Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.064
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 36: 193-220, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29328787

RESUMEN

Retroviruses are genome invaders that have shared a long history of coevolution with vertebrates and their immune system. Found endogenously in genomes as traces of past invasions, retroviruses are also considerable threats to human health when they exist as exogenous viruses such as HIV. The immune response to retroviruses is engaged by germline-encoded sensors of innate immunity that recognize viral components and damage induced by the infection. This response develops with the induction of antiviral effectors and launching of the clonal adaptive immune response, which can contribute to protective immunity. However, retroviruses efficiently evade the immune response, owing to their rapid evolution. The failure of specialized immune cells to respond, a form of neglect, may also contribute to inadequate antiretroviral immune responses. Here, we discuss the mechanisms by which immune responses to retroviruses are mounted at the molecular, cellular, and organismal levels. We also discuss how intrinsic, innate, and adaptive immunity may cooperate or conflict during the generation of immune responses.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Retroviridae/inmunología , Inmunidad Adaptativa , Animales , Humanos , Evasión Inmune , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunidad Innata , Infecciones por Retroviridae/metabolismo
2.
PLoS Pathog ; 20(8): e1012505, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39208378

RESUMEN

Replication of the complex retrovirus mouse mammary tumor virus (MMTV) is antagonized by murine Apobec3 (mA3), a member of the Apobec family of cytidine deaminases. We have shown that MMTV-encoded Rem protein inhibits proviral mutagenesis by the Apobec enzyme, activation-induced cytidine deaminase (AID) during viral replication in BALB/c mice. To further study the role of Rem in vivo, we have infected C57BL/6 (B6) mice with a superantigen-independent lymphomagenic strain of MMTV (TBLV-WT) or a mutant strain that is defective in Rem and its cleavage product Rem-CT (TBLV-SD). Compared to BALB/c, B6 mice were more susceptible to TBLV infection and tumorigenesis. Furthermore, unlike MMTV, TBLV induced T-cell tumors in B6 µMT mice, which lack membrane-bound IgM and conventional B-2 cells. At limiting viral doses, loss of Rem expression in TBLV-SD-infected B6 mice accelerated tumorigenesis compared to TBLV-WT in either wild-type B6 or AID-knockout mice. Unlike BALB/c results, high-throughput sequencing indicated that proviral G-to-A or C-to-T mutations were unchanged regardless of Rem expression in B6 tumors. However, knockout of both AID and mA3 reduced G-to-A mutations. Ex vivo stimulation showed higher levels of mA3 relative to AID in B6 compared to BALB/c splenocytes, and effects of agonists differed in the two strains. RNA-Seq revealed increased transcripts related to growth factor and cytokine signaling in TBLV-SD-induced tumors relative to TBLV-WT-induced tumors, consistent with another Rem function. Thus, Rem-mediated effects on tumorigenesis in B6 mice are independent of Apobec-mediated proviral hypermutation.


Asunto(s)
Citidina Desaminasa , Virus del Tumor Mamario del Ratón , Infecciones por Retroviridae , Animales , Femenino , Ratones , Desaminasas APOBEC/genética , Desaminasas APOBEC/metabolismo , Carcinogénesis/genética , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Virus del Tumor Mamario del Ratón/genética , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Infecciones por Retroviridae/genética , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/virología , Infecciones Tumorales por Virus/inmunología , Replicación Viral
3.
Immunity ; 47(2): 310-322.e7, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28813660

RESUMEN

Select humans and animals control persistent viral infections via adaptive immune responses that include production of neutralizing antibodies. The precise genetic basis for the control remains enigmatic. Here, we report positional cloning of the gene responsible for production of retrovirus-neutralizing antibodies in mice of the I/LnJ strain. It encodes the beta subunit of the non-classical major histocompatibility complex class II (MHC-II)-like molecule H2-O, a negative regulator of antigen presentation. The recessive and functionally null I/LnJ H2-Ob allele supported the production of virus-neutralizing antibodies independently of the classical MHC haplotype. Subsequent bioinformatics and functional analyses of the human H2-Ob homolog, HLA-DOB, revealed both loss- and gain-of-function alleles, which could affect the ability of their carriers to control infections with human hepatitis B (HBV) and C (HCV) viruses. Thus, understanding of the previously unappreciated role of H2-O (HLA-DO) in immunity to infections may suggest new approaches in achieving neutralizing immunity to viruses.


