinations of reticulocytes and their binding proteins are difficult in areas of the world that do not have access to advanced equipment or stem cell lines. These issues are particularly relevant to malaria vaccine candidate studies that are directed against surface proteins that the parasites use to gain entry into erythrocytes. Described here is a simple and inexpensive method to increase the reticulocyte count of cord blood samples. Exposure of cord blood to hypotonic saline (0.2%) for 5 min selectively lyses the non-reticulocytes resulting in an average 3.6-fold increase in reticulocyte count. Our studies show that this enrichment process does not damage the hemoglobin of the remaining erythrocytes which are still capable of supporting Plasmodium falciparum invasion and growth. This economical and rapid method of enrichment could facilitate studies of in vitro laboratory culturing of other malaria parasite species which preferentially invade reticulocytes such as P. vivax.


Asunto(s)
Sangre Fetal/citología , Soluciones Hipotónicas/farmacología , Reticulocitos/citología , Eritrocitos/metabolismo , Eritrocitos/parasitología , Femenino , Sangre Fetal/efectos de los fármacos , Sangre Fetal/parasitología , Hemoglobinas/análisis , Hemoglobinas/metabolismo , Humanos , Recién Nacido , Plasmodium falciparum/crecimiento & desarrollo , Embarazo , Recuento de Reticulocitos , Reticulocitos/efectos de los fármacos
18.
J Clin Invest ; 122(8): 2967-82, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22797302

RESUMEN

Viruses are leading causes of severe acute lower respiratory infections (LRIs). These infections evoke incomplete immunity, as individuals can be repeatedly reinfected throughout life. We report that acute viral LRI causes rapid pulmonary CD8+ cytotoxic T lymphocyte (TCD8) functional impairment via programmed death-1/programmed death ligand-1 (PD-1/PD-L1) signaling, a pathway previously associated with prolonged antigenic stimulation during chronic infections and cancer. PD-1-mediated TCD8 impairment occurred acutely in mice following infection with human metapneumovirus or influenza virus. Viral antigen was sufficient for PD-1 upregulation, but induction of PD-L1 was required for impairment. During secondary viral infection or epitope-only challenge, memory TCD8 rapidly reexpressed PD-1 and exhibited severe functional impairment. Inhibition of PD-1 signaling using monoclonal antibody blockade prevented TCD8 impairment, reduced viral titers during primary infection, and enhanced protection of immunized mice against challenge infection. Additionally, PD-1 and PD-L1 were upregulated in the lungs of patients with 2009 H1N1 influenza virus, respiratory syncytial virus, or parainfluenza virus infection. These results indicate that PD-1 mediates TCD8 functional impairment during acute viral infection and may contribute to recurrent viral LRIs. Therefore, the PD-1/PD-L1 pathway may represent a therapeutic target in the treatment of respiratory viruses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/metabolismo , Virosis/inmunología , Virosis/metabolismo , Enfermedad Aguda , Animales , Antígenos Virales , Antígeno HLA-B7/genética , Humanos , Imidazoles , Memoria Inmunológica , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana/inmunología , Gripe Humana/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Metapneumovirus , Ratones , Ratones Transgénicos , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Piridinas , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitial Respiratorio Humano , Transducción de Señal , Regulación hacia Arriba
19.
Cytometry A ; 73(6): 546-54, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18302186

RESUMEN

The complex life cycle of Plasmodium falciparum (Pf) makes it difficult to limit infections and reduce the risk of severe malaria. Improved understanding of Pf blood-stage growth and development would provide new opportunities to evaluate and interfere with successful completion of the parasite's life cycle. Cultured blood stage Pf was incubated with Hoechst 33342 (HO) and thiazole orange (TO) to stain DNA and total nucleic acids, respectively. Correlated HO and TO fluorescence emissions were then measured by flow cytometry. Complex bivariate data patterns were analyzed by manual cluster gating to quantify parasite life cycle stages. The permutations of viable staining with both reagents were tested for optimal detection of parasitized RBC (pRBC). Pf cultures were exposed to HO and TO simultaneously to achieve optimal staining of pRBC and consistent quantification of early and late stages of the replicative cycle (rings through schizonts). Staining of Pf nucleic acids allows for analysis of parasite development in the absence of fixatives, lysis, or radioactivity to enable examination of erythrocytes from parasite invasion through schizont rupture using sensitive and rapid assay procedures. Investigation of the mechanisms by which anti-malarial drugs and antibodies act against different Pf lifecycle stages will be aided by this cytometric strategy.


Asunto(s)
Bencimidazoles , Benzotiazoles , Citometría de Flujo/métodos , Colorantes Fluorescentes , Plasmodium falciparum/crecimiento & desarrollo , Quinolinas , Animales , ADN Protozoario/análisis , Eritrocitos/microbiología , Humanos , Estadios del Ciclo de Vida/fisiología , Malaria Falciparum/diagnóstico , ARN Protozoario/análisis , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...