Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 410
Filtrar
1.
Commun Biol ; 7(1): 350, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38514810

RESUMO

Koalas (Phascolarctos cinereus) have experienced a history of retroviral epidemics leaving their trace as heritable endogenous retroviruses (ERVs) in their genomes. A recently identified ERV lineage, named phaCin-ß, shows a pattern of recent, possibly current, activity with high insertional polymorphism in the population. Here, we investigate geographic patterns of three focal ERV lineages of increasing estimated ages, from the koala retrovirus (KoRV) to phaCin-ß and to phaCin-ß-like, using the whole-genome sequencing of 430 koalas from the Koala Genome Survey. Thousands of ERV loci were found across the population, with contrasting patterns of polymorphism. Northern individuals had thousands of KoRV integrations and hundreds of phaCin-ß ERVs. In contrast, southern individuals had higher phaCin-ß frequencies, possibly reflecting more recent activity and a founder effect. Overall, our findings suggest high ERV burden in koalas, reflecting historic retrovirus-host interactions. Importantly, the ERV catalogue supplies improved markers for conservation genetics in this endangered species.


Assuntos
Retrovirus Endógenos , Gammaretrovirus , Phascolarctidae , Infecções por Retroviridae , Humanos , Animais , Retrovirus Endógenos/genética , Phascolarctidae/genética , Infecções por Retroviridae/genética , Gammaretrovirus/genética , Sequenciamento Completo do Genoma
2.
Nat Commun ; 15(1): 2154, 2024 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-38461177

RESUMO

Five to ten percent of mammalian genomes is occupied by multiple clades of endogenous retroviruses (ERVs), that may count thousands of members. New ERV clades arise by retroviral infection of the germline followed by expansion by reinfection and/or retrotransposition. ERV mobilization is a source of deleterious variation, driving the emergence of ERV silencing mechanisms, leaving "DNA fossils". Here we show that the ERVK[2-1-LTR] clade is still active in the bovine and a source of disease-causing alleles. We develop a method to measure the rate of ERVK[2-1-LTR] mobilization, finding an average of 1 per ~150 sperm cells, with >10-fold difference between animals. We perform a genome-wide association study and identify eight loci affecting ERVK[2-1-LTR] mobilization. We provide evidence that polymorphic ERVK[2-1-LTR] elements in four of these loci cause the association. We generate a catalogue of full length ERVK[2-1-LTR] elements, and show that it comprises 15% of C-type autonomous elements, and 85% of D-type non-autonomous elements lacking functional genes. We show that >25% of the variance of mobilization rate is determined by the number of C-type elements, yet that de novo insertions are dominated by D-type elements. We propose that D-type elements act as parasite-of-parasite gene drives that may contribute to the observed demise of ERV elements.


Assuntos
Retrovirus Endógenos , Infecções por Retroviridae , Animais , Bovinos , Masculino , Retrovirus Endógenos/genética , Estudo de Associação Genômica Ampla , Sêmen , Espermatozoides , Infecções por Retroviridae/genética , Mamíferos/genética
3.
N Z Vet J ; 71(1): 1-7, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36178295

RESUMO

Feline leukaemia virus (FeLV) is a retrovirus that infects domestic and wild cats around the world. FeLV infection is associated with the development of neoplasms, bone marrow disorders and immunosuppression. Viral subgroups arise from mutations in the FeLV genome or from recombination of FeLV with ancestral endogenous retroviruses in the cat genome. The retroviral endogenisation process has allowed generation of a diversity of endogenous viruses, both functional and defective. These elements may be part of the normal functioning of the feline genome and may also interact with FeLV to form recombinant FeLV subgroups, enhance pathogenicity of viral subgroups, or inhibit and/or regulate other retroviral infections. Recombination of the env gene occurs most frequently and appears to be the most significant in terms of both the quantity and diversification of pathogenic effects in the viral population, as well as affecting cell tropism and types of disease that occur in infected cats. This review focuses on available information regarding genetic diversity, pathogenesis and diagnosis of FeLV as a result of the interaction between endogenous and exogenous viruses.


Assuntos
Doenças do Gato , Retrovirus Endógenos , Leucemia Felina , Infecções por Retroviridae , Gatos , Animais , Vírus da Leucemia Felina/genética , Vírus da Leucemia Felina/metabolismo , Retrovirus Endógenos/genética , Leucemia Felina/genética , Genes env , Infecções por Retroviridae/veterinária , Infecções por Retroviridae/genética , Doenças do Gato/genética
4.
Genome Biol Evol ; 15(1)2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36542479

RESUMO

Koala populations show marked differences in inbreeding levels and in the presence or absence of the endogenous Koala retrovirus (KoRV). These genetic differences among populations may lead to severe disease impacts threatening koala population viability. In addition, the recent colonization of the koala genome by KoRV provides a unique opportunity to study the process of retroviral adaptation to vertebrate genomes and the impact this has on speciation, genome structure, and function. The genome build described here is from an animal from the bottlenecked Southern population free of endogenous and exogenous KoRV. It provides a more contiguous genome build than the previous koala reference derived from an animal from a more outbred Northern population and is the first koala genome from a KoRV polymerase-free animal.


Assuntos
Retrovirus Endógenos , Gammaretrovirus , Phascolarctidae , Infecções por Retroviridae , Animais , Phascolarctidae/genética , Austrália/epidemiologia , Infecções por Retroviridae/epidemiologia , Infecções por Retroviridae/genética , Retroviridae/genética , Gammaretrovirus/genética
5.
Front Immunol ; 13: 1057791, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36518758

RESUMO

Human Endogenous Retroviruses (HERVs) are derived from ancient exogenous retroviral infections that have infected our ancestors' germline cells, underwent endogenization process, and were passed throughout the generations by retrotransposition and hereditary transmission. HERVs comprise 8% of the human genome and are critical for several physiological activities. Yet, HERVs reactivation is involved in pathological process as cancer and autoimmune diseases. In this review, we summarize the multiple aspects of HERVs' role within the human genome, as well as virological and molecular aspects, and their fusogenic property. We also discuss possibilities of how the HERVs are possibly transactivated and participate in modulating the inflammatory response in health conditions. An update on their role in several autoimmune, inflammatory, and aging-related diseases is also presented.


Assuntos
Doenças Autoimunes , Retrovirus Endógenos , Neoplasias , Infecções por Retroviridae , Humanos , Retrovirus Endógenos/genética , Infecções por Retroviridae/genética , Neoplasias/genética , Genoma Humano , Doenças Autoimunes/genética
6.
Mol Cell ; 82(20): 3763-3768, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36270247

RESUMO

Germ cells are subject to exogenous retrovirus infections occasionally resulting in the genomic integration of retroviral gene sequences. These endogenized retroviruses (ERVs) are found throughout mammalian genomes. Initially thought to be inert, it is now appreciated that ERVs have often been co-opted for complex physiological processes. However, unregulated ERV transposition and expression are a threat to cellular fitness and genomic integrity, and so mammalian cells must control ERVs through pre- and post-transcriptional mechanisms. Here, we provide a field guide to the molecular machinery that identifies and silences ERVs.


Assuntos
Retrovirus Endógenos , Infecções por Retroviridae , Animais , Retrovirus Endógenos/genética , Infecções por Retroviridae/genética , Genômica , Mamíferos/genética
7.
Int J Mol Sci ; 23(17)2022 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-36077510

RESUMO

Endogenous retroviruses (ERVs), deriving from exogenous retroviral infections of germ line cells occurred millions of years ago, represent ~8% of human genome. Most ERVs are highly inactivated because of the accumulation of mutations, insertions, deletions, and/or truncations. However, it is becoming increasingly apparent that ERVs influence host biology through genetic and epigenetic mechanisms under particular physiological and pathological conditions, which provide both beneficial and deleterious effects for the host. For instance, certain ERVs expression is essential for human embryonic development. Whereas abnormal activation of ERVs was found to be involved in numbers of human diseases, such as cancer and neurodegenerative diseases. Therefore, understanding the mechanisms of regulation of ERVs would provide insights into the role of ERVs in health and diseases. Here, we provide an overview of mechanisms of transcriptional regulation of ERVs and their dysregulation in human diseases.


Assuntos
Retrovirus Endógenos , Infecções por Retroviridae , Retrovirus Endógenos/genética , Epigênese Genética , Genoma Humano , Humanos , Infecções por Retroviridae/genética
8.
Proc Natl Acad Sci U S A ; 119(26): e2114441119, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35749360

RESUMO

Host genetic resistance to viral infection controls the pathogenicity and epidemic dynamics of infectious diseases. Refrex-1 is a restriction factor against feline leukemia virus subgroup D (FeLV-D) and an endogenous retrovirus (ERV) in domestic cats (ERV-DC). Refrex-1 is encoded by a subset of ERV-DC loci with truncated envelope genes and secreted from cells as a soluble protein. Here, we identified the copper transporter CTR1 as the entry receptor for FeLV-D and genotype I ERV-DCs. We also identified CTR1 as a receptor for primate ERVs from crab-eating macaques and rhesus macaques, which were found in a search of intact envelope genes capable of forming infectious viruses. Refrex-1 counteracted infection by FeLV-D and ERV-DCs via competition for the entry receptor CTR1; the antiviral effects extended to primate ERVs with CTR1-dependent entry. Furthermore, truncated ERV envelope genes found in chimpanzee, bonobo, gorilla, crab-eating macaque, and rhesus macaque genomes could also block infection by feline and primate retroviruses. Genetic analyses showed that these ERV envelope genes were acquired in a species- or genus-specific manner during host evolution. These results indicated that soluble envelope proteins could suppress retroviral infection across species boundaries, suggesting that they function to control retroviral spread. Our findings revealed that several mammalian species acquired antiviral machinery from various ancient retroviruses, leading to convergent evolution for host defense.


Assuntos
Transportador de Cobre 1 , Genes env , Vírus da Leucemia Felina , Leucemia Felina , Infecções por Retroviridae , Animais , Gatos , Transportador de Cobre 1/genética , Evolução Molecular , Interações Hospedeiro-Patógeno , Vírus da Leucemia Felina/fisiologia , Leucemia Felina/genética , Leucemia Felina/virologia , Macaca mulatta , Infecções por Retroviridae/genética , Infecções por Retroviridae/virologia
9.
Dis Model Mech ; 15(4)2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35142349

RESUMO

Endogenous retroviruses (ERVs) are fossils left in our genome from retrovirus infections of the past. Their sequences are part of every vertebrate genome and their random integrations are thought to have contributed to evolution. Although ERVs are mainly silenced by the host genome, they have been found to be activated in multiple disease states, such as auto-inflammatory disorders and neurological diseases. However, the numerous copies in mammalian genomes and the lack of tools to study them make defining their role in health and diseases challenging. In this study, we identified eight copies of the zebrafish endogenous retrovirus zferv. We created and characterised the first in vivo ERV reporter line in any species. Using a combination of live imaging, flow cytometry and single-cell RNA sequencing, we mapped zferv expression to early T cells and neurons. Thus, this new tool identified tissues expressing ERV in zebrafish, highlighting a potential role of ERV during brain development and strengthening the hypothesis that ERV play a role in immunity and neurological diseases. This transgenic line is therefore a suitable tool to study the function of ERV in health and diseases.


Assuntos
Retrovirus Endógenos , Infecções por Retroviridae , Animais , Animais Geneticamente Modificados , Retrovirus Endógenos/genética , Mamíferos , Neurônios , Infecções por Retroviridae/genética , Peixe-Zebra/genética
10.
Virus Genes ; 58(1): 70-74, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34787790

RESUMO

Endogenous retroviruses (ERVs) are genomic elements derived from retroviral infections in ancestral germ lines. Most ERVs are inactivated by genetic or epigenetic mechanisms, such as DNA methylation. RD-114-virus-related sequence (RDRS) C2a is a feline endogenous retrovirus present in all domestic cats; however, its expression and function are not clearly known. DNA methylation at CpG dinucleotides is a hallmark of silenced ERVs. This study aimed to investigate whether long terminal repeats (LTRs) of RDRS C2a function as a gene regulatory region. The DNA methylation status of RDRS C2a was examined by bisulfite sequencing, and CpG sites in 5' LTR of RDRS C2a were found hypomethylated, whereas those in 3' LTR were hypermethylated in feline cells. Several transcription factor-binding sites were identified in LTRs of RDRS C2a. Luciferase assay suggested that 5' LTR of RDRS C2a exhibited strong transcriptional activity, which was suppressed by in vitro DNA methylation. The study indicates that 5' LTR of RDRS C2a possibly functions as a promoter for itself or neighboring genes.


Assuntos
Retrovirus Endógenos , Infecções por Retroviridae , Animais , Gatos , Metilação de DNA/genética , Retrovirus Endógenos/genética , Regiões Promotoras Genéticas , Infecções por Retroviridae/genética , Sequências Repetidas Terminais/genética
11.
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
12.
Viruses ; 13(2)2021 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-33672541

RESUMO

Viruses are obligate parasites that rely on host cellular factors to replicate and spread. The endosomal sorting complexes required for transport (ESCRT) system, which is classically associated with sorting and downgrading surface proteins, is one of the host machineries hijacked by viruses across diverse families. Knowledge gained from research into ESCRT and viruses has, in turn, greatly advanced our understanding of many other cellular functions in which the ESCRT pathway is involved, e.g., cytokinesis. This review highlights the interplay between the ESCRT pathway and the viral factors of enveloped viruses with a special emphasis on retroviruses.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Infecções por Retroviridae/metabolismo , Infecções por Retroviridae/virologia , Retroviridae/fisiologia , Animais , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Humanos , Transporte Proteico , Retroviridae/genética , Infecções por Retroviridae/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
13.
Viruses ; 13(2)2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33673243

RESUMO

In normal cells APOBEC3 (A3A-A3H) enzymes as part of the innate immune system deaminate cytosine to uracil on single-stranded DNA (ssDNA) to scramble DNA in order to give protection against a range of exogenous retroviruses, DNA-based parasites, and endogenous retroelements. However, some viruses and cancer cells use these enzymes, especially A3A and A3B, to escape the adaptive immune response and thereby lead to the evolution of drug resistance. We have synthesized first-in-class inhibitors featuring modified ssDNA. We present models based on small-angle X-ray scattering (SAXS) data that (1) confirm that the mode of binding of inhibitor to an active A3B C-terminal domain construct in the solution state is the same as the mode of binding substrate to inactive mutants of A3A and A3B revealed in X-ray crystal structures and (2) give insight into the disulfide-linked inactive dimer formed under the oxidizing conditions of purification.


Assuntos
Citidina Desaminase/química , Citidina Desaminase/metabolismo , DNA de Cadeia Simples/química , Antígenos de Histocompatibilidade Menor/química , Antígenos de Histocompatibilidade Menor/metabolismo , RNA Viral/química , Infecções por Retroviridae/enzimologia , Retroviridae/genética , Domínio Catalítico , Citidina Desaminase/genética , DNA de Cadeia Simples/metabolismo , Dimerização , Humanos , Antígenos de Histocompatibilidade Menor/genética , Mutação , RNA Viral/genética , RNA Viral/metabolismo , Retroviridae/metabolismo , Infecções por Retroviridae/genética , Infecções por Retroviridae/virologia , Espalhamento a Baixo Ângulo
14.
Cancer Sci ; 112(6): 2081-2088, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33728716

RESUMO

Cell identity is controlled by regulatory elements, such as promoters, enhancers, and insulators, within the genome. These regulatory elements interact in the nucleus and form tissue-specific chromatin structures. Dysregulation of these elements and their interactions can lead to loss of cell identity and promote the development of diseases such as cancer. Tumor cells acquire aberrantly activated enhancers at oncogenic driver genes through various mechanisms. Small genomic changes such as mutations, insertions, and amplifications can form aberrant enhancers. Genomic rearrangements at the chromosomal level, including translocations and inversions, are also often observed in cancers. These rearrangements can result in repositioning of enhancers to locations near tumor-type-specific oncogenes. Chromatin structural changes caused by genomic or epigenomic changes lead to mis-interaction between enhancers and proto-oncogenes, ultimately contributing to tumorigenesis through activation of oncogenic signals. Additional epigenomic mechanisms can also cause aberrant enhancer activation, including those associated with overexpression of oncogenic transcription factors and the mutation of transcriptional cofactors. Exogenous viral DNA can also lead to enhancer aberrations. Here, we review the mechanisms underlying aberrant oncogene activation through enhancer activation and rewiring, both of which are caused by genomic or epigenomic alterations in non-coding regions.


Assuntos
Elementos Facilitadores Genéticos/genética , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias/genética , Cromatina/genética , Cromatina/metabolismo , Epigênese Genética , Humanos , Mutação , Oncogenes/genética , Infecções por Retroviridae/genética , Infecções por Retroviridae/virologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Translocação Genética
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.
Nat Commun ; 12(1): 1316, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33637755

RESUMO

Repeated retroviral infections of vertebrate germlines have made endogenous retroviruses ubiquitous features of mammalian genomes. However, millions of years of evolution obscure many of the immediate repercussions of retroviral endogenisation on host health. Here we examine retroviral endogenisation during its earliest stages in the koala (Phascolarctos cinereus), a species undergoing germline invasion by koala retrovirus (KoRV) and affected by high cancer prevalence. We characterise KoRV integration sites (IS) in tumour and healthy tissues from 10 koalas, detecting 1002 unique IS, with hotspots of integration occurring in the vicinity of known cancer genes. We find that tumours accumulate novel IS, with proximate genes over-represented for cancer associations. We detect dysregulation of genes containing IS and identify a highly-expressed transduced oncogene. Our data provide insights into the tremendous mutational load suffered by the host during active retroviral germline invasion, a process repeatedly experienced and overcome during the evolution of vertebrate lineages.


Assuntos
Células Germinativas , Neoplasias/genética , Infecções por Retroviridae/genética , Retroviridae/genética , Animais , Retrovirus Endógenos , Evolução Molecular , Gammaretrovirus/genética , Regulação Neoplásica da Expressão Gênica , Genoma Humano , Humanos , Neoplasias/virologia , Phascolarctidae/genética , Phascolarctidae/virologia , Proteínas Repressoras/genética , Infecções por Retroviridae/virologia , Proteína bcl-X/genética
17.
J Virol ; 94(18)2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32641479

RESUMO

Apolipoprotein B editing enzyme, catalytic polypeptide 3 (APOBEC3) family members are cytidine deaminases that play important roles in intrinsic responses to retrovirus infection. Complex retroviruses like human immunodeficiency virus type 1 (HIV-1) encode the viral infectivity factor (Vif) protein to counteract APOBEC3 proteins. Vif induces degradation of APOBEC3G and other APOBEC3 proteins and thereby prevents their packaging into virions. It is not known if murine leukemia virus (MLV) encodes a Vif-like protein. Here, we show that the MLV P50 protein, produced from an alternatively spliced gag RNA, interacts with the C terminus of mouse APOBEC3 and prevents its packaging without causing its degradation. By infecting APOBEC3 knockout (KO) and wild-type (WT) mice with Friend or Moloney MLV P50-deficient viruses, we found that APOBEC3 restricts the mutant viruses more than WT viruses in vivo Replication of P50-mutant viruses in an APOBEC3-expressing stable cell line was also much slower than that of WT viruses, and overexpressing P50 in this cell line enhanced mutant virus replication. Thus, MLV encodes a protein, P50, that overcomes APOBEC3 restriction by preventing its packaging into virions.IMPORTANCE MLV has existed in mice for at least a million years, in spite of the existence of host restriction factors that block infection. Although MLV is considered a simple retrovirus compared to lentiviruses, it does encode proteins generated from alternatively spliced RNAs. Here, we show that P50, generated from an alternatively spliced RNA encoded in gag, counteracts APOBEC3 by blocking its packaging. MLV also encodes a protein, glycoGag, that increases capsid stability and limits APOBEC3 access to the reverse transcription complex (RTC). Thus, MLV has evolved multiple means of preventing APOBEC3 from blocking infection, explaining its survival as an infectious pathogen in mice.


Assuntos
Citidina Desaminase/genética , Regulação Viral da Expressão Gênica , Produtos do Gene gag/genética , Leucemia Experimental/genética , Vírus da Leucemia Murina de Moloney/genética , Infecções por Retroviridae/genética , Infecções Tumorais por Vírus/genética , Processamento Alternativo , Animais , Capsídeo/metabolismo , Citidina Desaminase/deficiência , Produtos do Gene gag/metabolismo , Células HEK293 , Interações Hospedeiro-Patógeno/genética , Humanos , Leucemia Experimental/metabolismo , Leucemia Experimental/virologia , Camundongos , Camundongos Knockout , Vírus da Leucemia Murina de Moloney/metabolismo , Vírus da Leucemia Murina de Moloney/patogenicidade , Células NIH 3T3 , Infecções por Retroviridae/metabolismo , Infecções por Retroviridae/virologia , Transdução de Sinais , Infecções Tumorais por Vírus/metabolismo , Infecções Tumorais por Vírus/virologia , Vírion/genética , Vírion/metabolismo , Vírion/patogenicidade , Replicação Viral
18.
PLoS Genet ; 16(6): e1008471, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32525879

RESUMO

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.


Assuntos
Evolução Molecular , Duplicação Gênica , Camundongos/genética , Proteínas/genética , Infecções por Retroviridae/genética , Animais , Resistência à Doença/genética , Camundongos/virologia , Retroviridae/patogenicidade , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia
19.
J Virol ; 94(11)2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32188730

RESUMO

Koala retrovirus (KoRV) is of an interest to virologists due to its currently active endogenization into the koala (Phascolarctos cinereus) genome. Although KoRV has frequently been isolated in wild and captive koala populations, its pathogenesis and transmission remain to be fully characterized, and most previous research has concentrated on adult koalas rather than on joeys. Here, we characterized KoRV isolates obtained from a deceased male joey and its parents (animals reared in a Japanese zoo) to investigate KoRV transmission mode and pathogenesis. We sequenced the KoRV long terminal repeat (LTR) and envelope genes isolated from the joey and its parents and found KoRV-A and KoRV-C in genomic DNA from both the parents and the joey. Notably, both parents were also positive for KoRV-B, whereas the joey was KoRV-B negative, further confirming that KoRV-B is an exogenous strain. The KoRV LTR sequence of the joey was considerably closer to that of its sire than its dam. For further characterization, total KoRV, KoRV-A, KoRV-B, and KoRV-C proviral loads were quantified in peripheral blood mononuclear cells from the parents and in blood samples from the joey. Total KoRV, KoRV-A, and KoRV-C proviral loads were also quantified for different tissues (bone, liver, kidney, lung, spleen, heart, and muscle) from the joey, revealing differences suggestive of a distinct tissue tropism (highest total KoRV proviral load in the spleen and lowest in bone). The amount of KoRV-C in the parents was less than that in the joey. Our findings contribute to an improved understanding of KoRV pathogenesis and transmission mode and highlight useful areas for future research.IMPORTANCE KoRV is unique among retroviruses in that one strain (KoRV-A) is undergoing endogenization, whereas the other main subtype (KoRV-B) and another subtype (KoRV-C) are reportedly exogenous strains. Its transmission and pathogenesis are of interest in the study of retroviruses and are crucial for any conservation strategy geared toward koala health. This study provides new evidence on the modes of KoRV transmission from parent koalas to their joey. We found vertical transmission of KoRV-A, confirming its endogenization, but with closer conservation between the joey and its sire than its dam (previous reports on joeys are rare but have postulated dam-to-joey vertical transmission). This is also the first report of a KoRV-B-negative joey from KoRV-B-positive parents, contrasting with the few previous reports of 100% transmission of KoRV-B from dams to joeys. Thus, the results in this study give some novel insights for the transmission mode of KoRV.


Assuntos
Evolução Molecular , Phascolarctidae/virologia , Infecções por Retroviridae , Retroviridae , Sequências Repetidas Terminais , Animais , Feminino , Japão , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Masculino , Retroviridae/genética , Retroviridae/metabolismo , Infecções por Retroviridae/genética , Infecções por Retroviridae/metabolismo , Infecções por Retroviridae/transmissão , Infecções por Retroviridae/veterinária
20.
Vet Microbiol ; 242: 108610, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32122614

RESUMO

Reticuloendotheliosis virus (REV) infection of multiple avian species can lead to a number of diseases such as runting syndrome, immunosuppression and oncogenesis, causing major economic losses. MicroRNAs play important roles in post-transcriptional regulation, effectively inhibiting protein synthesis, and participating in many biological processes in cells, including proliferation, differentiation, apoptosis, lipometabolism, virus infection and replication, and tumorigenesis. Based on our previous high-throughput sequencing results, we explore the regulatory mechanisms of microRNA-155(miR-155) in chicken embryo fibroblasts (CEFs) in response to REV infection. Our results revealed expression of miR-155 in CEFs after REV infection upregulated in a time- and dose-dependent manner, indicating miR-155 plays a role in REV infection in CEFs indeed. After transfected with miR-155-mimic and miR-155-inhibitor, we found overexpression of miR-155 targeted caspase-6 and FOXO3a to inhibit apoptosis and accelerate cell cycle, thus improving viability of REV-infected CEFs. This result also verified the protective role of miR-155 in the viability of CEFs in the presence of REV. Knockdown of miR-155 also supported these above conclusions. Our findings uncover a new mechanism of REV pathogenesis in CEFs, and also provide a theoretical basis for uncovering new effective treatment and prevention methods for RE based on miR-155.


Assuntos
Fibroblastos/virologia , Regulação da Expressão Gênica , Interações entre Hospedeiro e Microrganismos/genética , MicroRNAs/genética , Vírus da Reticuloendoteliose/fisiologia , Infecções por Retroviridae/veterinária , Animais , Apoptose , Caspase 6/genética , Embrião de Galinha/virologia , Técnicas de Silenciamento de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Infecções por Retroviridae/genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...