Asunto(s)
Anticuerpos Neutralizantes , Antígenos HLA-D/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Inmunidad Humoral , Virus del Tumor Mamario del Ratón/inmunología , Virus Rauscher/inmunología , Infecciones por Retroviridae/inmunología , Animales , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Presentación de Antígeno/genética , Biología Computacional , Femenino , Predisposición Genética a la Enfermedad , Antígenos HLA-D/genética , Células HeLa , Hepatitis B/inmunología , Hepatitis B/transmisión , Hepatitis C/inmunología , Hepatitis C/transmisión , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Inmunidad Humoral/genética , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Mutación/genética , Polimorfismo Genético , Infecciones por Retroviridae/transmisión
4.
J Virol ; 97(5): e0193022, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37093008

RESUMEN

Inbred mouse lines vary in their ability to mount protective antiretroviral immune responses, and even closely related strains can exhibit opposing phenotypes upon retroviral infection. Here, we found that 129S mice inherit a previously unknown mechanism for the production of anti-murine leukemia virus (MLV) antibodies and control of infection. The resistant phenotype in 129S1 mice is controlled by two dominant loci that are independent from known MLV resistance genes. We also show that production of anti-MLV antibodies in 129S7 mice, but not 129S1 mice, is independent of interferon gamma signaling. Thus, our data indicate that 129S mice inherit an unknown mechanism for control of MLV infection and demonstrate that there is genetic variability in 129S substrains that affects their ability to mount antiviral immune responses. IMPORTANCE Understanding the genetic basis for production of protective antiviral immune responses is crucial for the development of novel vaccines and adjuvants. Additionally, characterizing the genetic and phenotypic variability in inbred mice has implications for the selection of strains for targeted mutagenesis, choice of controls, and for broader understanding of the requirements for protective immunity.


Asunto(s)
Ratones Endogámicos , Infecciones por Retroviridae , Animales , Ratones , Inmunidad , Interferón gamma , Virus de la Leucemia Murina/genética , Ratones Endogámicos/genética , Ratones Endogámicos/inmunología , Infecciones por Retroviridae/inmunología
5.
J Virol ; 97(1): e0179522, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36598198

RESUMEN

Activation-induced cytidine deaminase/apolipoprotein B mRNA editing catalytic polypeptide-like (AID/APOBEC) proteins are cytosine deaminases implicated in diverse biological functions. APOBEC1 (A1) proteins have long been thought to regulate lipid metabolism, whereas the evolutionary significance of A1 proteins in antiviral defense remains largely obscure. Endogenous retroviruses (ERVs) document past retroviral infections and are ubiquitous within the vertebrate genomes. Here, we identify the A1 gene repertoire, characterize the A1-mediated mutation footprints in ERVs, and interrogate the evolutionary arms race between A1 genes and ERVs across vertebrate species. We find that A1 genes are widely present in tetrapods, recurrently amplified and lost in certain lineages, suggesting that A1 genes might have originated during the early evolution of tetrapods. A1-mediated mutation footprints can be detected in ERVs across tetrapods. Moreover, A1 genes appear to have experienced episodic positive selection in many tetrapod lineages. Taken together, we propose that a long-running arms race between A1 genes and retroviruses might have persisted throughout the evolutionary course of tetrapods. IMPORTANCE APOBEC3 (A3) genes have been thought to function in defense against retroviruses, whereas the evolutionary significance of A1 proteins in antiviral defense remains largely obscure. In this study, we identify the A1 gene repertoire, characterize the A1-mediated mutation footprints in endogenous retroviruses (ERVs), and explore the evolutionary arms race between A1 genes and ERVs across vertebrate species. We found A1 proteins originated during the early evolution of tetrapods, and detected the footprints of A1-induced hypermutations in retroviral fossils. A1 genes appear to have experienced pervasive positive selection in tetrapods. Our study indicates a long-running arms race between A1 genes and retroviruses taking place throughout the evolutionary course of tetrapods.


Asunto(s)
Desaminasas APOBEC-1 , Retrovirus Endógenos , Evolución Molecular , Infecciones por Retroviridae , Animales , Desaminasas APOBEC-1/genética , Desaminasas APOBEC-1/inmunología , Retrovirus Endógenos/clasificación , Retrovirus Endógenos/genética , Retrovirus Endógenos/inmunología , Mutación , Filogenia , Infecciones por Retroviridae/inmunología , Vertebrados/inmunología
6.
Virol J ; 21(1): 177, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39107806

RESUMEN

BACKGROUND: Reticuloendotheliosis virus (REV), a member of the family Retroviridae, is a hot area of research, and a previous study showed that exosomes purified from REV-positive semen were not blocked by REV-specific neutralizing antibodies and established productive infections. METHODS: To further verify the infectivity of exosomes from REV-infected cells, we isolated and purified exosomes from REV-infected DF-1 cells and identified them using Western blot and a transmission electron microscope. We then inoculated 7-day-old embryonated eggs, 1-day-old chicks and 23-week-old hens with and without antibody treatment. REV was administered simultaneously as a control. RESULTS: In the absence of antibodies, the results indicated that REV-exosomes and REV could infect chicks, resulting in viremia and viral shedding, compared with the infection caused by REV, REV-exosomes reduced the hatching rate and increased mortality after hatching, causing severe growth inhibition and immune organ damage in 1-day-old chicks; both REV and REV-exosomes also could infect hens, however, lead to transient infection. In the presence of antibodies, REV-exosomes were not blocked by REV-specific neutralizing antibodies and infected 7-day-old embryonated eggs. However, REV could not infect 1-day-old chicks and 23-week-old hens. CONCLUSION: In this study, we compared the infectious ability of REV-exosomes and REV, REV-exosomes could escape from REV-specific neutralizing antibodies in embryonated eggs, providing new insights into the immune escape mechanism of REV.


Asunto(s)
Anticuerpos Antivirales , Pollos , Exosomas , Enfermedades de las Aves de Corral , Virus de la Reticuloendoteliosis , Infecciones por Retroviridae , Esparcimiento de Virus , Animales , Exosomas/virología , Exosomas/inmunología , Anticuerpos Antivirales/inmunología , Pollos/virología , Virus de la Reticuloendoteliosis/inmunología , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/transmisión , Enfermedades de las Aves de Corral/inmunología , Infecciones por Retroviridae/virología , Infecciones por Retroviridae/transmisión , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/veterinaria , Anticuerpos Neutralizantes/inmunología , Línea Celular , Viremia/virología , Femenino
7.
PLoS Genet ; 16(6): e1008471, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32525879

RESUMEN

Viruses and their hosts are locked in an evolutionary race where resistance to infection is acquired by the hosts while viruses develop strategies to circumvent these host defenses. Forming one arm of the host defense armory are cell autonomous restriction factors like Fv1. Originally described as protecting laboratory mice from infection by murine leukemia virus (MLV), Fv1s from some wild mice have also been found to restrict non-MLV retroviruses, suggesting an important role in the protection against viruses in nature. We surveyed the Fv1 genes of wild mice trapped in Thailand and characterized their restriction activities against a panel of retroviruses. An extra copy of the Fv1 gene, named Fv7, was found on chromosome 6 of three closely related Asian species of mice: Mus caroli, M. cervicolor, and M. cookii. The presence of flanking repeats suggested it arose by LINE-mediated retroduplication within their most recent common ancestor. A high degree of natural variation was observed in both Fv1 and Fv7 and, on top of positive selection at certain residues, insertions and deletions were present that changed the length of the reading frames. These genes exhibited a range of restriction phenotypes, with activities directed against gamma-, spuma-, and lentiviruses. It seems likely, at least in the case of M. caroli, that the observed gene duplication may expand the breadth of restriction beyond the capacity of Fv1 alone and that one or more such viruses have recently driven or continue to drive the evolution of the Fv1 and Fv7 genes.


Asunto(s)
Evolución Molecular , Duplicación de Gen , Ratones/genética , Proteínas/genética , Infecciones por Retroviridae/genética , Animales , Resistencia a la Enfermedad/genética , Ratones/virología , Retroviridae/patogenicidad , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología
8.
J Virol ; 95(22): e0124421, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34468176

RESUMEN

Apolipoprotein B mRNA editing enzyme catalytic subunit 3 (APOBEC3) proteins are critical for the control of infection by retroviruses. These proteins deaminate cytidines in negative-strand DNA during reverse transcription, leading to G-to-A changes in coding strands. Uracil DNA glycosylase (UNG) is a host enzyme that excises uracils in genomic DNA, which the base excision repair machinery then repairs. Whether UNG removes uracils found in retroviral DNA after APOBEC3-mediated mutation is not clear, and whether this occurs in vivo has not been demonstrated. To determine if UNG plays a role in the repair of retroviral DNA, we used APOBEC3G (A3G) transgenic mice which we showed previously had extensive deamination of murine leukemia virus (MLV) proviruses. The A3G transgene was crossed onto an Ung and mouse Apobec3 knockout background (UNG-/-APO-/-), and the mice were infected with MLV. We found that virus infection levels were decreased in A3G UNG-/-APO-/- compared with A3G APO-/- mice. Deep sequencing of the proviruses showed that there were significantly higher levels of G-to-A mutations in proviral DNA from A3G transgenic UNG-/-APO-/- than A3G transgenic APO-/- mice, suggesting that UNG plays a role in the repair of uracil-containing proviruses. In in vitro studies, we found that cytoplasmic viral DNA deaminated by APOBEC3G was uracilated. In the absence of UNG, the uracil-containing proviruses integrated at higher levels into the genome than those made in the presence of UNG. Thus, UNG also functions in the nucleus prior to integration by nicking uracil-containing viral DNA, thereby blocking integration. These data show that UNG plays a critical role in the repair of the damage inflicted by APOBEC3 deamination of reverse-transcribed DNA. IMPORTANCE While APOBEC3-mediated mutation of retroviruses is well-established, what role the host base excision repair enzymes play in correcting these mutations is not clear. This question is especially difficult to address in vivo. Here, we use a transgenic mouse developed by our lab that expresses human APOBEC3G and also lacks the endogenous uracil DNA glycosylase (Ung) gene and show that UNG removes uracils introduced by this cytidine deaminase in MLV reverse transcripts, thereby reducing G-to-A mutations in proviruses. Furthermore, our data suggest that UNG removes uracils at two stages in infection-first, in unintegrated nuclear viral reverse-transcribed DNA, resulting in its degradation; and second, in integrated proviruses, resulting in their repair. These data suggest that retroviruses damaged by host cytidine deaminases take advantage of the host DNA repair system to overcome this damage.


Asunto(s)
Desaminasa APOBEC-3G/inmunología , ADN Viral/inmunología , Infecciones por Retroviridae , Retroviridae , Uracil-ADN Glicosidasa/inmunología , Animales , Reparación del ADN , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Células 3T3 NIH , Retroviridae/genética , Retroviridae/inmunología , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología
9.
PLoS Pathog ; 16(3): e1008340, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32226027

RESUMEN

Combination immunotherapy (CIT) is currently applied as a treatment for different cancers and is proposed as a cure strategy for chronic viral infections. Whether such therapies are efficient during an acute infection remains elusive. To address this, inhibitory receptors were blocked and regulatory T cells depleted in acutely Friend retrovirus-infected mice. CIT resulted in a dramatic expansion of cytotoxic CD4+ and CD8+ T cells and a subsequent reduction in viral loads. Despite limited viral replication, mice developed fatal immunopathology after CIT. The pathology was most severe in the gastrointestinal tract and was mediated by granzyme B producing CD4+ and CD8+ T cells. A similar post-CIT pathology during acute Influenza virus infection of mice was observed, which could be prevented by vaccination. Melanoma patients who developed immune-related adverse events under immune checkpoint CIT also presented with expanded granzyme-expressing CD4+ and CD8+ T cell populations. Our data suggest that acute infections may induce immunopathology in patients treated with CIT, and that effective measures for infection prevention should be applied.


Asunto(s)
Anticuerpos/administración & dosificación , Melanoma/inmunología , Melanoma/terapia , Infecciones por Retroviridae/inmunología , Linfocitos T Reguladores/inmunología , Infecciones Tumorales por Virus/inmunología , Animales , Antígeno B7-H1/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Virus de la Leucemia Murina de Friend/fisiología , Humanos , Inmunoterapia/efectos adversos , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Infecciones por Retroviridae/patología , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología
10.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35163254

RESUMEN

Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections that have become fixed in the human genome. While HERV genes are typically silenced in healthy somatic cells, there are numerous reports of HERV transcription and translation across a wide spectrum of cancers, while T and B cell responses against HERV proteins have been detected in cancer patients. This review systematically categorizes the published evidence on the expression of and adaptive immune response against specific HERVs in distinct cancer types. A systematic literature search was performed using Medical Search Headings (MeSH) in the PubMed/Medline database. Papers were included if they described the translational activity of HERVs. We present multiple tables that pair the protein expression of specific HERVs and cancer types with information on the quality of the evidence. We find that HERV-K is the most investigated HERV. HERV-W (syncytin-1) is the second-most investigated, while other HERVs have received less attention. From a therapeutic perspective, HERV-K and HERV-E are the only HERVs with experimental demonstration of effective targeted therapies, but unspecific approaches using antiviral and demethylating agents in combination with chemo- and immunotherapies have also been investigated.


Asunto(s)
Formación de Anticuerpos/inmunología , Retrovirus Endógenos/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Infecciones por Retroviridae/inmunología , Proteínas Virales/inmunología , Animales , Interacciones Huésped-Patógeno/inmunología , Humanos
11.
Curr Issues Mol Biol ; 43(1): 52-64, 2021 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-33946297

RESUMEN

Koala populations are currently declining and under threat from koala retrovirus (KoRV) infection both in the wild and in captivity. KoRV is assumed to cause immunosuppression and neoplastic diseases, favoring chlamydiosis in koalas. Currently, 10 KoRV subtypes have been identified, including an endogenous subtype (KoRV-A) and nine exogenous subtypes (KoRV-B to KoRV-J). The host's immune response acts as a safeguard against pathogens. Therefore, a proper understanding of the immune response mechanisms against infection is of great importance for the host's survival, as well as for the development of therapeutic and prophylactic interventions. A vaccine is an important protective as well as being a therapeutic tool against infectious disease, and several studies have shown promise for the development of an effective vaccine against KoRV. Moreover, CRISPR/Cas9-based genome editing has opened a new window for gene therapy, and it appears to be a potential therapeutic tool in many viral infections, which could also be investigated for the treatment of KoRV infection. Here, we discuss the recent advances made in the understanding of the immune response in KoRV infection, as well as the progress towards vaccine development against KoRV infection in koalas.


Asunto(s)
Citocinas/inmunología , Phascolarctidae/virología , Infecciones por Retroviridae/prevención & control , Retroviridae/inmunología , Receptores Toll-Like/inmunología , Vacunación/métodos , Animales , Citocinas/metabolismo , Phascolarctidae/inmunología , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Receptores Toll-Like/metabolismo
12.
BMC Immunol ; 22(1): 49, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34320928

RESUMEN

BACKGROUND: Targeted immunotherapy is mostly associated with cancer treatment wherein designed molecules engage signaling pathways and mutant proteins critical to the survival of the cell. One of several genetic approaches is the use of in silico methods to develop immune epitopes targeting specific antigenic regions on related mutant proteins. In a recent study we showed a functional association between the gamma retrovirus HERV-H Long Terminal Associating (HHLA1, HHLA2 and HHLA3) proteins and melanoma associated antigen of the B class proteins (MAGEB5), with a resultant decrease in expression of HLA class I and II immune variants. HLA-C and HLA-DRB5 were the main HLA class I and II Immune variants, respectively, that showed expression changes across viral samples of interest. Specific immune variants for HLA-C and HLA-DRB5 were filtered for the top ten based on their relative frequency of counts across the samples. RESULTS: Protein variants for HHLA1, HHLA2, HHLA3 and MAGEB5 were used to predict antigenic epitope peptides to immune peptide-MHC class I and II binding using artificial neural networks. For IC50 peptide scores (PS) ≥ 0.5 with a transformed binding ability between 0 and 1, the top 5 epitopes identified for all targeted genes HHLA1,2 & 3 and MAGEB5 were qualified as strong or weak binders according to the threshold. Domain analysis using NCBI Conserved Domain Database (CDD) identified HHLA2 with immunoglobulin-like domains (Ig_C1-set) and MAGEB5 with the MAGE Homology Domain (MHD). Linear regression showed a statistical correlation (P < 0.001) for HHLA2 and MAGEB5 predicted epitope peptides to HLA-C but not HLA-DRB5. The prediction model identified HLA-C variant 9 (HLA-C9, BAA08825.1 HLA-B*1511) at 1.1% as the most valuable immune target for clinical considerations. Identification of the 9-mer epitope peptide within the domain showed for HHLA2: YANRTSLFY (PS = 0.5837) and VLAYYLSSSQNTIIN (PS = 0.77) for HLA-C and HLA-DRB5, respectively and for MAGEB5, peptides: FVRLTYLEY (PS = 0.5293) and YPAHYQFLWGPRAYT (PS = 0.62) for HLA-C and HLA-DRB5, respectively. CONCLUSION: Specific immune responses to targeted epitope peptides and their prediction models, suggested co-expression and co-evolution for HHLA2 and MAGEB5 in viral related diseases. HHLA2 and MAGEB5 could be considered markers for virus related tumors and targeted therapy for oncogenic diseases.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Epítopos de Linfocito T/inmunología , Inmunoglobulinas/inmunología , Inmunoterapia/métodos , Melanoma/inmunología , Proteínas de Neoplasias/metabolismo , Infecciones por Retroviridae/inmunología , Retroviridae/fisiología , Antígenos de Neoplasias/genética , Mapeo Epitopo , Epítopos de Linfocito T/genética , Evolución Molecular , Regulación de la Expresión Génica , Estudios de Asociación Genética , Antígenos HLA/genética , Antígenos HLA-C/genética , Cadenas HLA-DRB5/genética , Humanos , Inmunoglobulinas/genética , Melanoma/genética , Mutación/genética , Proteínas de Neoplasias/genética , Polimorfismo Genético
13.
J Virol ; 94(24)2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32999021

RESUMEN

Viruses, including retroviruses, can be passed from mothers to their progeny during birth and breastfeeding. It is assumed that newborns may develop immune tolerance to milk-transmitted pathogens similarly to food antigens. I/LnJ mice are uniquely resistant to retroviruses acquired as newborns or as adults as they produce virus-neutralizing antibodies (Abs). A loss-of-function allele of H2-Ob (Ob), originally mapped within the virus infectivity controller 1 (vic1) locus, is responsible for production of antiretrovirus Abs in I/LnJ mice. Importantly, Ob-deficient and vic1 I/LnJ congenic mice on other genetic backgrounds produce antivirus Abs when infected as adults, but not as newborns. We report here that I/LnJ mice carry an additional genetic locus, virus infectivity controller 2 (vic2), that abrogates neonatal immune tolerance to retroviruses. Further genetic analysis mapped the vic2 locus to the telomeric end of chromosome 15. Identification of the vic2 gene and understanding of the related signaling pathways would make blocking of neonatal immune tolerance to retroviruses an achievable goal.IMPORTANCE This work describes a previously unknown genetic mechanism that allows neonates to respond to infections as efficiently as adults.


Asunto(s)
Tolerancia Inmunológica/genética , Infecciones por Retroviridae/inmunología , Retroviridae/inmunología , Animales , Anticuerpos Neutralizantes , Mapeo Cromosómico , Femenino , Ratones , Ratones Congénicos , Ratones Endogámicos BALB C , Ratones Endogámicos , Infecciones por Retroviridae/virología
14.
J Virol ; 94(9)2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32075930

RESUMEN

The major histocompatibility complex (MHC) class I region of humans, chimpanzees (Pan troglodytes), and bonobos (Pan paniscus) is highly similar, and orthologues of HLA-A, -B, and -C are present in both Pan species. Based on functional characteristics, the different HLA-A allotypes are classified into different supertypes. One of them, the HLA A03 supertype, is widely distributed among different human populations. All contemporary known chimpanzee and bonobo MHC class I A allotypes cluster genetically into one of the six HLA-A families, the HLA-A1/A3/A11/A30 family. We report here that the peptide-binding motif of the Patr-A*05:01 allotype, which is commonly present in a cohort of western African chimpanzees, has a strong preference for binding peptides with basic amino acids at the carboxyl terminus. This phenomenon is shared with the family members of the HLA A03 supertype. Based on the chemical similarities in the peptide-binding pocket, we inferred that the preference for binding peptides with basic amino acids at the carboxyl terminus is widely present among the human, chimpanzee, and bonobo MHC-A allotypes. Subsequent in silico peptide-binding predictions illustrated that these allotypes have the capacity to target conserved parts of the proteome of human immunodeficiency virus type 1 (HIV-1) and the simian immunodeficiency virus SIVcpz.IMPORTANCE Most experimentally infected chimpanzees seem to control an HIV-1 infection and are therefore considered to be relatively resistant to developing AIDS. Contemporary free-ranging chimpanzees may carry SIVcpz, and there is evidence for AIDS-like symptoms in these free-ranging animals, whereas SIV infections in bonobos appear to be absent. In humans, the natural control of an HIV-1 infection is strongly associated with the presence of particular HLA class I allotypes. The ancestor of the contemporary living chimpanzees and bonobos survived a selective sweep targeting the MHC class I repertoire. We have put forward a hypothesis that this may have been caused by an ancestral retroviral infection similar to SIVcpz. Characterization of the relevant MHC allotypes may contribute to understanding the shaping of their immune repertoire. The abundant presence of MHC-A allotypes that prefer peptides with basic amino acids at the C termini suggests that these molecules may contribute to the control of retroviral infections in humans, chimpanzees, and bonobos.


Asunto(s)
Genes MHC Clase I/inmunología , Antígeno HLA-A3/inmunología , Primates/inmunología , Alelos , Secuencia de Aminoácidos , Animales , VIH-1/inmunología , Antígeno HLA-A3/metabolismo , Antígenos de Histocompatibilidad , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Pan paniscus/inmunología , Pan troglodytes/inmunología , Péptidos/metabolismo , Filogenia , Unión Proteica/inmunología , Infecciones por Retroviridae/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología
15.
PLoS Pathog ; 15(9): e1008043, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31568492

RESUMEN

Immunization vectors based on cytomegalovirus (CMV) have attracted a lot of interest in recent years because of their high efficacy in the simian immunodeficiency virus (SIV) macaque model, which has been attributed to their ability to induce strong, unusually broad, and unconventionally restricted CD8+ T cell responses. To evaluate the ability of CMV-based vectors to mediate protection by other immune mechanisms, we evaluated a mouse CMV (MCMV)-based vector encoding Friend virus (FV) envelope (Env), which lacks any known CD8+ T cell epitopes, for its protective efficacy in the FV mouse model. When we immunized highly FV-susceptible mice with the Env-encoding MCMV vector (MCMV.env), we could detect high frequencies of Env-specific CD4+ T cells after a single immunization. While the control of an early FV challenge infection was highly variable, an FV infection applied later after immunization was tightly controlled by almost all immunized mice. Protection of mice correlated with their ability to mount a robust anamnestic neutralizing antibody response upon FV infection, but Env-specific CD4+ T cells also produced appreciable levels of interferon γ. Depletion and transfer experiments underlined the important role of antibodies for control of FV infection but also showed that while no Env-specific CD8+ T cells were induced by the MCMV.env vaccine, the presence of CD8+ T cells at the time of FV challenge was required. The immunity induced by MCMV.env immunization was long-lasting, but was restricted to MCMV naïve animals. Taken together, our results demonstrate a novel mode of action of a CMV-based vaccine for anti-retrovirus immunization that confers strong protection from retrovirus challenge, which is conferred by CD4+ T cells and antibodies.


Asunto(s)
Virus de la Leucemia Murina de Friend/inmunología , Muromegalovirus/inmunología , Vacunas Virales/inmunología , Traslado Adoptivo , Animales , Anticuerpos Antivirales/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Femenino , Virus de la Leucemia Murina de Friend/genética , Virus de la Leucemia Murina de Friend/patogenicidad , Productos del Gen env/genética , Productos del Gen env/inmunología , Vectores Genéticos , Inmunización , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Muromegalovirus/genética , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/prevención & control , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/genética
16.
Nat Immunol ; 10(4): 420-6, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19234474

RESUMEN

Immunoglobulin class-switch recombination (CSR) requires activation-induced cytidine deaminase (AID). Deamination of DNA by AID in transcribed switch (S) regions leads to double-stranded breaks in DNA that serve as obligatory CSR intermediates. Here we demonstrate that the catalytic and regulatory subunits of protein kinase A (PKA) were specifically recruited to S regions to promote the localized phosphorylation of AID, which led to binding of replication protein A and subsequent propagation of the CSR cascade. Accordingly, inactivation of PKA resulted in considerable disruption of CSR because of decreased AID phosphorylation and recruitment of replication protein A to S regions. We propose that PKA nucleates the formation of active AID complexes specifically on S regions to generate the high density of DNA lesions required for CSR.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/inmunología , Citidina Desaminasa/inmunología , Cambio de Clase de Inmunoglobulina , Recombinación Genética/inmunología , Proteína de Replicación A/inmunología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Roturas del ADN de Doble Cadena , Ratones , Ratones Mutantes , Fosforilación , Unión Proteica , Infecciones por Retroviridae/inmunología
17.
J Infect Dis ; 221(10): 1724-1733, 2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-31822908

RESUMEN

BACKGROUND: Zoonotic simian foamy viruses (SFVs) establish persistent infections in humans, for whom the long-term consequences for health are poorly described. In this study, we aimed to characterize blood-cell phenotypes and plasma biomarkers associated with gorilla SFV infection in humans. METHODS: We used a case-control design to compare 15 Cameroonian hunters infected with gorilla SFV (cases) to 15 controls matched for age and ethnicity. A flow cytometry-based phenotypic study and quantification of plasma immune biomarkers were carried out on blood samples from all participants. Wilcoxon signed-rank tests were used to compare cases and controls. RESULTS: Cases had a significantly higher percentage of CD8 T lymphocytes than controls (median, 17.6% vs 13.7%; P = .03) but similar levels of B, natural killer, and CD4 T lymphocytes. Cases also had a lower proportion of recent CD4 thymic emigrants (10.9% vs 18.6%, P = .05), a higher proportion of programmed death receptor 1 (PD-1) expressing memory CD4 T lymphocytes (31.7% vs 24.7%, P = .01), and higher plasma levels of the soluble CD163 scavenger receptor (0.84 vs .59 µg/mL, P = .003) than controls. CONCLUSIONS: We show, for the first time, that chronic infection with SFV is associated with T lymphocyte differentiation and monocyte activation.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/fisiología , Infecciones por Retroviridae/inmunología , Virus Espumoso de los Simios , Zoonosis , Adulto , Anciano , Animales , Estudios de Casos y Controles , Regulación de la Expresión Génica/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/metabolismo , Masculino , Persona de Mediana Edad , Primates , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo
18.
Annu Rev Genet ; 46: 677-700, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23145935

RESUMEN

Mammalian genes and genomes have been shaped by ancient and ongoing challenges from viruses. These genetic imprints can be identified via evolutionary analyses to reveal fundamental details about when (how old), where (which protein domains), and how (what are the functional consequences of adaptive changes) host-virus arms races alter the proteins involved. Just as extreme amino acid conservation can serve to identify key immutable residues in enzymes, positively selected residues point to molecular recognition interfaces between host and viral proteins that have adapted and counter-adapted in a long series of classical Red Queen conflicts. Common rules for the strategies employed by both hosts and viruses have emerged from case studies of innate immunity genes in primates. We are now poised to use these rules to transition from a retrospective view of host-virus arms races to specific predictions about which host genes face pathogen antagonism and how those genetic conflicts transform host and virus evolution.


Asunto(s)
Evolución Molecular , Interacciones Huésped-Patógeno , Desaminasas APOBEC , Inmunidad Adaptativa , Animales , Factores de Restricción Antivirales , Proteínas Portadoras/genética , Citidina Desaminasa , Citosina Desaminasa/genética , Epistasis Genética , Humanos , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteolisis , Retroviridae/genética , Retroviridae/inmunología , Retroviridae/patogenicidad , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Proteína 1 que Contiene Dominios SAM y HD , Selección Genética , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Proteínas Virales/genética , Proteínas Virales/metabolismo
19.
J Virol ; 93(20)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31341050

RESUMEN

Endogenous retroviruses (ERV) are found throughout vertebrate genomes, and failure to silence their activation can have deleterious consequences on the host. Mutation and subsequent disruption of ERV loci is therefore an indispensable component of the cell-intrinsic defenses that maintain the integrity of the host genome. Abundant in vitro and in silico evidence have revealed that APOBEC3 cytidine-deaminases, including human APOBEC3G (hA3G), can potently restrict retrotransposition; yet, in vivo data demonstrating such activity is lacking, since no replication-competent human ERV have been identified. In mice deficient for Toll-like receptor 7 (TLR7), transcribed ERV loci can recombine and generate infectious ERV. In this study, we show that ectopic expression of hA3G can prevent the emergence of replication-competent, infectious ERV in Tlr7-/- mice. Mice encode one copy of Apobec3 in their genome. ERV reactivation in Tlr7-/- mice was comparable in the presence or absence of Apobec3 In contrast, expression of a human APOBEC3G transgene abrogated emergence of infectious ERV in the Tlr7-/- background. No ERV RNA was detected in the plasma of hA3G+Apobec3-/-Tlr7-/- mice, and infectious ERV virions could not be amplified through coculture with permissive cells. These data reveal that hA3G can potently restrict active ERV in vivo and suggest that expansion of the APOBEC3 locus in primates may have helped to provide for the continued restraint of ERV in the human genome.IMPORTANCE Although APOBEC3 proteins are known to be important antiviral restriction factors in both mice and humans, their roles in the restriction of endogenous retroviruses (ERV) have been limited to in vitro studies. Here, we report that human APOBEC3G expressed as a transgene in mice prevents the emergence of infectious ERV from endogenous loci. This study reveals that APOBEC3G can powerfully restrict active retrotransposons in vivo and demonstrates how transgenic mice can be used to investigate host mechanisms that inhibit retrotransposons and reinforce genomic integrity.


Asunto(s)
Desaminasa APOBEC-3G/metabolismo , Retrovirus Endógenos/fisiología , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/virología , Replicación Viral , Animales , Dosificación de Gen , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Ratones , Ratones Noqueados , Sistemas de Lectura Abierta , Infecciones por Retroviridae/inmunología , Receptores Toll-Like/metabolismo
20.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31434732

RESUMEN

Lymph-borne Friend murine leukemia virus (FrMLV) exploits the sentinel macrophages in the draining popliteal lymph node (pLN) to infect highly permissive innate-like B-1 cells and establish infection in mice. The reason for FrMLV sensitivity of B-1 cells and their impact on viral spread is unknown. Here we demonstrate that Toll-like receptor 7 (TLR7) sensing and type I interferon (IFN-I) signaling in B-1 cells contribute to FrMLV susceptibility. FrMLV infection in B-1 cell-deficient mice (bumble; IκBNS dysfunctional) was significantly lower than that in the wild-type mice and was rescued by adoptive transfer of wild-type B-1 cells. This rescue of FrMLV infection in bumble mice was dependent on intact TLR7 sensing and IFN-I signaling within B-1 cells. Analyses of infected cell types revealed that the reduced infection in bumble mice was due predominantly to compromised virus spread to the B-2 cell population. Our data reveal how FrMLV exploits innate immune sensing and activation in the B-1 cell population for infection and subsequent spread to other lymphocytes.IMPORTANCE Viruses establish infection in hosts by targeting highly permissive cell types. The retrovirus Friend murine leukemia virus (FrMLV) infects a subtype of B cells called B-1 cells that permit robust virus replication. The reason for their susceptibility had remained unknown. We found that innate sensing of incoming virus and the ensuing type I interferon response within B-1 cells are responsible for their observed susceptibility. Our data provide insights into how retroviruses coevolved with the host to co-opt innate immune sensing pathways designed to fight virus infections for establishing infection. Understanding early events in viral spread can inform antiviral intervention strategies that prevent the colonization of a host.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Virus de la Leucemia Murina de Friend/fisiología , Glicoproteínas de Membrana/inmunología , Infecciones por Retroviridae/inmunología , Receptor Toll-Like 7/inmunología , Infecciones Tumorales por Virus/inmunología , Traslado Adoptivo , Animales , Subgrupos de Linfocitos B/virología , Inmunidad Innata , Interferón Tipo I/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Infecciones por Retroviridae/virología , Transducción de Señal/inmunología , Receptor Toll-Like 7/genética , Infecciones Tumorales por Virus/virología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA