Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.935
Filtrar
1.
Mem Inst Oswaldo Cruz ; 119: e240071, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39292108

RESUMO

BACKGROUND: Human immunodeficiency virus (HIV)-1 infection can activate the expression of human endogenous retroviruses (HERVs), particularly HERV-K (HML-2). HIV controllers (HICs) are rare people living with HIV (PLWHs) who naturally control HIV-1 replication and overexpress some cellular restriction factors that negatively regulate the LTR-driven transcription of HIV-1 proviruses. OBJECTIVES: To understand the ability of HICs to control the expression of endogenous retroviruses. METHODS: We measured endogenous retrovirus type K6 (ERVK-6) RNA expression in peripheral blood mononuclear cells (PBMCs) of HICs (n = 23), antiretroviral (ART)-suppressed subjects (n = 8), and HIV-1-negative (NEG) individuals (n = 10) and correlated the transcript expression of ERVK-6 with multiple HIV-1 cellular restriction factors. FINDINGS: Our study revealed that ERVK-6 RNA expression in PBMCs from HICs was significantly downregulated compared with that in both the ART and NEG control groups. Moreover, we detected that ERVK-6 RNA levels in PBMCs across all groups were negatively correlated with the expression levels of p21 and MCPIP1, two cellular restriction factors that limit the activation of macrophages and T cells by downregulating the activity of NF-kB. MAIN CONCLUSIONS: These findings support the hypothesis that HICs activate innate antiviral mechanisms that may simultaneously downregulate the transcription of both exogenous (HIV-1) and endogenous (ERVK-6) retroviruses. Future studies with larger cohorts should be performed to confirm this hypothesis and to explore the role of p21 and MCPIP1 in regulating HERV-K expression in physiological and pathological conditions.


Assuntos
Retrovirus Endógenos , Infecções por HIV , HIV-1 , RNA Viral , Ribonucleases , Humanos , HIV-1/genética , HIV-1/imunologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , Infecções por HIV/genética , Retrovirus Endógenos/genética , Retrovirus Endógenos/imunologia , Masculino , Adulto , Feminino , Ribonucleases/genética , Ribonucleases/metabolismo , RNA Viral/genética , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Pessoa de Meia-Idade , Inibidor de Quinase Dependente de Ciclina p21/genética , Fatores de Transcrição/genética , Imunidade Inata/genética , Estudos de Casos e Controles , Replicação Viral/genética
2.
Cells ; 13(17)2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39273061

RESUMO

Zika virus (ZIKV) is an arbovirus with maternal, sexual, and TORCH-related transmission capabilities. After 2015, Brazil had the highest number of ZIVK-infected pregnant women who lost their babies or delivered them with Congenital ZIKV Syndrome (CZS). ZIKV triggers an immune defense in the placenta. This immune response counts with the participation of interleukins and transcription factors. Additionally, it has the potential involvement of human endogenous retroviruses (HERVS). Interleukins are immune response regulators that aid immune tolerance and support syncytial structure development in the placenta, where syncytin receptors facilitate vital cell-to-cell fusion events. HERVs are remnants of ancient viral infections that integrate into the genome and produce syncytin proteins crucial for placental development. Since ZIKV can infect trophoblast cells, we analyzed the relationship between ZIKV infection, HERV, interleukin, and transcription factor modulations in the placenta. To investigate the impact of ZIKV on trophoblast cells, we examined two cell types (BeWo and HTR8) infected with ZIKV-MR766 (African) and ZIKV-IEC-Paraíba (Asian-Brazilian) using Taqman and RT2 Profiler PCR Array assays. Our results indicate that early ZIKV infection (24-72 h) does not induce differential interleukins, transcription factors, and HERV expression. However, we show that the expression of a few of these host defense genes appears to be linked independently of ZIKV infection. Future studies involving additional trophoblastic cell lineages and extended infection timelines will illuminate the dynamic interplay between ZIKV, HERVs, interleukins, and transcription factors in the placenta.


Assuntos
Retrovirus Endógenos , Interleucinas , Fatores de Transcrição , Trofoblastos , Infecção por Zika virus , Zika virus , Humanos , Trofoblastos/virologia , Trofoblastos/metabolismo , Feminino , Infecção por Zika virus/virologia , Infecção por Zika virus/genética , Retrovirus Endógenos/genética , Gravidez , Interleucinas/genética , Interleucinas/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Placenta/virologia , Placenta/metabolismo , Linhagem Celular
3.
J Transl Med ; 22(1): 756, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39135093

RESUMO

BACKGROUND: Decoding human genomic sequences requires comprehensive analysis of DNA sequence functionality. Through computational and experimental approaches, researchers have studied the genotype-phenotype relationship and generate important datasets that help unravel complicated genetic blueprints. Thus, the recently developed artificial intelligence methods can be used to interpret the functions of those DNA sequences. METHODS: This study explores the use of deep learning, particularly pre-trained genomic models like DNA_bert_6 and human_gpt2-v1, in interpreting and representing human genome sequences. Initially, we meticulously constructed multiple datasets linking genotypes and phenotypes to fine-tune those models for precise DNA sequence classification. Additionally, we evaluate the influence of sequence length on classification results and analyze the impact of feature extraction in the hidden layers of our model using the HERV dataset. To enhance our understanding of phenotype-specific patterns recognized by the model, we perform enrichment, pathogenicity and conservation analyzes of specific motifs in the human endogenous retrovirus (HERV) sequence with high average local representation weight (ALRW) scores. RESULTS: We have constructed multiple genotype-phenotype datasets displaying commendable classification performance in comparison with random genomic sequences, particularly in the HERV dataset, which achieved binary and multi-classification accuracies and F1 values exceeding 0.935 and 0.888, respectively. Notably, the fine-tuning of the HERV dataset not only improved our ability to identify and distinguish diverse information types within DNA sequences but also successfully identified specific motifs associated with neurological disorders and cancers in regions with high ALRW scores. Subsequent analysis of these motifs shed light on the adaptive responses of species to environmental pressures and their co-evolution with pathogens. CONCLUSIONS: These findings highlight the potential of pre-trained genomic models in learning DNA sequence representations, particularly when utilizing the HERV dataset, and provide valuable insights for future research endeavors. This study represents an innovative strategy that combines pre-trained genomic model representations with classical methods for analyzing the functionality of genome sequences, thereby promoting cross-fertilization between genomics and artificial intelligence.


Assuntos
Genoma Humano , Genômica , Fenótipo , Humanos , Genômica/métodos , Modelos Genéticos , Retrovirus Endógenos/genética , Aprendizado Profundo , Genótipo
4.
Mol Biol Evol ; 41(8)2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39101626

RESUMO

Retroviruses are an ancient viral family that have globally coevolved with vertebrates and impacted their evolution. In Australia, a continent that has been geographically isolated for millions of years, little is known about retroviruses in wildlife, despite the devastating impacts of a retrovirus on endangered koala populations. We therefore sought to identify and characterize Australian retroviruses through reconstruction of endogenous retroviruses from marsupial genomes, in particular the Tasmanian devil due to its high cancer incidence. We screened 19 marsupial genomes and identified over 80,000 endogenous retrovirus fragments which we classified into eight retrovirus clades. The retroviruses were similar to either Betaretrovirus (5/8) or Gammaretrovirus (3/8) retroviruses, but formed distinct phylogenetic clades compared to extant retroviruses. One of the clades (MEBrv 3) lost an envelope but retained retrotranspositional activity, subsequently amplifying throughout all Dasyuridae genomes. Overall, we provide insights into Australian retrovirus evolution and identify a highly active endogenous retrovirus within Dasyuridae genomes.


Assuntos
Retrovirus Endógenos , Genoma , Marsupiais , Filogenia , Animais , Retrovirus Endógenos/genética , Marsupiais/virologia , Austrália , Evolução Molecular
5.
Cell Stem Cell ; 31(9): 1298-1314.e8, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39146934

RESUMO

Endogenous retroviruses (ERVs) occupy a significant part of the human genome, with some encoding proteins that influence the immune system or regulate cell-cell fusion in early extra-embryonic development. However, whether ERV-derived proteins regulate somatic development is unknown. Here, we report a somatic developmental function for the primate-specific ERVH48-1 (SUPYN/Suppressyn). ERVH48-1 encodes a fragment of a viral envelope that is expressed during early embryonic development. Loss of ERVH48-1 led to impaired mesoderm and cardiomyocyte commitment and diverted cells to an ectoderm-like fate. Mechanistically, ERVH48-1 is localized to sub-cellular membrane compartments through a functional N-terminal signal peptide and binds to the WNT antagonist SFRP2 to promote its polyubiquitination and degradation, thus limiting SFRP2 secretion and blocking repression of WNT/ß-catenin signaling. Knockdown of SFRP2 or expression of a chimeric SFRP2 with the ERVH48-1 signal peptide rescued cardiomyocyte differentiation. This study demonstrates how ERVH48-1 modulates WNT/ß-catenin signaling and cell type commitment in somatic development.


Assuntos
Diferenciação Celular , Retrovirus Endógenos , Proteínas de Membrana , Miócitos Cardíacos , Via de Sinalização Wnt , Humanos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/citologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Retrovirus Endógenos/metabolismo , Retrovirus Endógenos/genética , Animais , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Primatas , Células HEK293 , Mesoderma/metabolismo
6.
Int J Immunopathol Pharmacol ; 38: 3946320241274255, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39122243

RESUMO

OBJECTIVES: Human endogenous retroviruses (HERVs) are integral components of the human genome, and their reactivation has been implicated in the pathogenesis of some malignancies. External viral co-infections are suspected to play a role in HERV transactivation. This study aimed to investigate the expression of HERV-K np9 elements and HERV-R env gene in pediatric acute lymphoblastic leukemia (ALL) patients. Additionally, we explored potential correlations between HERV expression and common viral infections prevalent in this group of patients. METHODS: Blood samples were collected from 43 pediatric ALL patients and 48 age- and sex-matched healthy controls. Quantitative real-time PCR (qRT-PCR) was used to assess the expression of HERV-K np9 and HERV-R env, along with herpes simplex virus (HSV), parvovirus B19, and polyomavirus BK. RESULTS: HERV-K np9 and HERV-R env showed significantly higher expression in the peripheral blood of ALL patients compared to healthy controls (p < .001 and p = .003, respectively). HSV positivity was associated with significantly increased HERV-K np9 expression. No significant correlations were observed between other investigated viruses and HERV gene expression. CONCLUSION: The overexpression of HERV-K np9 and HERV-R env in pediatric ALL patients suggest their potential role in leukemogenesis. Our findings also suggest a possible link between HSV infection and HERV reactivation in this population. Future investigations are needed to understand the precise roles of these genes and viral infections in the development of ALL.


Assuntos
Retrovirus Endógenos , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Retrovirus Endógenos/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/virologia , Masculino , Feminino , Criança , Pré-Escolar , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , Adolescente , Estudos de Casos e Controles
7.
Viruses ; 16(8)2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-39205280

RESUMO

Endogenous viral elements (EVEs) have been reported to exist widely in the genomes of eukaryotic organisms, and they are closely associated with the growth, development, genetics, adaptation, and evolution of their hosts. In this study, two methods-homologous sequence search and genome alignment-were used to explore the endogenous viral sequences in the genomes of Fragaria species. Results revealed abundant endogenous pararetroviruses (EPRVs) in the genomes of Fragaria species, including 786 sequences belonging to five known taxa such as Caulimovirus and other unclassified taxa. Differences were observed in the detected EPRVs between the two methods, with the homologous sequence search having a greater number of EPRVs. On the contrary, genome alignment identified various types and sources of virus-like sequences. Furthermore, through genome alignment, a 267-bp sequence with 95% similarity to the gene encoding the aphid-transmitted protein of Strawberry vein banding virus (Caulimovirus venafragariae) was discovered in the F. chiloensis genome, which was likely a recent insertion. In addition, the statistical analysis of the genome alignment results indicated a remarkably higher abundance of virus-like sequences in the genomes of polyploid strawberries compared with diploid ones. Moreover, the differences in virus-like sequences were observed between the genomes of Fragaria species and those of their close relatives. This study enriched the diversity of viruses that infect strawberries, and laid a theoretical foundation for further research on the origin of endogenous viruses in the strawberry genome, host-virus interactions, adaptation, evolution, and their functions.


Assuntos
Fragaria , Filogenia , Fragaria/virologia , Genoma de Planta , Retrovirus Endógenos/genética , Retrovirus Endógenos/classificação , Vírus de Plantas/genética , Vírus de Plantas/classificação , Vírus de Plantas/isolamento & purificação , Caulimovirus/genética , Caulimovirus/classificação , Genoma Viral
8.
Viruses ; 16(8)2024 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-39205286

RESUMO

Endogenous retroviruses (ERVs) are the remnants of retroviral germline infections and are highly abundant in the genomes of vertebrates. At one time considered to be nothing more than inert 'junk' within genomes, ERVs have been tolerated within host genomes over vast timescales, and their study continues to reveal complex co-evolutionary histories within their respective host species. For example, multiple instances have been characterized of ERVs having been 'borrowed' for normal physiology, from single copies to ones involved in various regulatory networks such as innate immunity and during early development. Within the cell, the accessibility of ERVs is normally tightly controlled by epigenetic mechanisms such as DNA methylation or histone modifications. However, these silencing mechanisms of ERVs are reversible, and epigenetic alterations to the chromatin landscape can thus lead to their aberrant expression, as is observed in abnormal cellular environments such as in tumors. In this review, we focus on ERV transcriptional control and draw parallels and distinctions concerning the loss of regulation in disease, as well as their precise regulation in early development.


Assuntos
Retrovirus Endógenos , Epigênese Genética , Retrovirus Endógenos/genética , Humanos , Animais , Metilação de DNA , Regulação Viral da Expressão Gênica , Transcrição Gênica , Transcrição Viral/genética , Infecções por Retroviridae/virologia
9.
Nat Commun ; 15(1): 7557, 2024 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-39214979

RESUMO

Histone modifications are associated with distinct transcriptional states, but it is unclear whether they instruct gene expression. To investigate this, we mutate histone H3.3 K9 and K27 residues in mouse embryonic stem cells (mESCs). Here, we find that H3.3K9 is essential for controlling specific distal intergenic regions and for proper H3K27me3 deposition at promoters. The H3.3K9A mutation resulted in decreased H3K9me3 at regions encompassing endogenous retroviruses and induced a gain of H3K27ac and nascent transcription. These changes in the chromatin environment unleash cryptic enhancers, resulting in the activation of distinctive transcriptional programs and culminating in protein expression normally restricted to specialized immune cell types. The H3.3K27A mutant disrupts the deposition and spreading of the repressive H3K27me3 mark, particularly impacting bivalent genes with higher basal levels of H3.3 at promoters. Therefore, H3.3K9 and K27 crucially orchestrate repressive chromatin states at cis-regulatory elements and bivalent promoters, respectively, and instruct proper transcription in mESCs.


Assuntos
Cromatina , Elementos Facilitadores Genéticos , Histonas , Lisina , Células-Tronco Embrionárias Murinas , Regiões Promotoras Genéticas , Histonas/metabolismo , Histonas/genética , Animais , Regiões Promotoras Genéticas/genética , Camundongos , Cromatina/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Elementos Facilitadores Genéticos/genética , Lisina/metabolismo , Mutação , Código das Histonas , Transcrição Gênica , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo
10.
Sci Rep ; 14(1): 20236, 2024 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-39215087

RESUMO

Systemic lupus erythematosus (SLE) is a multifactorial disease characterized by the convergence of genetic, immunological, and viral elements resulting in a complex interaction of both internal and external factors. The role of the Epstein-Barr virus (EBV) and human endogenous retroviruses (HERV-E) as triggers and maintenance elements in the pathogenesis of SLE has been widely recognized. Previous studies have independently evaluated the effects of EBV and HERV-E in this disease. In this work, for the first time, these viral factors are jointly investigated in SLE patients. This study aimed at assessing the differential expression of immune regulatory genes and the incidence of specific viral pathogens (EBV and HERV-E), alongside the detailed characterization of surface markers in T- and B-lymphocytes in patients with SLE and control participants. A comparative analysis between patients with SLE and control participants was performed, evaluating the expression of phenotypic markers and genes involved in the immune response (TNF-α, IL-2, IL-6, IL-10, IFNG, TLR3), as well as HERV-E gag and EBV viral genes (LMP1 and BZLF1).A significant association between SLE and EBV was found in this study. A notable increase in EBV LMP1 gene expression was observed in patients with SLE . Also, a significant overexpression of HERV-E was observed, in addition to a considerable increase in the distribution of the cell surface marker CD27 + on T- and B-lymphocytes, observed in individuals with SLE compared to the control group. This study provides evidence regarding the role that EBV virus plays in lymphocytes in the context of SLE, highlighting how both the virus and the host gene expression may influence disease pathogenesis by altering immune regulatory pathways mediated by TNF-α, IFN-γ, and IL-10, as well as parallel overexpression of HERV-E gag. The decrease in TLR3 could indicate a compromised antiviral response, which could facilitate viral reactivation and contribute to disease activity.


Assuntos
Retrovirus Endógenos , Herpesvirus Humano 4 , Leucócitos Mononucleares , Lúpus Eritematoso Sistêmico , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/virologia , Retrovirus Endógenos/genética , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 4/genética , Adulto , Feminino , Masculino , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Leucócitos Mononucleares/metabolismo , Perfilação da Expressão Gênica , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/virologia , Infecções por Vírus Epstein-Barr/genética , Pessoa de Meia-Idade , Linfócitos B/imunologia , Linfócitos B/virologia , Estudos de Casos e Controles , Linfócitos T/imunologia , Citocinas/metabolismo , Citocinas/genética
11.
Sci Adv ; 10(29): eado1218, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39018396

RESUMO

Cancer cells exhibit rewired transcriptional regulatory networks that promote tumor growth and survival. However, the mechanisms underlying the formation of these pathological networks remain poorly understood. Through a pan-cancer epigenomic analysis, we found that primate-specific endogenous retroviruses (ERVs) are a rich source of enhancers displaying cancer-specific activity. In colorectal cancer and other epithelial tumors, oncogenic MAPK/AP1 signaling drives the activation of enhancers derived from the primate-specific ERV family LTR10. Functional studies in colorectal cancer cells revealed that LTR10 elements regulate tumor-specific expression of multiple genes associated with tumorigenesis, such as ATG12 and XRCC4. Within the human population, individual LTR10 elements exhibit germline and somatic structural variation resulting from a highly mutable internal tandem repeat region, which affects AP1 binding activity. Our findings reveal that ERV-derived enhancers contribute to transcriptional dysregulation in response to oncogenic signaling and shape the evolution of cancer-specific regulatory networks.


Assuntos
Neoplasias Colorretais , Retrovirus Endógenos , Regulação Neoplásica da Expressão Gênica , Transdução de Sinais , Humanos , Neoplasias Colorretais/genética , Neoplasias Colorretais/virologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Retrovirus Endógenos/genética , Elementos Facilitadores Genéticos , Linhagem Celular Tumoral , Transcrição Gênica , Animais , Carcinogênese/genética , Redes Reguladoras de Genes
12.
Front Cell Infect Microbiol ; 14: 1404431, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39081866

RESUMO

Introduction: Endogenous retroviruses (ERVs), which originated from exogenous retroviral infections of germline cells millions of years ago and were inherited by subsequent generations as per Mendelian inheritance patterns, predominantly comprise non-protein-coding sequences due to the accumulation of mutations, insertions, deletions, and truncations. Nevertheless, recent studies have revealed that ERVs play a crucial role in diverse biological processes by encoding various proteins. Methods: In this study, we successfully identified an ERV envelope (env) gene in a mink species. A phylogenetic tree of mink ERV-V env and reference sequences was constructed using Bayesian methods and maximum-likelihood inference. Results: Phylogenetic analyses indicated a significant degree of sequence conservation and positive selection within the env-surface open reading frame. Additionally, qRT-PCR revealed diverse patterns of mink ERV-V env expression in various tissues. The expression of mink ERV-V env gene in testicular tissue strongly correlated with the seasonal reproductive cycles of minks. Discussion: Our study suggests that the ERV-V env gene in mink may have been repurposed for host functions.


Assuntos
Retrovirus Endógenos , Vison , Filogenia , Retrovirus Endógenos/genética , Animais , Vison/virologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Estações do Ano , Reprodução/genética , Masculino , Testículo/virologia , Teorema de Bayes
13.
Curr Opin Virol ; 67: 101427, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39047314

RESUMO

The koala retrovirus, KoRV, is one of the few models for understanding the health consequences of retroviral colonization of the germline. Such colonization events transition exogenous infectious retroviruses to Mendelian traits or endogenous retroviruses (ERVs). KoRV is currently in a transitional state from exogenous retrovirus to ERV, which in koalas (Phascolarctos cinereus) has been associated with strongly elevated levels of neoplasia. In this review, we describe what is currently known about the associations and underlying mechanisms of KoRV-induced neoplasia.


Assuntos
Retrovirus Endógenos , Neoplasias , Phascolarctidae , Infecções por Retroviridae , Animais , Neoplasias/virologia , Phascolarctidae/virologia , Retrovirus Endógenos/genética , Retrovirus Endógenos/fisiologia , Retrovirus Endógenos/patogenicidade , Infecções por Retroviridae/virologia , Infecções por Retroviridae/veterinária , Humanos , Retroviridae/fisiologia , Retroviridae/patogenicidade , Retroviridae/genética
14.
Genes Dev ; 38(11-12): 554-568, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38960717

RESUMO

Retrotransposon control in mammals is an intricate process that is effectuated by a broad network of chromatin regulatory pathways. We previously discovered ChAHP, a protein complex with repressive activity against short interspersed element (SINE) retrotransposons that is composed of the transcription factor ADNP, chromatin remodeler CHD4, and HP1 proteins. Here we identify ChAHP2, a protein complex homologous to ChAHP, in which ADNP is replaced by ADNP2. ChAHP2 is predominantly targeted to endogenous retroviruses (ERVs) and long interspersed elements (LINEs) via HP1ß-mediated binding of H3K9 trimethylated histones. We further demonstrate that ChAHP also binds these elements in a manner mechanistically equivalent to that of ChAHP2 and distinct from DNA sequence-specific recruitment at SINEs. Genetic ablation of ADNP2 alleviates ERV and LINE1 repression, which is synthetically exacerbated by additional depletion of ADNP. Together, our results reveal that the ChAHP and ChAHP2 complexes function to control both nonautonomous and autonomous retrotransposons by complementary activities, further adding to the complexity of mammalian transposon control.


Assuntos
Retroelementos , Animais , Humanos , Camundongos , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Cromossômicas não Histona/genética , Retrovirus Endógenos/genética , Regulação da Expressão Gênica/genética , Histonas/metabolismo , Histonas/genética , Elementos Nucleotídeos Longos e Dispersos/genética , Ligação Proteica , Retroelementos/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Complexos Multiproteicos/metabolismo
16.
BMC Genomics ; 25(1): 707, 2024 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-39033293

RESUMO

Most studied, investigating transcriptional changes in myocardial biopsies focus on human genes. However, the presence and potential consequence of persistent expression of viral genes within the myocardium is unclear. The aim of the study was to analyze viral gene expression in RNAseq data from endomyocardial biopsies. The NCBI Bioproject library was screened for published projects that included bulk RNA sequencing data from endomyocardial biopsies from both healthy and diseased patients with a sample size greater than 20. Diseased patients with hypertrophic, dilated, and ischemic cardiomyopathies were included. A total of 507 patients with 507 samples from 6 bioprojects were included and mapped to the human genome (hg38). Unmappable sequences were extracted and mapped to an artificial 'super-virus' genome comprising 12,182 curated viral reference genomes. Subsequently, the sequences were reiteratively permutated and mapped again to account for randomness. In total, sequences from 68 distinct viruses were found, all of which were potentially human pathogenic. No increase in cardiotropic viruses was found in patients with dilated cardiomyopathy. However, the expression levels of the particle forming human endogenous retrovirus K were significantly increased (q < 0.0003, ANOVA). Higher expression levels were associated with increased expression in mitochondrial pathways such as oxidative phosphorylation (p < 0.0001). In Conclusion, expression of human endogenous retrovirus K is significantly increased in patients with dilated cardiomyopathy, which in turn was associated with transcriptional alterations in major cellular pathways.


Assuntos
Cardiomiopatias , Miocárdio , Humanos , Cardiomiopatias/virologia , Cardiomiopatias/genética , Cardiomiopatias/patologia , Cardiomiopatias/metabolismo , Biópsia , Miocárdio/metabolismo , Miocárdio/patologia , Retrovirus Endógenos/genética , Perfilação da Expressão Gênica , Transcriptoma
17.
Elife ; 132024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39037763

RESUMO

HERV-K(HML-2), the youngest clade of human endogenous retroviruses (HERVs), includes many intact or nearly intact proviruses, but no replication competent HML-2 proviruses have been identified in humans. HML-2-related proviruses are present in other primates, including rhesus macaques, but the extent and timing of HML-2 activity in macaques remains unclear. We have identified 145 HML-2-like proviruses in rhesus macaques, including a clade of young, rhesus-specific insertions. Age estimates, intact open reading frames, and insertional polymorphism of these insertions are consistent with recent or ongoing infectious activity in macaques. 106 of the proviruses form a clade characterized by an ~750 bp sequence between env and the 3' long terminal repeat (LTR), derived from an ancient recombination with a HERV-K(HML-8)-related virus. This clade is found in Old World monkeys (OWM), but not great apes, suggesting it originated after the ape/OWM split. We identified similar proviruses in white-cheeked gibbons; the gibbon insertions cluster within the OWM recombinant clade, suggesting interspecies transmission from OWM to gibbons. The LTRs of the youngest proviruses have deletions in U3, which disrupt the Rec Response Element (RcRE), required for nuclear export of unspliced viral RNA. We show that the HML-8-derived region functions as a Rec-independent constitutive transport element (CTE), indicating the ancestral Rec-RcRE export system was replaced by a CTE mechanism.


Just as we study fossils to understand how animals and plants have evolved, we can study ancient viruses to understand how diseases have emerged and changed over long periods. Unlike fossils, viruses do not leave visible traces in the ground but, instead, they leave viral genes known as endogenous viral elements (or EVEs) that become permanently incorporated in their host's DNA. HML-2s are the youngest known EVEs in the human genome. They have evolved gradually by accumulating lots of small genetic changes and no longer actively infect humans. But these virus remnants have long been suspected to play a role in prostate cancer, lupus and other human diseases. Rhesus macaques and other monkeys also have HML-2s but these are less well studied than human HML-2s. Monkeys are often used as models of human biology in research studies, therefore, understanding how HML-2s have evolved in rhesus macaques may enable researchers to establish this monkey as a model for investigating the role of HML-2s in humans. To investigate this possibility, Williams et al. searched for HML-like EVEs in rhesus macaque genomes published in previous studies. The experiments found that, unlike human HML-2s, the macaque HML-2s underwent a sudden genetic transformation millions of years ago. They acquired a new gene from another virus that completely changed how the macaque HML-2s leave a compartment within the cells of their host that contains most of the host's genome ­ a key step in the life cycle of viruses. The data also suggest that HML-2s may still be actively infecting macaques today and that these EVEs jumped from monkeys into gibbons. This is the first known example of HML-2s moving between different types of primates and it indicates there may be a risk that macaque HML-2s could infect humans. In the future, the findings of Williams et al. may help researchers develop new approaches to treat prostate cancer and other diseases linked with HML-2s in humans.


Assuntos
Retrovirus Endógenos , Macaca mulatta , Provírus , Recombinação Genética , Animais , Retrovirus Endógenos/genética , Macaca mulatta/virologia , Provírus/genética , Humanos , Infecções por Retroviridae/transmissão , Infecções por Retroviridae/virologia , Infecções por Retroviridae/veterinária , RNA Viral/genética , Filogenia
18.
Schizophr Res ; 271: 220-227, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39053037

RESUMO

OBJECTIVE: Human endogenous retroviruses (HERV) are the remnants of infections that occurred million years ago. They gradually integrated into the human genome, comprising 8 % of it. There are growing reports suggesting their potential role in various diseases, including schizophrenia. Schizophrenia, a serious psychiatric disorder, is caused by the interaction of genetic and environmental factors. In the present paper, we investigated studies focusing on the association between schizophrenia and HERV-W. METHODS: We registered this study at PROSPERO (registration number: CRD42022301122). The entire steps of this study were based on the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. We searched PubMed, Scopus, Web of Science, and Google Scholar up to 1 August 2022. Heterogeneity was estimated through I2 statistics, and the association was measured using the first estimate and penalization methods. RESULTS: Finally, 13 eligible studies were analyzed, including 698 cases and 728 controls. The overall odds ratio indicated a significant association in both the first estimate (OR = 9.34, 95 % CI = 4.92-17.75; P = 0.002) and penalization (OR = 7.38, 95 % CI = 4.15-13.10; P = 0.003) methods. In the subgroup analysis, among HERV-W fragments, the HERV-W envelope protein or RNA (OR = 11.41, 95 % CI: 5.67-22.97; P = 0.03) showed the strongest association with schizophrenia. CONCLUSION: Our meta-analysis showed that HERV-W is significantly associated with schizophrenia. More studies are required to determine the pathophysiological mechanism and the diagnostic, prognostic, and therapeutic value of HERV-W in schizophrenia.


Assuntos
Retrovirus Endógenos , Esquizofrenia , Esquizofrenia/virologia , Esquizofrenia/epidemiologia , Humanos , Retrovirus Endógenos/genética , Ativação Viral
19.
Ageing Res Rev ; 99: 102392, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38925481

RESUMO

The present perspective article proposes an etiopathological model for multiple sclerosis pathogenesis and progression associated with the activation of human endogenous retroviruses. We reviewed preclinical, clinical, epidemiological, and evolutionary evidence indicating how the complex, multi-level interplay of genetic traits and environmental factors contributes to multiple sclerosis. We propose that endogenous retroviruses transactivation acts as a critical node in disease development. We also discuss the rationale for combined anti-retroviral therapy in multiple sclerosis as a disease-modifying therapeutic strategy. Finally, we propose that the immuno-pathogenic process triggered by endogenous retrovirus activation can be extended to aging and aging-related neurodegeneration. In this regard, endogenous retroviruses can be envisioned to act as epigenetic noise, favoring the proliferation of disorganized cellular subpopulations and accelerating system-specific "aging". Since inflammation and aging are two sides of the same coin (plastic dis-adaptation to external stimuli with system-specific degree of freedom), the two conditions may be epiphenomenal products of increased epigenomic entropy. Inflammation accelerates organ-specific aging, disrupting communication throughout critical systems of the body and producing symptoms. Overlapping neurological symptoms and syndromes may emerge from the activity of shared molecular networks that respond to endogenous retroviruses' reactivation.


Assuntos
Retrovirus Endógenos , Esclerose Múltipla , Humanos , Retrovirus Endógenos/patogenicidade , Retrovirus Endógenos/genética , Esclerose Múltipla/virologia , Esclerose Múltipla/etiologia , Envelhecimento , Animais , Epigênese Genética
20.
Nat Commun ; 15(1): 4920, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858353

RESUMO

The differentiation of the stroma is a hallmark event during postnatal uterine development. However, the spatiotemporal changes that occur during this process and the underlying regulatory mechanisms remain elusive. Here, we comprehensively delineated the dynamic development of the neonatal uterus at single-cell resolution and characterized two distinct stromal subpopulations, inner and outer stroma. Furthermore, single-cell RNA sequencing revealed that uterine ablation of Pr-set7, the sole methyltransferase catalyzing H4K20me1, led to a reduced proportion of the inner stroma due to massive cell death, thus impeding uterine development. By combining RNA sequencing and epigenetic profiling of H4K20me1, we demonstrated that PR-SET7-H4K20me1 either directly repressed the transcription of interferon stimulated genes or indirectly restricted the interferon response via silencing endogenous retroviruses. Declined H4K20me1 level caused viral mimicry responses and ZBP1-mediated apoptosis and necroptosis in stromal cells. Collectively, our study provides insight into the epigenetic machinery governing postnatal uterine stromal development mediated by PR-SET7.


Assuntos
Epigênese Genética , Histona-Lisina N-Metiltransferase , Células Estromais , Útero , Feminino , Animais , Útero/metabolismo , Células Estromais/metabolismo , Camundongos , Histona-Lisina N-Metiltransferase/metabolismo , Histona-Lisina N-Metiltransferase/genética , Interferons/metabolismo , Interferons/genética , Retrovirus Endógenos/genética , Apoptose/genética , Camundongos Endogâmicos C57BL , Morte Celular/genética , Necroptose/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Histonas/metabolismo , Análise de Célula Única , Camundongos Knockout , Diferenciação Celular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...