Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Mol Sci ; 24(3)2023 Jan 25.
Artículo en Inglés | MEDLINE | ID: covidwho-2288568

RESUMEN

Alveolar macrophages (AMs) form the first defense line against various respiratory pathogens, and their immune response has a profound impact on the outcome of respiratory infection. Enhancer of zeste homolog 2 (EZH2), which catalyzes the trimethylation of H3K27 for epigenetic repression, has gained increasing attention for its immune regulation function, yet its exact function in AMs remains largely obscure. Using porcine 3D4/21 AM cells as a model, we characterized the transcriptomic and epigenomic alterations after the inhibition of EZH2. We found that the inhibition of EZH2 causes transcriptional activation of numerous immune genes and inhibits the subsequent infection by influenza A virus. Interestingly, specific families of transposable elements, particularly endogenous retrovirus elements (ERVs) and LINEs which belong to retrotransposons, also become derepressed. While some of the derepressed ERV families are pig-specific, a few ancestral families are known to be under EZH2-mediated repression in humans. Given that derepression of ERVs can promote innate immune activation through "viral mimicry", we speculate that ERVs may also contribute to the coinciding immune activation in AMs after the inhibition of EZH2. Overall, this study improves the understanding of the EZH2-related immune regulation in AMs and provides novel insights into the epigenetic regulation of retrotransposons in pigs.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2 , Complejo Represivo Polycomb 2 , Humanos , Animales , Porcinos , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Complejo Represivo Polycomb 2/genética , Retroelementos/genética , Epigénesis Genética , Macrófagos Alveolares/metabolismo , Pulmón/metabolismo
2.
Int J Mol Sci ; 24(4)2023 Feb 04.
Artículo en Inglés | MEDLINE | ID: covidwho-2282433

RESUMEN

Type 1 diabetes (T1D) is one of the most common chronic diseases of the endocrine system, associated with several life-threatening comorbidities. While the etiopathogenesis of T1D remains elusive, a combination of genetic susceptibility and environmental factors, such as microbial infections, are thought to be involved in the development of the disease. The prime model for studying the genetic component of T1D predisposition encompasses polymorphisms within the HLA (human leukocyte antigen) region responsible for the specificity of antigen presentation to lymphocytes. Apart from polymorphisms, genomic reorganization caused by repeat elements and endogenous viral elements (EVEs) might be involved in T1D predisposition. Such elements are human endogenous retroviruses (HERVs) and non-long terminal repeat (non-LTR) retrotransposons, including long and short interspersed nuclear elements (LINEs and SINEs). In line with their parasitic origin and selfish behaviour, retrotransposon-imposed gene regulation is a major source of genetic variation and instability in the human genome, and may represent the missing link between genetic susceptibility and environmental factors long thought to contribute to T1D onset. Autoreactive immune cell subtypes with differentially expressed retrotransposons can be identified with single-cell transcriptomics, and personalized assembled genomes can be constructed, which can then serve as a reference for predicting retrotransposon integration/restriction sites. Here we review what is known to date about retrotransposons, we discuss the involvement of viruses and retrotransposons in T1D predisposition, and finally we consider challenges in retrotransposons analysis methods.


Asunto(s)
Diabetes Mellitus Tipo 1 , Retrovirus Endógenos , Humanos , Retroelementos , Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad , Elementos de Nucleótido Esparcido Corto
3.
BMC Genom Data ; 23(1): 27, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1840943

RESUMEN

BACKGROUND: The factors driving the late phase of COVID-19 are still poorly understood. However, autoimmunity is an evolving theme in COVID-19's pathogenesis. Additionally, deregulation of human retroelements (RE) is found in many viral infections, and has also been reported in COVID-19. RESULTS: Unexpectedly, coronaviruses (CoV) - including SARS-CoV-2 - harbour many RE-identical sequences (up to 35 base pairs), and some of these sequences are part of SARS-CoV-2 epitopes associated to COVID-19 severity. Furthermore, RE are expressed in healthy controls and human cells and become deregulated after SARS-CoV-2 infection, showing mainly changes in long interspersed nuclear element (LINE1) expression, but also in endogenous retroviruses. CONCLUSION: CoV and human RE share coding sequences, which are targeted by antibodies in COVID-19 and thus could induce an autoimmune loop by molecular mimicry.


Asunto(s)
COVID-19 , SARS-CoV-2 , Epítopos , Humanos , Imitación Molecular , Retroelementos/genética , SARS-CoV-2/genética
4.
Genes (Basel) ; 13(5)2022 04 20.
Artículo en Inglés | MEDLINE | ID: covidwho-1792755

RESUMEN

The major advantage of mRNA vaccines over more conventional approaches is their potential for rapid development and large-scale deployment in pandemic situations. In the current COVID-19 crisis, two mRNA COVID-19 vaccines have been conditionally approved and broadly applied, while others are still in clinical trials. However, there is no previous experience with the use of mRNA vaccines on a large scale in the general population. This warrants a careful evaluation of mRNA vaccine safety properties by considering all available knowledge about mRNA molecular biology and evolution. Here, I discuss the pervasive claim that mRNA-based vaccines cannot alter genomes. Surprisingly, this notion is widely stated in the mRNA vaccine literature but never supported by referencing any primary scientific papers that would specifically address this question. This discrepancy becomes even more puzzling if one considers previous work on the molecular and evolutionary aspects of retroposition in murine and human populations that clearly documents the frequent integration of mRNA molecules into genomes, including clinical contexts. By performing basic comparisons, I show that the sequence features of mRNA vaccines meet all known requirements for retroposition using L1 elements-the most abundant autonomously active retrotransposons in the human genome. In fact, many factors associated with mRNA vaccines increase the possibility of their L1-mediated retroposition. I conclude that is unfounded to a priori assume that mRNA-based therapeutics do not impact genomes and that the route to genome integration of vaccine mRNAs via endogenous L1 retroelements is easily conceivable. This implies that we urgently need experimental studies that would rigorously test for the potential retroposition of vaccine mRNAs. At present, the insertional mutagenesis safety of mRNA-based vaccines should be considered unresolved.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Animales , Biología , COVID-19/prevención & control , Vacunas contra la COVID-19/genética , Humanos , Elementos de Nucleótido Esparcido Largo , Ratones , ARN Mensajero/genética , Retroelementos , Vacunas Sintéticas/genética , Vacunas de ARNm
5.
JCI Insight ; 6(24)2021 12 22.
Artículo en Inglés | MEDLINE | ID: covidwho-1501860

RESUMEN

SARS-CoV-2 promotes an imbalanced host response that underlies the development and severity of COVID-19. Infections with viruses are known to modulate transposable elements (TEs), which can exert downstream effects by modulating host gene expression, innate immune sensing, or activities encoded by their protein products. We investigated the impact of SARS-CoV-2 infection on TE expression using RNA-Seq data from cell lines and from primary patient samples. Using a bioinformatics tool, Telescope, we showed that SARS-CoV-2 infection led to upregulation or downregulation of TE transcripts, a subset of which differed from cells infected with SARS, Middle East respiratory syndrome coronavirus (MERS-CoV or MERS), influenza A virus (IAV), respiratory syncytial virus (RSV), and human parainfluenza virus type 3 (HPIV3). Differential expression of key retroelements specifically identified distinct virus families, such as Coronaviridae, with unique retroelement expression subdividing viral species. Analysis of ChIP-Seq data showed that TEs differentially expressed in SARS-CoV-2 infection were enriched for binding sites for transcription factors involved in immune responses and for pioneer transcription factors. In samples from patients with COVID-19, there was significant TE overexpression in bronchoalveolar lavage fluid and downregulation in PBMCs. Thus, although the host gene transcriptome is altered by infection with SARS-CoV-2, the retrotranscriptome may contain the most distinctive features of the cellular response to SARS-CoV-2 infection.


Asunto(s)
COVID-19/genética , Retrovirus Endógenos/genética , Elementos de Nucleótido Esparcido Largo/genética , Células A549 , Línea Celular , Secuenciación de Inmunoprecipitación de Cromatina , Biología Computacional , Infecciones por Coronavirus/genética , Elementos Transponibles de ADN/genética , Regulación hacia Abajo , Interacciones Microbiota-Huesped/genética , Humanos , Técnicas In Vitro , Virus de la Influenza A , Gripe Humana/genética , Coronavirus del Síndrome Respiratorio de Oriente Medio , Virus de la Parainfluenza 3 Humana , RNA-Seq , Infecciones por Virus Sincitial Respiratorio/genética , Virus Sincitiales Respiratorios , Infecciones por Respirovirus/genética , Retroelementos/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/genética , Transcriptoma , Regulación hacia Arriba
6.
J Virol ; 95(12)2021 05 24.
Artículo en Inglés | MEDLINE | ID: covidwho-1501541

RESUMEN

Long disregarded as junk DNA or genomic dark matter, endogenous retroviruses (ERVs) have turned out to represent important components of the antiviral immune response. These remnants of once-infectious retroviruses not only regulate cellular immune activation, but may even directly target invading viral pathogens. In this Gem, we summarize mechanisms by which retroviral fossils protect us from viral infections. One focus will be on recent advances in the role of ERVs as regulators of antiviral gene expression.


Asunto(s)
Retrovirus Endógenos/fisiología , Retroelementos , Virosis/inmunología , Animales , Retrovirus Endógenos/genética , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Humanos , Inmunidad Celular , Regiones Promotoras Genéticas , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Receptores Virales/antagonistas & inhibidores , Receptores Virales/metabolismo , Proteínas Virales/metabolismo , Virión/metabolismo , Virosis/genética , Virosis/virología
7.
Retrovirology ; 18(1): 34, 2021 10 29.
Artículo en Inglés | MEDLINE | ID: covidwho-1486582

RESUMEN

Zhang et al. (Proc Natl Acad Sci 118:e2105968118, 2021) recently reported that SARS-CoV-2 RNA can be retrotranscribed and integrated into the DNA of human cells by the L1 retrotransposon machinery. This phenomenon could cause persistence of viral sequences in patients and may explain the prolonged PCR-positivity of SARS-CoV-2 infected patients, even long after the phase of active virus replication has ended. This commentary does critically review the available data on this topic and discusses them in the context of findings made for other exogenous viruses and ancestral endogenous retroviral elements.


Asunto(s)
COVID-19 , Retroelementos , Humanos , ARN Viral , SARS-CoV-2 , Replicación Viral
8.
Science ; 374(6567): eabj3624, 2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: covidwho-1440797

RESUMEN

Inherited genetic factors can influence the severity of COVID-19, but the molecular explanation underpinning a genetic association is often unclear. Intracellular antiviral defenses can inhibit the replication of viruses and reduce disease severity. To better understand the antiviral defenses relevant to COVID-19, we used interferon-stimulated gene (ISG) expression screening to reveal that 2'-5'-oligoadenylate synthetase 1 (OAS1), through ribonuclease L, potently inhibits severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We show that a common splice-acceptor single-nucleotide polymorphism (Rs10774671) governs whether patients express prenylated OAS1 isoforms that are membrane-associated and sense-specific regions of SARS-CoV-2 RNAs or if they only express cytosolic, nonprenylated OAS1 that does not efficiently detect SARS-CoV-2. In hospitalized patients, expression of prenylated OAS1 was associated with protection from severe COVID-19, suggesting that this antiviral defense is a major component of a protective antiviral response.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , COVID-19/genética , COVID-19/fisiopatología , ARN Bicatenario/metabolismo , ARN Viral/metabolismo , SARS-CoV-2/fisiología , Regiones no Traducidas 5' , Células A549 , Animales , COVID-19/enzimología , COVID-19/inmunología , Quirópteros/genética , Quirópteros/virología , Coronaviridae/enzimología , Coronaviridae/genética , Coronaviridae/fisiología , Endorribonucleasas/metabolismo , Humanos , Interferones/inmunología , Isoenzimas/genética , Isoenzimas/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Polimorfismo de Nucleótido Simple , Prenilación de Proteína , ARN Bicatenario/química , ARN Bicatenario/genética , ARN Viral/química , ARN Viral/genética , Retroelementos , SARS-CoV-2/genética , Índice de Severidad de la Enfermedad , Replicación Viral
9.
Front Cell Infect Microbiol ; 11: 609160, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1140637

RESUMEN

There is an increased global outbreak of diseases caused by coronaviruses affecting respiratory tracts of birds and mammals. Recent dangerous coronaviruses are MERS-CoV, SARS-CoV, and SARS-CoV-2, causing respiratory illness and even failure of several organs. However, profound impact of coronavirus on host cells remains elusive. In this study, we analyzed transcriptome of MERS-CoV, SARS-CoV, and SARS-CoV-2 infected human lung-derived cells, and observed that infection of these coronaviruses all induced increase of retrotransposon expression with upregulation of TET genes. Upregulation of retrotransposon was also observed in SARS-CoV-2 infected human intestinal organoids. Retrotransposon upregulation may lead to increased genome instability and enhanced expression of genes with readthrough from retrotransposons. Therefore, people with higher basal level of retrotransposon such as cancer patients and aged people may have increased risk of symptomatic infection. Additionally, we show evidence supporting long-term epigenetic inheritance of retrotransposon upregulation. We also observed chimeric transcripts of retrotransposon and SARS-CoV-2 RNA for potential human genome invasion of viral fragments, with the front and the rear part of SARS-CoV-2 genome being easier to form chimeric RNA. Thus, we suggest that primers and probes for nucleic acid detection should be designed in the middle of virus genome to identify live virus with higher probability. In summary, we propose our hypothesis that coronavirus invades human cells and interacts with retrotransposon, eliciting more severe symptoms in patients with underlying diseases. In the treatment of patients with coronavirus infection, it may be necessary to pay more attention to the potential harm contributed by retrotransposon dysregulation.


Asunto(s)
Infecciones por Coronavirus/virología , Coronavirus/genética , Genoma Viral/genética , Retroelementos/genética , Transcriptoma , Línea Celular Tumoral , Humanos , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , SARS-CoV-2/genética
10.
Hum Genomics ; 15(1): 2, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: covidwho-1004357

RESUMEN

SARS-CoV-2 has spread rapidly across the world and is negatively impacting the global human population. COVID-19 patients display a wide variety of symptoms and clinical outcomes, including those attributed to genetic ancestry. Alu retrotransposons have played an important role in human evolution, and their variants influence host response to viral infection. Intronic Alus regulate gene expression through several mechanisms, including both genetic and epigenetic pathways. With respect to SARS-CoV-2, an intronic Alu within the ACE gene is hypothesized to be associated with COVID-19 susceptibility and morbidity. Here, we review specific Alu polymorphisms that are of particular interest when considering host response to SARS-CoV-2 infection, especially polymorphic Alu insertions in genes associated with immune response and coagulation/fibrinolysis cascade. We posit that additional research focused on Alu-related pathways could yield novel biomarkers capable of predicting clinical outcomes as well as patient-specific treatment strategies for COVID-19 and related infectious diseases.


Asunto(s)
COVID-19/genética , Predisposición Genética a la Enfermedad , Retroelementos , COVID-19/fisiopatología , COVID-19/virología , Humanos , Morbilidad , SARS-CoV-2/aislamiento & purificación
11.
Nat Genet ; 52(12): 1294-1302, 2020 12.
Artículo en Inglés | MEDLINE | ID: covidwho-880696

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) is an entry receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and a regulator of several physiological processes. ACE2 has recently been proposed to be interferon (IFN) inducible, suggesting that SARS-CoV-2 may exploit this phenomenon to enhance viral spread and questioning the efficacy of IFN treatment in coronavirus disease 2019. Using a recent de novo transcript assembly that captured previously unannotated transcripts, we describe a new isoform of ACE2, generated by co-option of intronic retroelements as promoter and alternative exon. The new transcript, termed MIRb-ACE2, exhibits specific expression patterns across the aerodigestive and gastrointestinal tracts and is highly responsive to IFN stimulation. In contrast, canonical ACE2 expression is unresponsive to IFN stimulation. Moreover, the MIRb-ACE2 translation product is a truncated, unstable ACE2 form, lacking domains required for SARS-CoV-2 binding and is therefore unlikely to contribute to or enhance viral infection.


Asunto(s)
Enzima Convertidora de Angiotensina 2/biosíntesis , Interferones/metabolismo , Retroelementos/genética , Enzima Convertidora de Angiotensina 2/genética , Animales , Línea Celular , Chlorocebus aethiops , Inducción Enzimática , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Células HEK293 , Humanos , Isoenzimas/biosíntesis , Isoenzimas/genética , Estabilidad Proteica , RNA-Seq , Receptores de Coronavirus/metabolismo , SARS-CoV-2/metabolismo , Distribución Tisular , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA