Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.059
Filtrar
1.
J Virol ; 97(5): e0193022, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37093008

RESUMO

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.


Assuntos
Camundongos Endogâmicos , Infecções por Retroviridae , Animais , Camundongos , Imunidade , Interferon gama , Vírus da Leucemia Murina/genética , Camundongos Endogâmicos/genética , Camundongos Endogâmicos/imunologia , Infecções por Retroviridae/imunologia
2.
J Virol ; 97(1): e0179522, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36598198

RESUMO

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.


Assuntos
Desaminase APOBEC-1 , Retrovirus Endógenos , Evolução Molecular , Infecções por Retroviridae , Animais , Desaminase APOBEC-1/genética , Desaminase APOBEC-1/imunologia , Retrovirus Endógenos/classificação , Retrovirus Endógenos/genética , Retrovirus Endógenos/imunologia , Mutação , Filogenia , Infecções por Retroviridae/imunologia , Vertebrados/imunologia
3.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-35163254

RESUMO

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.


Assuntos
Formação de Anticorpos/imunologia , Retrovirus Endógenos/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Infecções por Retroviridae/imunologia , Proteínas Virais/imunologia , Animais , Interações Hospedeiro-Patógeno/imunologia , Humanos
4.
Front Immunol ; 12: 729017, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34603301

RESUMO

Piscine orthoreovirus (PRV-1) infection causes heart and skeletal muscle inflammation (HSMI) in farmed Atlantic salmon (Salmo salar). The virus is also associated with focal melanized changes in white skeletal muscle where PRV-1 infection of macrophages appears to be important. In this study, we studied the macrophage polarization into M1 (pro-inflammatory) and M2 (anti-inflammatory) phenotypes during experimentally induced HSMI. The immune response in heart with HSMI lesions was characterized by CD8+ and MHC-I expressing cells and not by polarized macrophages. Fluorescent in situ hybridization (FISH) assays revealed localization of PRV-1 in a few M1 macrophages in both heart and skeletal muscle. M2 type macrophages were widely scattered in the heart and were more abundant in heart compared to the skeletal muscle. However, the M2 macrophages did not co-stain for PRV-1. There was a strong cellular immune response to the infection in the heart compared to that of the skeletal muscle, seen as increased MHC-I expression, partly in cells also containing PRV-1 RNA, and a high number of cytotoxic CD8+ granzyme producing cells that targeted PRV-1. In skeletal muscle, MHC-I expressing cells and CD8+ cells were dispersed between myocytes, but these cells did not stain for PRV-1. Gene expression analysis by RT-qPCR complied with the FISH results and confirmed a drop in level of PRV-1 following the cell mediated immune response. Overall, the results indicated that M1 macrophages do not contribute to the initial development of HSMI. However, large numbers of M2 macrophages reside in the heart and may contribute to the subsequent fast recovery following clearance of PRV-1 infection.


Assuntos
Linfócitos T CD8-Positivos/virologia , Doenças dos Peixes/virologia , Coração/virologia , Macrófagos/virologia , Orthoreovirus/patogenicidade , Infecções por Retroviridae/virologia , Salmo salar/virologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Doenças dos Peixes/imunologia , Doenças dos Peixes/metabolismo , Interações Hospedeiro-Patógeno , Imunidade Celular , Macrófagos/imunologia , Macrófagos/metabolismo , Músculo Esquelético/imunologia , Músculo Esquelético/metabolismo , Músculo Esquelético/virologia , Miocárdio/imunologia , Miocárdio/metabolismo , Orthoreovirus/imunologia , Fenótipo , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/metabolismo , Salmo salar/imunologia , Salmo salar/metabolismo , Fatores de Tempo , Carga Viral
5.
Front Immunol ; 12: 728677, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34691035

RESUMO

Multiple Sclerosis (MS) is an inflammatory disease of the central nervous system. Sardinia, an Italian island, is one of the areas with the highest global prevalence of MS. Genetic factors have been widely explored to explain this greater prevalence among some populations; the genetic makeup of the Sardinians appears to make them more likely to develop autoimmune diseases. A strong association between MS and some infections have been reported globally. The most robust evidence indicating the role of infections is MS development concerns the Epstein-Barr virus (EBV). Anti-EBV antibodies in patients once infected by EBV are associated with the development of MS years later. These features have also been noted in Sardinian patients with MS. Many groups have found an increased expression of the Human endogenous retroviruses (HERV) family in patients with MS. A role in pathogenesis, prognosis, and prediction of treatment response has been proposed for HERV. A European multi-centre study has shown that their presence was variable among populations, ranging from 59% to 100% of patients, with higher HERV expression noted in Sardinian patients with MS. The mycobacterium avium subspecies paratuberculosis (MAP) DNA and antibodies against MAP2694 protein were found to be associated with MS in Sardinian patients. More recently, this association has also been reported in Japanese patients with MS. In this study, we analysed the role of infectious factors in Sardinian patients with MS and compared it with the findings reported in other populations.


Assuntos
Autoimunidade , Infecções por Vírus Epstein-Barr/epidemiologia , Saúde Global , Esclerose Múltipla/epidemiologia , Paratuberculose/epidemiologia , Infecções por Retroviridae/epidemiologia , Retrovirus Endógenos/imunologia , Retrovirus Endógenos/patogenicidade , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 4/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Itália/epidemiologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/microbiologia , Esclerose Múltipla/virologia , Mycobacterium avium subsp. paratuberculosis/imunologia , Mycobacterium avium subsp. paratuberculosis/patogenicidade , Paratuberculose/imunologia , Paratuberculose/microbiologia , Prognóstico , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Medição de Risco , Fatores de Risco
6.
J Virol ; 95(22): e0124421, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34468176

RESUMO

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.


Assuntos
Desaminase APOBEC-3G/imunologia , DNA Viral/imunologia , Infecções por Retroviridae , Retroviridae , Uracila-DNA Glicosidase/imunologia , Animais , Reparo do DNA , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Células NIH 3T3 , Retroviridae/genética , Retroviridae/imunologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia
7.
Nat Commun ; 12(1): 5376, 2021 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-34508086

RESUMO

Natural killer (NK) cells are important early responders against viral infections. Changes in metabolism are crucial to fuel NK cell responses, and altered metabolism is linked to NK cell dysfunction in obesity and cancer. However, very little is known about the metabolic requirements of NK cells during acute retroviral infection and their importance for antiviral immunity. Here, using the Friend retrovirus mouse model, we show that following infection NK cells increase nutrient uptake, including amino acids and iron, and reprogram their metabolic machinery by increasing glycolysis and mitochondrial metabolism. Specific deletion of the amino acid transporter Slc7a5 has only discrete effects on NK cells, but iron deficiency profoundly impaires NK cell antiviral functions, leading to increased viral loads. Our study thus shows the requirement of nutrients and metabolism for the antiviral activity of NK cells, and has important implications for viral infections associated with altered iron levels such as HIV and SARS-CoV-2.


Assuntos
Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Infecções por Retroviridae/imunologia , Animais , Medula Óssea , COVID-19 , Citocinas , HIV , Infecções por HIV , Transportador 1 de Aminoácidos Neutros Grandes/genética , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias , Retroviridae , Infecções por Retroviridae/virologia , SARS-CoV-2 , Carga Viral
8.
BMC Immunol ; 22(1): 49, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34320928

RESUMO

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.


Assuntos
Antígenos de Neoplasias/metabolismo , Epitopos de Linfócito T/imunologia , Imunoglobulinas/imunologia , Imunoterapia/métodos , Melanoma/imunologia , Proteínas de Neoplasias/metabolismo , Infecções por Retroviridae/imunologia , Retroviridae/fisiologia , Antígenos de Neoplasias/genética , Mapeamento de Epitopos , Epitopos de Linfócito T/genética , Evolução Molecular , Regulação da Expressão Gênica , Estudos de Associação Genética , Antígenos HLA/genética , Antígenos HLA-C/genética , Cadeias HLA-DRB5/genética , Humanos , Imunoglobulinas/genética , Melanoma/genética , Mutação/genética , Proteínas de Neoplasias/genética , Polimorfismo Genético
9.
Viruses ; 13(5)2021 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-34063186

RESUMO

Immunotherapy has been shown to be highly effective in some types of cancer caused by viruses. Gene therapy involves insertion or modification of a therapeutic gene, to correct for inappropriate gene products that cause/may cause diseases. Both these types of therapy have been used as alternative ways to avoid cancers caused by oncoviruses. In this review, we summarize recent studies on immunotherapy and gene therapy including the topics of oncolytic immunotherapy, immune checkpoint inhibitors, gene replacement, antisense oligonucleotides, RNA interference, clustered regularly interspaced short palindromic repeats Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-based gene editing, transcription activator-like effector nucleases (TALENs) and custom treatment for Epstein-Barr virus, human T-lymphotropic virus 1, hepatitis B virus, human papillomavirus, hepatitis C virus, herpesvirus associated with Kaposi's sarcoma, Merkel cell polyomavirus, and cytomegalovirus.


Assuntos
Terapia Genética , Imunoterapia , Infecções por Retroviridae/terapia , Retroviridae/fisiologia , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Edição de Genes , Humanos , Retroviridae/genética , Infecções por Retroviridae/genética , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia
10.
Curr Issues Mol Biol ; 43(1): 52-64, 2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-33946297

RESUMO

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.


Assuntos
Citocinas/imunologia , Phascolarctidae/virologia , Infecções por Retroviridae/prevenção & controle , Retroviridae/imunologia , Receptores Toll-Like/imunologia , Vacinação/métodos , Animais , Citocinas/metabolismo , Phascolarctidae/imunologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Receptores Toll-Like/metabolismo
11.
Viruses ; 13(3)2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33801908

RESUMO

An evolutionary arms race has been ongoing between retroviruses and their primate hosts for millions of years. Within the last century, a zoonotic transmission introduced the Human Immunodeficiency Virus (HIV-1), a retrovirus, to the human population that has claimed the lives of millions of individuals and is still infecting over a million people every year. To counteract retroviruses such as this, primates including humans have evolved an innate immune sensor for the retroviral capsid lattice known as TRIM5α. Although the molecular basis for its ability to restrict retroviruses is debated, it is currently accepted that TRIM5α forms higher-order assemblies around the incoming retroviral capsid that are not only disruptive for the virus lifecycle, but also trigger the activation of an antiviral state. More recently, it was discovered that TRIM5α restriction is broader than previously thought because it restricts not only the human retroelement LINE-1, but also the tick-borne flaviviruses, an emergent group of RNA viruses that have vastly different strategies for replication compared to retroviruses. This review focuses on the underlying mechanisms of TRIM5α-mediated restriction of retroelements and flaviviruses and how they differ from the more widely known ability of TRIM5α to restrict retroviruses.


Assuntos
Capsídeo/imunologia , Imunidade Inata , Vírus de RNA/imunologia , Vírus de RNA/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Fatores de Restrição Antivirais , Capsídeo/metabolismo , Proteínas de Transporte/genética , Flavivirus/imunologia , Flavivirus/metabolismo , Humanos , Vírus de RNA/classificação , Vírus de RNA/genética , Retroviridae/imunologia , Retroviridae/metabolismo , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/prevenção & controle , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/imunologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
12.
Viruses ; 13(3)2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33803830

RESUMO

Non-human primates (NHP) are an important source of viruses that can spillover to humans and, after adaptation, spread through the host population. Whereas HIV-1 and HTLV-1 emerged as retroviral pathogens in humans, a unique class of retroviruses called foamy viruses (FV) with zoonotic potential are occasionally detected in bushmeat hunters or zookeepers. Various FVs are endemic in numerous mammalian natural hosts, such as primates, felines, bovines, and equines, and other animals, but not in humans. They are apathogenic, and significant differences exist between the viral life cycles of FV and other retroviruses. Importantly, FVs replicate in the presence of many well-defined retroviral restriction factors such as TRIM5α, BST2 (Tetherin), MX2, and APOBEC3 (A3). While the interaction of A3s with HIV-1 is well studied, the escape mechanisms of FVs from restriction by A3 is much less explored. Here we review the current knowledge of FV biology, host restriction factors, and FV-host interactions with an emphasis on the consequences of FV regulatory protein Bet binding to A3s and outline crucial open questions for future studies.


Assuntos
Desaminases APOBEC/metabolismo , Interações entre Hospedeiro e Microrganismos , Proteínas dos Retroviridae/metabolismo , Spumavirus/genética , Spumavirus/fisiologia , Animais , Linhagem Celular , Humanos , Mutação , Primatas/virologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Proteínas dos Retroviridae/classificação , Proteínas dos Retroviridae/genética , Spumavirus/imunologia
13.
Viruses ; 13(3)2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33801276

RESUMO

The SAM and HD domain-containing protein 1 (SAMHD1) is a dNTP triphosphohydrolase that plays a crucial role for a variety of different cellular functions. Besides balancing intracellular dNTP concentrations, facilitating DNA damage repair, and dampening excessive immune responses, SAMHD1 has been shown to act as a major restriction factor against various virus species. In addition to its well-described activity against retroviruses such as HIV-1, SAMHD1 has been identified to reduce the infectivity of different DNA viruses such as the herpesviruses CMV and EBV, the poxvirus VACV, or the hepadnavirus HBV. While some viruses are efficiently restricted by SAMHD1, others have developed evasion mechanisms that antagonize the antiviral activity of SAMHD1. Within this review, we summarize the different cellular functions of SAMHD1 and highlight the countermeasures viruses have evolved to neutralize the restriction factor SAMHD1.


Assuntos
Infecções por Vírus de DNA/imunologia , Interações Hospedeiro-Patógeno/imunologia , Infecções por Retroviridae/imunologia , Proteína 1 com Domínio SAM e Domínio HD/imunologia , Vírus de DNA/imunologia , Humanos , Retroviridae/imunologia , Interferência Viral
14.
Emerg Microbes Infect ; 10(1): 964-981, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33858301

RESUMO

Antiviral monoclonal antibodies (mAbs) can generate protective immunity through Fc-FcγRs interactions. We previously showed a role for immune complexes (ICs) in the enhancement of antiviral T-cell responses through FcγR-mediated activation of dendritic cells (DCs). Here we addressed how mAb therapy in retrovirus-infected mice affects the activation of neutrophils and inflammatory monocytes, two FcγR-expressing innate effector cells rapidly recruited to sites of infection. We found that both cell-types activated in vitro by viral ICs secreted chemokines able to recruit monocytes and neutrophils themselves. Moreover, inflammatory cytokines potentiated chemokines and cytokines release by IC-activated cells and induced FcγRIV upregulation. Similarly, infection and mAb-treatment upregulated FcγRIV on neutrophils and inflammatory monocytes and enhanced their cytokines/chemokines secretion. Notably, upon antibody therapy neutrophils and inflammatory monocytes displayed distinct functional activation states and sequentially modulated the antiviral immune response by secreting Th1-type polarizing cytokines and chemokines, which occurred in a FcγRIV-dependent manner. Consistently, FcγRIV- blocking in mAb-treated, infected mice led to reduced immune protection. Our work provides new findings on the immunomodulatory role of neutrophils and monocytes in the enhancement of immune responses upon antiviral mAb therapy.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Monócitos/imunologia , Neutrófilos/imunologia , Infecções por Retroviridae/tratamento farmacológico , Animais , Anticorpos Monoclonais/imunologia , Complexo Antígeno-Anticorpo/metabolismo , Antígenos Ly/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Camundongos , Receptores de IgG/metabolismo , Infecções por Retroviridae/imunologia , Resultado do Tratamento
15.
Commun Biol ; 4(1): 318, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33750893

RESUMO

Bone morphogenetic protein (BMP) is a kind of classical multi-functional growth factor that plays a vital role in the formation and maintenance of bone, cartilage, muscle, blood vessels, and the regulation of adipogenesis and thermogenesis. However, understanding of the role of BMPs in antiviral immunity is still limited. Here we demonstrate that Bmp8a is a newly-identified positive regulator for antiviral immune responses. The bmp8a-/- zebrafish, when infected with viruses, show reduced antiviral immunity and increased viral load and mortality. We also show for the first time that Bmp8a interacts with Alk6a, which promotes the phosphorylation of Tbk1 and Irf3 through p38 MAPK pathway, and induces the production of type I interferons (IFNs) in response to viral infection. Our study uncovers a previously unrecognized role of Bmp8a in regulation of antiviral immune responses and provides a target for controlling viral infection.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Interferon Tipo I/metabolismo , Infecções por Retroviridae/virologia , Retroviridae/patogenicidade , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/virologia , Animais , Animais Geneticamente Modificados , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Técnicas de Inativação de Genes , Interações Hospedeiro-Patógeno , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/imunologia , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Retroviridae/crescimento & desenvolvimento , Retroviridae/imunologia , Infecções por Retroviridae/genética , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/metabolismo , Transdução de Sinais , Carga Viral , Replicação Viral , Peixe-Zebra/genética , Peixe-Zebra/imunologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
mBio ; 12(1)2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33531395

RESUMO

PD-1-targeted therapies have shown modest antiviral effects in preclinical models of chronic viral infection. Thus, novel therapy protocols are necessary to enhance T cell immunity and viral control to overcome T cell dysfunction and immunosuppression. Here, we demonstrate that nanoparticle-based therapeutic vaccination improved PD-1-targeted therapy during chronic infection with Friend retrovirus (FV). Prevention of inhibitory signals by blocking PD-L1 in combination with therapeutic vaccination with nanoparticles containing the microbial compound CpG and a CD8+ T cell Gag epitope peptide synergistically enhanced functional virus-specific CD8+ T cell responses and improved viral clearance. We characterized the CD8+ T cell populations that were affected by this combination therapy, demonstrating that new effector cells were generated and that exhausted CD8+ T cells were reactivated at the same time. While CD8+ T cells with high PD-1 (PD-1hi) expression turned into a large population of granzyme B-expressing CD8+ T cells after combination therapy, CXCR5-expressing follicular cytotoxic CD8+ T cells also expanded to a high degree. Thus, our study describes a very efficient approach to enhance virus control and may help us to understand the mechanisms of combination immunotherapy reactivating CD8+ T cell immunity. A better understanding of CD8+ T cell immunity during combination therapy will be important for developing efficient checkpoint therapies against chronic viral infections and cancer.IMPORTANCE Despite significant efforts, vaccines are not yet available for every infectious pathogen, and the search for a protective approach to prevent the establishment of chronic infections, i.e., with HIV, continues. Immune checkpoint therapies targeting inhibitory receptors, such as PD-1, have shown impressive results against solid tumors. However, immune checkpoint therapies have not yet been licensed to treat chronic viral infections, since a blockade of inhibitory receptors alone provides only limited benefit, as demonstrated in preclinical models of chronic viral infection. Thus, there is a high interest in the development of potent combination immunotherapies. Here, we tested whether the combination of a PD-L1 blockade and therapeutic vaccination with functionalized nanoparticles is a potent therapy during chronic Friend retrovirus infection. We demonstrate that the combination therapy induced a synergistic reinvigoration of the exhausted virus-specific CD8+ T cell immunity. Taken together, our results provide further information on how to improve PD-1-targeted therapies during chronic viral infection and cancer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vírus da Leucemia Murina de Friend/imunologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Ativação Linfocitária , Infecções por Retroviridae/terapia , Vacinação , Animais , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Receptores CXCR5/análise , Infecções por Retroviridae/imunologia
17.
Front Immunol ; 12: 809774, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35126368

RESUMO

Type I Interferons (IFNs), including numerous IFNα subtypes and IFNß, are key molecules during innate and adaptive immune responses against viral infections. These cytokines exert various non-redundant biological activities, although binding to the same receptor. Persistent viral infections are often characterized by increased IFN signatures implicating a potential role of type I IFNs in disease pathogenesis. Using the well-established Friend retrovirus (FV) mouse model, we compared the therapeutic efficacy of IFNα11 and IFNß in acute and chronic retroviral infection. We observed a strong antiviral activity of both IFNs during acute FV infection, whereas only IFNα11 and not IFNß could also control persistent FV infection. The therapeutic treatment with IFNα11 induced the expression of antiviral IFN-stimulated genes (ISG) and improved cytotoxic T cell responses. Finally, dysfunctional CD8+ T cells solely regained cytotoxicity after IFNα11 treatment. Our data provide evidence for opposing activities of type I IFNs during chronic retroviral infections. IFNß was shown to be involved in immune dysfunction in chronic infections, whereas IFNα11 had a strong antiviral potential and reactivated exhausted T cells during persistent retroviral infection. In contrast, during acute infection, both type I IFNs were able to efficiently suppress FV replication.


Assuntos
Antivirais/farmacologia , Interferon-alfa/farmacologia , Infecções por Retroviridae/tratamento farmacológico , Infecções por Retroviridae/virologia , Animais , Biomarcadores , Linhagem Celular , Citotoxicidade Imunológica/efeitos dos fármacos , Gerenciamento Clínico , Modelos Animais de Doenças , Feminino , Vírus da Leucemia Murina de Friend/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Fatores Imunológicos/farmacologia , Interferon Tipo I/farmacologia , Interferon beta/farmacologia , Vírus da Leucemia Murina/fisiologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Infecções por Retroviridae/imunologia , Resultado do Tratamento , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
18.
Am J Reprod Immunol ; 85(2): e13345, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32939907

RESUMO

Cells produce cytoplasmic vesicles to facilitate the processing and transport of RNAs, proteins, and other signaling molecules among intracellular organelles. Moreover, most cells release a range of extracellular vesicles (EVs) that mediate intercellular communication in both physiological and pathological settings. In addition to a better understanding of their biological functions, the diagnostic and therapeutic prospects of EVs, particularly the nano-sized small EVs (sEVs, exosomes), are currently being rigorously pursued. While EVs and viruses such as retroviruses might have evolved independently, they share a number of similar characteristics, including biogenesis pathways, size distribution, cargo, and cell-targeting mechanisms. The interplay of EVs with viruses has profound effects on viral replication and infectivity. Our research indicates that sEVs, produced by primary human trophoblasts, can endow other non-placental cell types with antiviral response. Better insights into the interaction of EVs with viruses may illuminate new ways to attenuate viral infections during pregnancy, and perhaps develop new antiviral therapeutics to protect the feto-placental unit during critical times of human development.


Assuntos
Vesículas Extracelulares/imunologia , Placenta/imunologia , Gravidez/imunologia , Infecções por Retroviridae/imunologia , Retroviridae/fisiologia , Trofoblastos/imunologia , Feminino , Humanos , Nanoestruturas , Especificidade de Órgãos , Virulência , Replicação Viral
19.
Annu Rev Anim Biosci ; 9: 225-248, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33290087

RESUMO

Endogenous retroviruses (ERVs) serve as markers of ancient viral infections and provide invaluable insight into host and viral evolution. ERVs have been exapted to assist in performing basic biological functions, including placentation, immune modulation, and oncogenesis. A subset of ERVs share high nucleotide similarity to circulating horizontally transmitted exogenous retrovirus (XRV) progenitors. In these cases, ERV-XRV interactions have been documented and include (a) recombination to result in ERV-XRV chimeras, (b) ERV induction of immune self-tolerance to XRV antigens, (c) ERV antigen interference with XRV receptor binding, and (d) interactions resulting in both enhancement and restriction of XRV infections. Whereas the mechanisms governing recombination and immune self-tolerance have been partially determined, enhancement and restriction of XRV infection are virus specific and only partially understood. This review summarizes interactions between six unique ERV-XRV pairs, highlighting important ERV biological functions and potential evolutionary histories in vertebrate hosts.


Assuntos
Retrovirus Endógenos/genética , Infecções por Retroviridae/virologia , Animais , Antígenos Virais , Aves/genética , Aves/virologia , Evolução Molecular , Mamíferos/genética , Mamíferos/virologia , Retroviridae/fisiologia , Infecções por Retroviridae/imunologia
20.
Virol J ; 17(1): 168, 2020 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-33129323

RESUMO

Koala retrovirus (KoRV) is believed to be in an active state of endogenization into the koala genome. KoRV is present as both an endogenous and exogenous infection in all koalas in northern Australia. KoRV has been linked to koala pathologies including neoplasia and increased susceptibility to Chlamydia. A KoRV vaccine recently trialled in 10 northern koalas improved antibody response and reduced viral load. This communication reports the expression of key immune genes underlining the innate and adaptive immune response to vaccination in these northern koalas. The results showed that prior to vaccination, IL-8 was expressed at the highest levels, with at least 200-fold greater expression compared to other cytokines, while CD8 mRNA expression was significantly higher than CD4 mRNA expression level. Interferon-γ was up-regulated at both 4- and 8-weeks post-vaccination while IL-8 was down-regulated at 8-weeks post-vaccination.


Assuntos
Citocinas/genética , Interferon gama/genética , Phascolarctidae/virologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/veterinária , Retroviridae/imunologia , Vacinas Virais/imunologia , Animais , Formação de Anticorpos , Austrália , Estudos de Coortes , Citocinas/imunologia , Retrovirus Endógenos/genética , Retrovirus Endógenos/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Phascolarctidae/imunologia , Retroviridae/genética , Infecções por Retroviridae/prevenção & controle , Regulação para Cima , Vacinas Virais/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...