Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
Intervalo de año de publicación
1.
J Gen Virol ; 104(8)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37643006

RESUMEN

Distinct cytomegaloviruses (CMVs) are widely distributed across their mammalian hosts in a highly host species-restricted pattern. To date, evidence demonstrating this has been limited largely to PCR-based approaches targeting small, conserved genomic regions, and only a few complete genomes of isolated viruses representing distinct CMV species have been sequenced. We have now combined direct isolation of infectious viruses from tissues with complete genome sequencing to provide a view of CMV diversity in a wild animal population. We targeted Natal multimammate mice (Mastomys natalensis), which are common in sub-Saharan Africa, are known to carry a variety of zoonotic pathogens, and are regarded as the primary source of Lassa virus (LASV) spillover into humans. Using transformed epithelial cells prepared from M. natalensis kidneys, we isolated CMVs from the salivary gland tissue of 14 of 37 (36 %) animals from a field study site in Mali. Genome sequencing showed that these primary isolates represent three different M. natalensis CMVs (MnatCMVs: MnatCMV1, MnatCMV2 and MnatCMV3), with some animals carrying multiple MnatCMVs or multiple strains of a single MnatCMV presumably as a result of coinfection or superinfection. Including primary isolates and plaque-purified isolates, we sequenced and annotated the genomes of two MnatCMV1 strains (derived from sequencing 14 viruses), six MnatCMV2 strains (25 viruses) and ten MnatCMV3 strains (21 viruses), totalling 18 MnatCMV strains isolated as 60 infectious viruses. Phylogenetic analysis showed that these MnatCMVs group with other murid viruses in the genus Muromegalovirus (subfamily Betaherpesvirinae, family Orthoherpesviridae), and that MnatCMV1 and MnatCMV2 are more closely related to each other than to MnatCMV3. The availability of MnatCMV isolates and the characterization of their genomes will serve as the prelude to the generation of a MnatCMV-based vaccine to target LASV in the M. natalensis reservoir.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Animales , Humanos , Ratones , Filogenia , Secuencia de Bases , Murinae
2.
Annu Rev Pharmacol Toxicol ; 59: 463-486, 2019 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-30134124

RESUMEN

Adverse drug reactions (ADRs) are a significant health care burden. Immune-mediated adverse drug reactions (IM-ADRs) are responsible for one-fifth of ADRs but contribute a disproportionately high amount of that burden due to their severity. Variation in human leukocyte antigen ( HLA) genes has emerged as a potential preprescription screening strategy for the prevention of previously unpredictable IM-ADRs. Immunopharmacogenomics combines the disciplines of immunogenomics and pharmacogenomics and focuses on the effects of immune-specific variation on drug disposition and IM-ADRs. In this review, we present the latest evidence for HLA associations with IM-ADRs, ongoing research into biological mechanisms of IM-ADRs, and the translation of clinical actionable biomarkers for IM-ADRs, with a focus on T cell-mediated ADRs.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inmunología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Hipersensibilidad a las Drogas/inmunología , Hipersensibilidad a las Drogas/prevención & control , Antígenos HLA/inmunología , Humanos , Farmacogenética/métodos , Linfocitos T/inmunología
3.
Immunol Cell Biol ; 98(4): 332-343, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31997396

RESUMEN

Vaccination against γ-herpesviruses has proved difficult. CD4+ T cells are essential to contain infection, but how best to prime them and whether this can reduce viral loads remain unclear. To address these questions, we used ovalbumin (OVA) as a model antigen, delivering it with murine cytomegalovirus (MCMV) to protect mice against OVA-expressing murine herpesvirus-4 (MuHV-4). Membrane-associated OVA (mOVA) was more effective than soluble OVA, both to prime CD4+ T cells and as an effector target. It was also a better target than an OVA epitope limited to infected cells, suggesting that protective CD4+ T cells recognize infected cell debris rather than infected cells themselves. While MCMV-mOVA protected acutely against MuHV-4-mOVA, long-term protection was incomplete, even when OVA-specific CD8+ T cells and B cells were also primed. Thus, even optimized single-target vaccines may poorly reduce long-term γ-herpesvirus infections.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Herpesviridae/inmunología , Vacunas contra Herpesvirus/inmunología , Inmunogenicidad Vacunal/inmunología , Ovalbúmina/inmunología , Rhadinovirus/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Infecciones por Herpesviridae/prevención & control , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células 3T3 NIH , Rhadinovirus/genética , Factores de Tiempo , Vacunación
4.
J Allergy Clin Immunol ; 144(1): 183-192, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30776417

RESUMEN

BACKGROUND: Vancomycin is a prevalent cause of the severe hypersensitivity syndrome drug reaction with eosinophilia and systemic symptoms (DRESS), which leads to significant morbidity and mortality and commonly occurs in the setting of combination antibiotic therapy, affecting future treatment choices. Variations in HLA class I in particular have been associated with serious T cell-mediated adverse drug reactions, which has led to preventive screening strategies for some drugs. OBJECTIVE: We sought to determine whether variation in the HLA region is associated with vancomycin-induced DRESS. METHODS: Probable vancomycin-induced DRESS cases were matched 1:2 with tolerant control subjects based on sex, race, and age by using BioVU, Vanderbilt's deidentified electronic health record database. Associations between DRESS and carriage of HLA class I and II alleles were assessed by means of conditional logistic regression. An extended sample set from BioVU was used to conduct a time-to-event analysis of those exposed to vancomycin with and without the identified HLA risk allele. RESULTS: Twenty-three subjects met the inclusion criteria for vancomycin-associated DRESS. Nineteen (82.6%) of 23 cases carried HLA-A*32:01 compared with 0 (0%) of 46 of the matched vancomycin-tolerant control subjects (P = 1 × 10-8) and 6.3% of the BioVU population (n = 54,249, P = 2 × 10-16). Time-to-event analysis of DRESS development during vancomycin treatment among the HLA-A*32:01-positive group indicated that 19.2% had DRESS and did so within 4 weeks. CONCLUSIONS: HLA-A*32:01 is strongly associated with vancomycin-induced DRESS in a population of predominantly European ancestry. HLA-A*32:01 testing could improve antibiotic safety, help implicate vancomycin as the causal drug, and preserve future treatment options with coadministered antibiotics.


Asunto(s)
Antibacterianos/efectos adversos , Síndrome de Hipersensibilidad a Medicamentos/inmunología , Antígenos HLA-A/inmunología , Vancomicina/efectos adversos , Adolescente , Adulto , Anciano , Antibacterianos/química , Síndrome de Hipersensibilidad a Medicamentos/etiología , Femenino , Antígenos HLA-A/química , Humanos , Masculino , Persona de Mediana Edad , Simulación del Acoplamiento Molecular , Vancomicina/química , Adulto Joven
5.
J Gen Virol ; 100(12): 1695-1700, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31609196

RESUMEN

Cytomegaloviruses (CMVs) use myeloid cells to move within their hosts. Murine CMV (MCMV) colonizes the salivary glands for long-term shedding, and reaches them via CD11c+ infected cells. A need to recruit patrolling monocytes for systemic spread has been proposed, based on poor salivary gland infection in fractalkine receptor (CX3CR1)-deficient mice. We found no significant CX3CR1 dependence of salivary gland infection. CCL2 and the viral m131/m129 chemokine homologue were also redundant for acute MCMV spread, arguing against a need for inflammation or infection to recruit additional monocytes to the entry site. M131/m129 promoted salivary gland infection, but only after the initial seeding of infected cells to this site. Our data support the idea that MCMV disseminates by infecting and mobilizing tissue-resident dendritic cells.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Quimiocina CCL2/metabolismo , Quimiocinas CC/metabolismo , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Interacciones Huésped-Patógeno , Muromegalovirus/fisiología , Proteínas Virales/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C/genética , Células Dendríticas/metabolismo , Células Dendríticas/virología , Ratones , Ratones Noqueados , Monocitos/metabolismo , Monocitos/virología , Unión Proteica , Replicación Viral
6.
J Immunol ; 196(5): 2205-2218, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26810224

RESUMEN

The Alphaherpesvirinae subfamily includes HSV types 1 and 2 and the sequence-divergent pathogen varicella zoster virus (VZV). T cells, controlled by TCR and HLA molecules that tolerate limited epitope amino acid variation, might cross-react between these microbes. We show that memory PBMC expansion with either HSV or VZV enriches for CD4 T cell lines that recognize the other agent at the whole-virus, protein, and peptide levels, consistent with bidirectional cross-reactivity. HSV-specific CD4 T cells recovered from HSV-seronegative persons can be explained, in part, by such VZV cross-reactivity. HSV-1-reactive CD8 T cells also cross-react with VZV-infected cells, full-length VZV proteins, and VZV peptides, as well as kill VZV-infected dermal fibroblasts. Mono- and cross-reactive CD8 T cells use distinct TCRB CDR3 sequences. Cross-reactivity to VZV is reconstituted by cloning and expressing TCRA/TCRB receptors from T cells that are initially isolated using HSV reagents. Overall, we define 13 novel CD4 and CD8 HSV-VZV cross-reactive epitopes and strongly imply additional cross-reactive peptide sets. Viral proteins can harbor both CD4 and CD8 HSV/VZV cross-reactive epitopes. Quantitative estimates of HSV/VZV cross-reactivity for both CD4 and CD8 T cells vary from 10 to 50%. Based on these findings, we hypothesize that host herpesvirus immune history may influence the pathogenesis and clinical outcome of subsequent infections or vaccinations for related pathogens and that cross-reactive epitopes and TCRs may be useful for multi-alphaherpesvirus vaccine design and adoptive cellular therapy.


Asunto(s)
Alphaherpesvirinae/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Reacciones Cruzadas/inmunología , Infecciones por Herpesviridae/inmunología , Presentación de Antígeno/inmunología , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Epítopos de Linfocito T/inmunología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 2/inmunología , Humanos , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Proteínas Virales/inmunología
7.
J Gen Virol ; 96(Pt 5): 1127-1137, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25575707

RESUMEN

Infection with multiple genetically distinct strains of pathogen is common and can lead to positive (complementation) or negative (competitive) within-host interactions. These interactions can alter aspects of the disease process and help shape pathogen evolution. Infection of the host with multiple strains of cytomegalovirus (CMV) occurs frequently in humans and mice. Profound, NK-cell-mediated (apparent) competition has been identified in C57BL/6 mice, and prevented the replication and shedding of certain co-infecting CMV strains. However, the frequency of such strong competition has not been established. Other within-host interactions such as complementation or alternative forms of competition remain possible. Moreover, high rates of recombination in both human CMV and murine CMV (MCMV) suggest prolonged periods of viral co-replication, rather than strong competitive suppression. An established model was employed to investigate the different possible outcomes of multi-strain infection in other mouse strains. In this study, co-replication of up to four strains of MCMV in the spleen, liver and salivary glands was observed in both MCMV-susceptible and MCMV-resistant mice. In the absence of apparent competition, no other forms of competition were unmasked. In addition, no evidence of complementation between viral strains was observed. Importantly, co-replication of MCMV strains was apparent for up to 90 days in the salivary glands. These data indicated that competition was not the default outcome of multi-strain CMV infection. Prolonged, essentially neutral, co-replication may be the norm, allowing for multi-strain transmission and prolonged opportunities for recombination.


Asunto(s)
Coinfección/virología , Infecciones por Herpesviridae/virología , Muromegalovirus/crecimiento & desarrollo , Subfamilia A de Receptores Similares a Lectina de Células NK/inmunología , Glándulas Salivales/virología , Animales , Hígado/virología , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Subfamilia A de Receptores Similares a Lectina de Células NK/deficiencia , Bazo/virología
8.
PLoS Pathog ; 9(1): e1003111, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23300458

RESUMEN

It is becoming increasingly clear that many diseases are the result of infection from multiple genetically distinct strains of a pathogen. Such multi-strain infections have the capacity to alter both disease and pathogen dynamics. Infection with multiple strains of human cytomegalovirus (HCMV) is common and has been linked to enhanced disease. Suggestions that disease enhancement in multi-strain infected patients is due to complementation have been supported by trans-complementation studies in mice during co-infection of wild type and gene knockout strains of murine CMV (MCMV). Complementation between naturally circulating strains of CMV has, however, not been assessed. In addition, many models of multi-strain infection predict that co-infecting strains will compete with each other and that this competition may contribute to selective transmission of more virulent pathogen strains. To assess the outcome of multi-strain infection, C57BL/6 mice were infected with up to four naturally circulating strains of MCMV. In this study, profound within-host competition was observed between co-infecting strains of MCMV. This competition was MCMV strain specific and resulted in the complete exclusion of certain strains of MCMV from the salivary glands of multi-strain infected mice. Competition was dependent on Ly49H(+) natural killer (NK) cells as well as the expression of the ligand for Ly49H, the MCMV encoded product, m157. Strains of MCMV which expressed an m157 gene product capable of ligating Ly49H were outcompeted by strains of MCMV expressing variant m157 genes. Importantly, within-host competition prevented the shedding of the less virulent strains of MCMV, those recognized by Ly49H, into the saliva of multi-strain infected mice. These data demonstrate that NK cells have the strain specific recognition capacity required to meditate within-host competition between strains of MCMV. Furthermore, this within-host competition has the capacity to shape the dynamics of viral shedding and potentially select for the transmission of more virulent virus strains.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Citomegalovirus/inmunología , Células Asesinas Naturales/inmunología , Subfamilia A de Receptores Similares a Lectina de Células NK/inmunología , Animales , Anticuerpos Bloqueadores/inmunología , Anticuerpos Monoclonales/inmunología , Antígenos Virales/biosíntesis , Antígenos Virales/inmunología , Células Cultivadas , Citomegalovirus/clasificación , Citomegalovirus/patogenicidad , Células Asesinas Naturales/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Glándulas Salivales/virología , Proteínas Virales/inmunología
9.
Front Pharmacol ; 13: 858557, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35431929

RESUMEN

Asbestos-induced preclinical mouse models of mesothelioma produce tumors that are very similar to those that develop in humans and thus represent an ideal platform to study this rare, universally fatal tumor type. Our team and a number of other research groups have established such models as a stepping stone to new treatments, including chemotherapy, immunotherapy and other approaches that have been/are being translated into clinical trials. In some cases this work has led to changes in mesothelioma treatment practice and over the last 30 years these models and studies have led to trials which have improved the response rate in mesothelioma from less than 10% to over 50%. Mouse models have had a vital role in that improvement and will continue to play a key role in the future success of mesothelioma immunotherapy. In this review we focus only on these original inbred mouse models, the large number of preclinical studies conducted using them and their contribution to current and future clinical therapy for mesothelioma.

10.
Oncoimmunology ; 11(1): 2038403, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35186441

RESUMEN

The process of tumorigenesis leaves a series of indelible genetic changes in tumor cells, that when expressed, have the potential to be tumor-specific immune targets. Neoantigen vaccines that capitalize on this potential immunogenicity have shown efficacy in preclinical models and have now entered clinical trials. Here we discuss the status of personalized neoantigen vaccines and the current major challenges to this nascent field. In particular, we focus on the types of antigens that can be targeted by vaccination and on the role that preexisting immunosuppression, and in particular T-cell exhaustion, will play in the development of effective cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/uso terapéutico , Humanos , Inmunoterapia , Neoplasias/tratamiento farmacológico , Vacunación
11.
Commun Biol ; 5(1): 133, 2022 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-35173258

RESUMEN

Pre-existing pathogen-specific memory T cell responses can contribute to multiple adverse outcomes including autoimmunity and drug hypersensitivity. How the specificity of the T cell receptor (TCR) is subverted or seconded in many of these diseases remains unclear. Here, we apply abacavir hypersensitivity (AHS) as a model to address this question because the disease is linked to memory T cell responses and the HLA risk allele, HLA-B*57:01, and the initiating insult, abacavir, are known. To investigate the role of pathogen-specific TCR specificity in mediating AHS we performed a genome-wide screen for HLA-B*57:01 restricted T cell responses to Epstein-Barr virus (EBV), one of the most prevalent human pathogens. T cell epitope mapping revealed HLA-B*57:01 restricted responses to 17 EBV open reading frames and identified an epitope encoded by EBNA3C. Using these data, we cloned the dominant TCR for EBNA3C and a previously defined epitope within EBNA3B. TCR specificity to each epitope was confirmed, however, cloned TCRs did not cross-react with abacavir plus self-peptide. Nevertheless, abacavir inhibited TCR interactions with their cognate ligands, demonstrating that TCR specificity may be subverted by a drug molecule. These results provide an experimental road map for future studies addressing the heterologous immune responses of TCRs including T cell mediated adverse drug reactions.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Didesoxinucleósidos , Epítopos de Linfocito T , Antígenos HLA-B , Herpesvirus Humano 4/genética , Humanos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Complemento 3d
12.
JCI Insight ; 7(1)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35014624

RESUMEN

Congenital cytomegalovirus (cCMV) infection is the leading infectious cause of neurodevelopmental disorders. However, the neuropathogenesis remains largely elusive due to a lack of informative animal models. In this study, we developed a congenital murine CMV (cMCMV) infection mouse model with high survival rate and long survival period that allowed long-term follow-up study of neurodevelopmental disorders. This model involves in utero intracranial injection and mimics many reported clinical manifestations of cCMV infection in infants, including growth restriction, hearing loss, and impaired cognitive and learning-memory abilities. We observed that abnormalities in MRI/CT neuroimaging were consistent with brain hemorrhage and loss of brain parenchyma, which was confirmed by pathological analysis. Neuropathological findings included ventriculomegaly and cortical atrophy associated with impaired proliferation and migration of neural progenitor cells in the developing brain at both embryonic and postnatal stages. Robust inflammatory responses during infection were shown by elevated inflammatory cytokine levels, leukocyte infiltration, and activation of microglia and astrocytes in the brain. Pathological analyses and CT neuroimaging revealed brain calcifications induced by cMCMV infection and cell death via pyroptosis. Furthermore, antiviral treatment with ganciclovir significantly improved neurological functions and mitigated brain damage as shown by CT neuroimaging. These results demonstrate that this model is suitable for investigation of mechanisms of infection-induced brain damage and long-term studies of neurodevelopmental disorders, including the development of interventions to limit CNS damage associated with cCMV infection.


Asunto(s)
Infecciones por Citomegalovirus , Modelos Animales de Enfermedad , Neuroimagen , Animales , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/diagnóstico por imagen , Infecciones por Citomegalovirus/fisiopatología , Infecciones por Citomegalovirus/terapia , Femenino , Estudios de Seguimiento , Ratones , Ratones Endogámicos ICR , Embarazo
13.
Synth Biol (Oxf) ; 6(1): ysab018, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712842

RESUMEN

Diverse applications rely on engineering microbes to carry and express foreign transgenes. This engineered baggage rarely benefits the microbe and is thus prone to rapid evolutionary loss when the microbe is propagated. For applications where a transgene must be maintained for extended periods of growth, slowing the rate of transgene evolution is critical and can be achieved by reducing either the rate of mutation or the strength of selection. Because the benefits realized by changing these quantities will not usually be equal, it is important to know which will yield the greatest improvement to the evolutionary half-life of the engineering. Here, we provide a method for jointly estimating the mutation rate of transgene loss and the strength of selection favoring these transgene-free, revertant individuals. The method requires data from serial transfer experiments in which the frequency of engineered genomes is monitored periodically. Simple mathematical models are developed that use these estimates to predict the half-life of the engineered transgene and provide quantitative predictions for how alterations to mutation and selection will influence longevity. The estimation method and predictive tools have been implemented as an interactive web application, MuSe.

14.
Pathogens ; 10(7)2021 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-34358016

RESUMEN

As the largest herpesviruses, the 230 kb genomes of cytomegaloviruses (CMVs) have increased our understanding of host immunity and viral escape mechanisms, although many of the annotated genes remain as yet uncharacterised. Here we identify the m15 locus of murine CMV (MCMV) as a viral modulator of natural killer (NK) cell immunity. We show that, rather than discrete transcripts from the m14, m15 and m16 genes as annotated, there are five 3'-coterminal transcripts expressed over this region, all utilising a consensus polyA tail at the end of the m16 gene. Functional inactivation of any one of these genes had no measurable impact on viral replication. However, disruption of all five transcripts led to significantly attenuated dissemination to, and replication in, the salivary glands of multiple strains of mice, but normal growth during acute infection. Disruption of the m15 locus was associated with heightened NK cell responses, including enhanced proliferation and IFNγ production. Depletion of NK cells, but not T cells, rescued salivary gland replication and viral shedding. These data demonstrate the identification of multiple transcripts expressed by a single locus which modulate, perhaps in a concerted fashion, the function of anti-viral NK cells.

15.
Hum Immunol ; 81(6): 300-304, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32173028

RESUMEN

Abacavir hypersensitivity syndrome (ABC HSS) is strongly associated with carriage of human leukocyte antigen (HLA)-B*57:01, which has a 100% negative predictive value for the development of ABC HSS. However, 45% of individuals who carry HLA-B*57:01 can tolerate ABC. We investigated immune and non-immune related genes in ABC HSS (n = 95) and ABC tolerant (n = 43) HLA-B*57:01 + patients to determine other factors required for the development of ABC HSS. Assignment of phenotype showed that ABC HSS subjects were significantly less likely than tolerants to carry only ERAP1 hypoactive trimming allotypes (p = 0.02). An altered self-peptide repertoire model by which abacavir activates T cells is in keeping with observation that endoplasmic reticulum aminopeptidase 1 (ERAP1) allotypes that favour efficient peptide trimming are more common in ABC HSS patients compared to patients who tolerate ABC. Independently, non-specific immune activation via soluble cluster of differentiation antigen 14 (sCD14) may also influence susceptibility to ABC HSS.


Asunto(s)
Fármacos Anti-VIH/uso terapéutico , Didesoxinucleósidos/uso terapéutico , Síndrome de Hipersensibilidad a Medicamentos/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Infecciones por VIH/tratamiento farmacológico , VIH-1/fisiología , Antígenos HLA-B/genética , Alérgenos/inmunología , Aminopeptidasas/genética , Fármacos Anti-VIH/inmunología , Didesoxinucleósidos/efectos adversos , Didesoxinucleósidos/inmunología , Síndrome de Hipersensibilidad a Medicamentos/etiología , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Infecciones por VIH/complicaciones , Humanos , Receptores de Lipopolisacáridos/genética , Masculino , Antígenos de Histocompatibilidad Menor/genética , Fenotipo , Estudios Retrospectivos
16.
J Exp Med ; 216(8): 1809-1827, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31142589

RESUMEN

CMVs efficiently target MHC I molecules to avoid recognition by cytotoxic T cells. However, the lack of MHC I on the cell surface renders the infected cell susceptible to NK cell killing upon missing self recognition. To counter this, mouse CMV (MCMV) rescues some MHC I molecules to engage inhibitory Ly49 receptors. Here we identify a new viral protein, MATp1, that is essential for MHC I surface rescue. Rescued altered-self MHC I molecules show increased affinity to inhibitory Ly49 receptors, resulting in inhibition of NK cells despite substantially reduced MHC I surface levels. This enables the virus to evade recognition by licensed NK cells. During evolution, this novel viral immune evasion mechanism could have prompted the development of activating NK cell receptors that are specific for MATp1-modified altered-self MHC I molecules. Our study solves a long-standing conundrum of how MCMV avoids recognition by NK cells, unravels a fundamental new viral immune evasion mechanism, and demonstrates how this forced the evolution of virus-specific activating MHC I-restricted Ly49 receptors.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Evasión Inmune/inmunología , Células Asesinas Naturales/inmunología , Muromegalovirus/metabolismo , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Proteínas Virales/metabolismo , Animales , Antígenos Ly/genética , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Femenino , Fibroblastos/metabolismo , Infecciones por Herpesviridae/virología , Antígenos de Histocompatibilidad Clase I/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor 1 Gatillante de la Citotoxidad Natural/genética
17.
J Exp Med ; 215(2): 575-594, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29282254

RESUMEN

Upon infection with an intracellular pathogen, cytotoxic CD8+ T cells develop diverse differentiation states characterized by function, localization, longevity, and the capacity for self-renewal. The program of differentiation is determined, in part, by FOXO1, a transcription factor known to integrate extrinsic input in order to specify survival, DNA repair, self-renewal, and proliferation. At issue is whether the state of T cell differentiation is specified by initial conditions of activation or is actively maintained. To study the spectrum of T cell differentiation, we have analyzed an infection with mouse cytomegalovirus, a persistent-latent virus that elicits different cytotoxic T cell responses characterized as acute resolving or inflationary. Our results show that FOXO1 is continuously required for all the phenotypic characteristics of memory-effector T cells such that with acute inactivation of the gene encoding FOXO1, T cells revert to a short-lived effector phenotype, exhibit reduced viability, and manifest characteristics of anergy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Anergia Clonal , Proteína Forkhead Box O1/inmunología , Memoria Inmunológica , Traslado Adoptivo , Animales , Antígenos Virales , Linfocitos T CD8-positivos/citología , Diferenciación Celular/inmunología , Supervivencia Celular/inmunología , Proteína Forkhead Box O1/deficiencia , Proteína Forkhead Box O1/genética , Factor Nuclear 1-alfa del Hepatocito/inmunología , Lectinas Tipo C , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Muromegalovirus/inmunología , Receptores Inmunológicos/inmunología
18.
Front Immunol ; 9: 1861, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30154789

RESUMEN

Tissue-resident memory T (TRM) cells provide first-line defense against invading pathogens encountered at barrier sites. In the lungs, TRM cells protect against respiratory infections, but wane more quickly than TRM cells in other tissues. This lack of a sustained TRM population in the lung parenchyma explains, at least in part, why infections with some pathogens, such as influenza virus and respiratory syncytial virus (RSV), recur throughout life. Intranasal (IN) vaccination with a murine cytomegalovirus (MCMV) vector expressing the M protein of RSV (MCMV-M) has been shown to elicit robust populations of CD8+ TRM cells that accumulate over time and mediate early viral clearance. To extend this finding, we compared the inflationary CD8+ T cell population elicited by MCMV-M vaccination with a conventional CD8+ T cell population elicited by an MCMV vector expressing the M2 protein of RSV (MCMV-M2). Vaccination with MCMV-M2 induced a population of M2-specific CD8+ TRM cells that waned rapidly, akin to the M2-specific CD8+ TRM cell population elicited by infection with RSV. In contrast to the natural immunodominance profile, however, coadministration of MCMV-M and MCMV-M2 did not suppress the M-specific CD8+ T cell response, suggesting that progressive expansion was driven by continuous antigen presentation, irrespective of the competitive or regulatory effects of M2-specific CD8+ T cells. Moreover, effective viral clearance mediated by M-specific CD8+ TRM cells was not affected by the coinduction of M2-specific CD8+ T cells. These data show that memory inflation is required for the maintenance of CD8+ TRM cells in the lungs after IN vaccination with MCMV.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Memoria Inmunológica , Pulmón/inmunología , Muromegalovirus/inmunología , Administración Intranasal , Animales , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Epítopos de Linfocito T/inmunología , Femenino , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/patología , Inmunización , Pulmón/metabolismo , Pulmón/patología , Ratones , Especificidad de Órganos , Especificidad del Receptor de Antígeno de Linfocitos T
19.
J Allergy Clin Immunol Pract ; 6(4): 1287-1296.e1, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29100867

RESUMEN

BACKGROUND: For severe cutaneous adverse reactions (SCARs) associated with multiple antibiotics dosed concurrently, clinical causality is challenging and diagnostic approaches are limited, leading to constricted future antibiotic choices. OBJECTIVE: To examine the combined utility of in vivo and ex vivo diagnostic approaches at assigning drug causality in a cohort of patients with antibiotic-associated (AA)-SCARs. METHODS: Patients with AA-SCARs were prospectively recruited between April 2015 and February 2017. In vivo testing (patch testing or delayed intradermal testing) was performed to the implicated antibiotic(s) at the highest nonirritating concentration and read at 24 hours through 1 week. Ex vivo testing used patient peripheral blood mononuclear cells (PBMCs) stimulated with a range of pharmacologically relevant concentrations of implicated antibiotics to measure dose-dependent IFN-γ release from CD4+ and CD8+ T cells via an enzyme-linked immunoSpot assay. RESULTS: In 19 patients with AA-SCARs, combined in vivo and ex vivo testing assigned antibiotic causality in 15 (79%) patients. Ten patients (53%) with AA-SCARs were positive on IFN-γ release enzyme-linked immunoSpot assay, with an overall reported sensitivity of 52% (95% CI, 29-76) and specificity of 100% (95% CI, 79-100), with improved sensitivity noted in acute (within 1 day to 6 weeks after SCAR onset) testing (75%) and in patients with higher phenotypic scores (59%). There was increased use of narrow-spectrum beta-lactams and antibiotics from within the implicated class following testing in patients with a positive ex vivo or in vivo test result. CONCLUSIONS: We demonstrate the potential utility of combined in vivo and ex vivo testing in patients with AA-SCARs to assign drug causality with high specificity.


Asunto(s)
Antibacterianos/efectos adversos , Erupciones por Medicamentos , Interferón gamma/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Erupciones por Medicamentos/diagnóstico , Erupciones por Medicamentos/etiología , Erupciones por Medicamentos/inmunología , Ensayo de Immunospot Ligado a Enzimas , Femenino , Humanos , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , Proyectos Piloto , Pruebas Cutáneas , Adulto Joven
20.
Expert Rev Vaccines ; 15(1): 31-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26524478

RESUMEN

Modern human activity fueled by economic development is profoundly altering our relationship with microorganisms. This altered interaction with microbes is believed to be the major driving force behind the increased rate of emerging infectious diseases from animals. The spate of recent infectious disease outbreaks, including Ebola virus disease and Middle East respiratory syndrome, emphasize the need for development of new innovative tools to manage these emerging diseases. Disseminating vaccines are one such novel approach to potentially interrupt animal to human (zoonotic) transmission of these pathogens.


Asunto(s)
Enfermedades Transmisibles Emergentes/epidemiología , Enfermedades Transmisibles Emergentes/prevención & control , Transmisión de Enfermedad Infecciosa/prevención & control , Inmunización/métodos , Vacunas/inmunología , Zoonosis/epidemiología , Zoonosis/prevención & control , Animales , Enfermedades Transmisibles Emergentes/transmisión , Humanos , Vacunas/administración & dosificación , Vacunas/aislamiento & purificación , Zoonosis/transmisión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA