Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
1.
Front Immunol ; 12: 736349, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867957

RESUMEN

The majority of human genome are non-coding genes. Recent research have revealed that about half of these genome sequences make up of transposable elements (TEs). A branch of these belong to the endogenous retroviruses (ERVs), which are germline viral infection that occurred over millions of years ago. They are generally harmless as evolutionary mutations have made them unable to produce viral agents and are mostly epigenetically silenced. Nevertheless, ERVs are able to express by still unknown mechanisms and recent evidences have shown links between ERVs and major proinflammatory diseases and cancers. The major challenge is to elucidate a detailed mechanistic understanding between them, so that novel therapeutic approaches can be explored. Here, we provide a brief overview of TEs, human ERVs and their links to microbiome, innate immune response, proinflammatory diseases and cancer. Finally, we recommend the employment of systems biology approaches for future HERV research.


Asunto(s)
Retrovirus Endógenos/patogenicidad , Inflamación/etiología , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/virología , Evolución Biológica , Elementos Transponibles de ADN/genética , Retrovirus Endógenos/genética , Retrovirus Endógenos/inmunología , Genoma Humano , Humanos , Inmunidad Innata , Inflamación/inmunología , Inflamación/virología , Aprendizaje Automático , Microbiota/inmunología , Modelos Biológicos , Neoplasias/etiología , Neoplasias/inmunología , Neoplasias/virología , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/virología , Biología de Sistemas
2.
Front Immunol ; 12: 757302, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790199

RESUMEN

Roles for viral infections and aberrant immune responses in driving localized neuroinflammation and neurodegeneration in multiple sclerosis (MS) are the focus of intense research. Epstein-Barr virus (EBV), as a persistent and frequently reactivating virus with major immunogenic influences and a near 100% epidemiological association with MS, is considered to play a leading role in MS pathogenesis, triggering localized inflammation near or within the central nervous system (CNS). This triggering may occur directly via viral products (RNA and protein) and/or indirectly via antigenic mimicry involving B-cells, T-cells and cytokine-activated astrocytes and microglia cells damaging the myelin sheath of neurons. The genetic MS-risk factor HLA-DR2b (DRB1*1501ß, DRA1*0101α) may contribute to aberrant EBV antigen-presentation and anti-EBV reactivity but also to mimicry-induced autoimmune responses characteristic of MS. A central role is proposed for inflammatory EBER1, EBV-miRNA and LMP1 containing exosomes secreted by viable reactivating EBV+ B-cells and repetitive release of EBNA1-DNA complexes from apoptotic EBV+ B-cells, forming reactive immune complexes with EBNA1-IgG and complement. This may be accompanied by cytokine- or EBV-induced expression of human endogenous retrovirus-W/-K (HERV-W/-K) elements and possibly by activation of human herpesvirus-6A (HHV-6A) in early-stage CNS lesions, each contributing to an inflammatory cascade causing the relapsing-remitting neuro-inflammatory and/or progressive features characteristic of MS. Elimination of EBV-carrying B-cells by antibody- and EBV-specific T-cell therapy may hold the promise of reducing EBV activity in the CNS, thereby limiting CNS inflammation, MS symptoms and possibly reversing disease. Other approaches targeting HHV-6 and HERV-W and limiting inflammatory kinase-signaling to treat MS are also being tested with promising results. This article presents an overview of the evidence that EBV, HHV-6, and HERV-W may have a pathogenic role in initiating and promoting MS and possible approaches to mitigate development of the disease.


Asunto(s)
Retrovirus Endógenos/patogenicidad , Herpesvirus Humano 4/patogenicidad , Herpesvirus Humano 6/patogenicidad , Esclerosis Múltiple/etiología , Enfermedades Neuroinflamatorias/virología , Anticuerpos Antivirales/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Autoinmunidad , Linfocitos B/inmunología , Barrera Hematoencefálica , Encéfalo/virología , Coinfección , ADN Viral/inmunología , Retrovirus Endógenos/fisiología , Antígenos Nucleares del Virus de Epstein-Barr/genética , Antígenos Nucleares del Virus de Epstein-Barr/inmunología , Productos del Gen env/fisiología , Predisposición Genética a la Enfermedad , Infecciones por Herpesviridae/complicaciones , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 6/inmunología , Humanos , Ganglios Linfáticos/virología , Modelos Inmunológicos , Imitación Molecular , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Esclerosis Múltiple/virología , Vaina de Mielina/inmunología , Vaina de Mielina/patología , Enfermedades Neuroinflamatorias/etiología , Proteínas Gestacionales/fisiología , Activación Transcripcional , Activación Viral , Latencia del Virus
3.
Front Immunol ; 12: 728677, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34691035

RESUMEN

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


Asunto(s)
Autoinmunidad , Infecciones por Virus de Epstein-Barr/epidemiología , Salud Global , Esclerosis Múltiple/epidemiología , Paratuberculosis/epidemiología , Infecciones por Retroviridae/epidemiología , Retrovirus Endógenos/inmunología , Retrovirus Endógenos/patogenicidad , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 4/patogenicidad , Interacciones Huésped-Patógeno , Humanos , Italia/epidemiología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/microbiología , Esclerosis Múltiple/virología , Mycobacterium avium subsp. paratuberculosis/inmunología , Mycobacterium avium subsp. paratuberculosis/patogenicidad , Paratuberculosis/inmunología , Paratuberculosis/microbiología , Pronóstico , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Medición de Riesgo , Factores de Riesgo
4.
Int J Mol Sci ; 22(14)2021 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-34298881

RESUMEN

Chronic neurodegenerative diseases are complex, and their pathogenesis is uncertain. Alzheimer's disease (AD) is a neurodegenerative brain alteration that is responsible for most dementia cases in the elderly. AD etiology is still uncertain; however, chronic neuroinflammation is a constant component of brain pathology. Infections have been associated with several neurological diseases and viruses of the Herpes family appear to be a probable cause of AD neurodegenerative alterations. Several different factors may contribute to the AD clinical progression. Exogeneous viruses or other microbes and environmental pollutants may directly induce neurodegeneration by activating brain inflammation. In this paper, we suggest that exogeneous brain insults may also activate retrotransposons and silent human endogenous retroviruses (HERVs). The initial inflammation of small brain areas induced by virus infections or other brain insults may activate HERV dis-regulation that contributes to neurodegenerative mechanisms. Chronic HERV activation in turn may cause progressive neurodegeneration that thereafter merges in cognitive impairment and dementia in genetically susceptible people. Specific treatment for exogenous end endogenous pathogens and decreasing pollutant exposure may show beneficial effect in early intervention protocol to prevent the progression of cognitive deterioration in the elderly.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/virología , Encéfalo/patología , Encéfalo/virología , Retrovirus Endógenos/patogenicidad , Virosis/patología , Virosis/virología , Animales , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/virología , Encefalitis/patología , Encefalitis/virología , Humanos
5.
JCI Insight ; 6(15)2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34185707

RESUMEN

We previously reported heightened expression of the human endogenous retroviral protein HERV-K deoxyuridine triphosphate nucleotidohydrolase (dUTPase) in circulating monocytes and pulmonary arterial (PA) adventitial macrophages of patients with PA hypertension (PAH). Furthermore, recombinant HERV-K dUTPase increased IL-6 in PA endothelial cells (PAECs) and caused pulmonary hypertension in rats. Here we show that monocytes overexpressing HERV-K dUTPase, as opposed to GFP, can release HERV-K dUTPase in extracellular vesicles (EVs) that cause pulmonary hypertension in mice in association with endothelial mesenchymal transition (EndMT) related to induction of SNAIL/SLUG and proinflammatory molecules IL-6 as well as VCAM1. In PAECs, HERV-K dUTPase requires TLR4-myeloid differentiation primary response-88 to increase IL-6 and SNAIL/SLUG, and HERV-K dUTPase interaction with melanoma cell adhesion molecule (MCAM) is necessary to upregulate VCAM1. TLR4 engagement induces p-p38 activation of NF-κB in addition to p-pSMAD3 required for SNAIL and pSTAT1 for IL-6. HERV-K dUTPase interaction with MCAM also induces p-p38 activation of NF-κB in addition to pERK1/2-activating transcription factor-2 (ATF2) to increase VCAM1. Thus in PAH, monocytes or macrophages can release HERV-K dUTPase in EVs, and HERV-K dUTPase can engage dual receptors and signaling pathways to subvert PAEC transcriptional machinery to induce EndMT and associated proinflammatory molecules.


Asunto(s)
Retrovirus Endógenos , Transición Epitelial-Mesenquimal/inmunología , Hipertensión Pulmonar , Macrófagos/inmunología , Monocitos/inmunología , Arteria Pulmonar , Pirofosfatasas/metabolismo , Animales , Antígeno CD146/metabolismo , Retrovirus Endógenos/metabolismo , Retrovirus Endógenos/patogenicidad , Células Endoteliales/metabolismo , Hipertensión Pulmonar/inmunología , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/virología , Inflamación/metabolismo , Inflamación/virología , Ratones , Arteria Pulmonar/inmunología , Arteria Pulmonar/patología , Transducción de Señal , Factores de Transcripción de la Familia Snail/metabolismo
6.
Biologicals ; 71: 1-8, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34039532

RESUMEN

Xenogenic cell-based therapeutic products are expected to alleviate the chronic shortage of human donor organs. For example, porcine islet cell products are currently under development for the treatment of human diabetes. As porcine cells possess endogenous retrovirus (PERV), which can replicate in human cells in vitro, the potential transmission of PERV has raised concerns in the case of products that use living pig cells as raw materials. Although several PERV sequences exist in the porcine genome, not all have the ability to infect human cells. Therefore, polymerase chain reaction analysis, which amplifies a portion of the target gene, may not accurately assess the infection risk. Here, we determined porcine genome sequences and evaluated the infectivity of PERVs using high-throughput sequencing technologies. RNA sequencing was performed on both PERV-infected human cells and porcine cells, and reads mapped to PERV sequences were examined. The normalized number of the reads mapped to PERV regions was able to predict the infectivity of PERVs, indicating that it would be useful for evaluation of the PERV infection risk prior to transplantation of porcine products.


Asunto(s)
Retrovirus Endógenos , Gammaretrovirus , Secuenciación de Nucleótidos de Alto Rendimiento , Animales , Retrovirus Endógenos/genética , Retrovirus Endógenos/patogenicidad , Gammaretrovirus/genética , Gammaretrovirus/patogenicidad , Islotes Pancreáticos/virología , Porcinos , Trasplante Heterólogo
7.
Retrovirology ; 18(1): 10, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33879155

RESUMEN

High quality reference genomes have facilitated the study of endogenous retroviruses (ERVs). However, there are an increasing number of published works which assume the ERVs in reference genomes are universal; even those of evolutionarily recent integrations. Consequently, these studies fail to properly characterise polymorphic ERVs, and even propose biological functions for ERVs that may not actually be present in the genomes of interest. Here, I outline the pitfalls of three studies of chicken endogenous Avian Leukosis Viruses (ALVEs or "ev genes": the "original" ERVs), all confounded by the assumption that the reference genome provides a representative ALVE baseline.


Asunto(s)
Pollos/genética , Retrovirus Endógenos/genética , Genoma , Animales , Leucosis Aviar/genética , Virus de la Leucosis Aviar/genética , Retrovirus Endógenos/patogenicidad
8.
J Integr Neurosci ; 20(1): 233-238, 2021 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-33834708

RESUMEN

The pathogenesis of multiple sclerosis (MS) remains poorly understood. Presumably, MS is caused by multiple environmental, epigenetic, and genetic factors. Among them, human endogenous retroviruses (HERVs), Epstein-Barr virus (EBV) and vitamin D have been suggested to play a role in the pathogenesis and course of MS. Because vitamin D can affect the immune system and infections, it can be hypothesized that there is a close interplay between vitamins, EBV and ERV in the pathogenesis of MS. Here, we summarize the important data on vitamin D, including polymorphisms in genes related to vitamin D metabolism, EBV and ERV, in the pathogenesis of MS and create hypotheses regarding their interactions. Data indicate that vitamin D has a strong impact on viral infections and interferes with EBV infection, while EBV is capable of activating silent ERVs. We believe that EBV could be the missing link between vitamin D and ERV in MS pathogenesis.


Asunto(s)
Retrovirus Endógenos/patogenicidad , Herpesvirus Humano 4/patogenicidad , Esclerosis Múltiple , Vitamina D/metabolismo , Humanos , Esclerosis Múltiple/etiología , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/virología
9.
APMIS ; 129(5): 241-253, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33683784

RESUMEN

Human endogenous retroviruses (HERVs) are remnants of ancient retroviral germline infections. Most HERV sequences are silenced in somatic cells, but interest is emerging on the involvement of HERV derived transcripts and proteins in human physiology and disease. A HERV-W encoded protein, syncytin-1, has been co-opted into fetal physiology, where it plays a role in trophoblast formation. Altered HERV transcription and expression of HERV derived proteins are associated with various cancer types and neurological diseases such as multiple sclerosis (MS). The implication of HERVs as potential mediators of both health and disease suggests important roles of regulatory mechanisms and alterations of these in physiological and pathological processes. The regulation of HERV sequences is mediated by a wide variety of mechanisms, and the focus of this review is on selected aspects of these, including epigenetic mechanisms such as CpG methylation and histone modifications of the HP1-H3K9me axis, viral transactivation events, and regulatory perspectives of transient stimuli in the microenvironment. Increasing knowledge of the regulation of HERV sequences will not only contribute to the understanding of complex pathogeneses, but also may pinpoint potential targets for better diagnosis and treatment in complex diseases as MS.


Asunto(s)
Retrovirus Endógenos/genética , Desarrollo Fetal/genética , Regulación Viral de la Expresión Génica , Neoplasias/patología , Enfermedades del Sistema Nervioso/patología , Retrovirus Endógenos/patogenicidad , Epigénesis Genética , Productos del Gen env/fisiología , Humanos , Inmunidad/genética , Inmunidad/inmunología , Neoplasias/genética , Neoplasias/inmunología , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/inmunología , Proteínas Gestacionales/fisiología , Activación Transcripcional
10.
Cancer Lett ; 507: 26-38, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33713739

RESUMEN

Early events in an experimental model of mesothelioma development include increased levels of editing in double-stranded RNA (dsRNA). We hypothesised that expression of endogenous retroviruses (ERV) contributes to dsRNA formation and type-I interferon signaling. ERV and interferon stimulated genes (ISGs) expression were significantly higher in tumor compared to non-tumor samples. 12 tumor specific ERV ("MesoERV1-12") were identified and verified by qPCR in mouse tissues. "MesoERV1-12" expression was lower in mouse embryonic fibroblasts (MEF) compared to mesothelioma cells. "MesoERV1-12" levels were significantly increased by demethylating agent 5-Aza-2'-deoxycytidine treatment and were accompanied by increased levels of dsRNA and ISGs. Basal ISGs expression was higher in mesothelioma cells compared to MEF and was significantly decreased by JAK inhibitor Ruxolitinib, by blocking Ifnar1 and by silencing Mavs. "MesoERV7" promoter was demethylated in asbestos-exposed compared to sham mice tissue as well as in mesothelioma cells and MEF upon 5-Aza-CdR treatment. These observations uncover novel aspects of asbestos-induced mesothelioma whereby ERV expression increases due to promoter demethylation and is paralleled by increased levels of dsRNA and activation of type-I IFN signaling. These features are important for early diagnosis and therapy.


Asunto(s)
Retrovirus Endógenos/patogenicidad , Interferón Tipo I/metabolismo , Mesotelioma/virología , Edición de ARN , ARN Bicatenario/metabolismo , Animales , Asbesto Crocidolita , Asbestosis/complicaciones , Línea Celular Tumoral , Metilación de ADN , Modelos Animales de Enfermedad , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Regulación Neoplásica de la Expresión Génica , Interacciones Huésped-Patógeno , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferón Tipo I/genética , Mesotelioma/etiología , Mesotelioma/genética , Mesotelioma/metabolismo , Ratones , Regiones Promotoras Genéticas , ARN Bicatenario/genética , Transducción de Señal
11.
J Mol Biol ; 432(24): 166711, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33197463

RESUMEN

Expression of the Human Endogenous Retrovirus Type K (HERV-K), the youngest and most active HERV, has been associated with various cancers and neurodegenerative diseases. As in all retroviruses, a fraction of HERV-K transcripts is exported from the nucleus in unspliced or incompletely spliced forms to serve as templates for translation of viral proteins. In a fraction of HERV-K loci (Type 2 proviruses), nuclear export of the unspliced HERV-K mRNA appears to be mediated by a cis-acting signal on the mRNA, the RcRE, and the protein Rec-these are analogous to the RRE-Rev system in HIV-1. Interestingly, the HIV-1 Rev protein is able to mediate the nuclear export of the HERV-K RcRE, contributing to elevated HERV-K expression in HIV-infected patients. We aimed to understand the structural basis for HIV Rev-HERV-K RcRE recognition. We examined the conformation of the RcRE RNA in solution using small-angle X-ray scattering (SAXS) and atomic force microscopy (AFM). We found that the 433-nt long RcRE can assume folded or extended conformations as observed by AFM. SAXS analysis of a truncated RcRE variant revealed an "A"-shaped topological structure similar to the one previously reported for the HIV-1 RRE. The effect of the overall topology was examined using several deletion variants. SAXS and biochemical analyses demonstrated that the "A" shape is necessary for efficient Rev-RcRE complex formation in vitro and nuclear export activity in cell culture. The findings provide insight into the mechanism of HERV-K expression and a structural explanation for HIV-1 Rev-mediated expression of HERV-K in HIV-infected patients. IMPORTANCE: Expression of the human endogenous retrovirus type K (HERV-K) has been associated with various cancers and autoimmune diseases. Nuclear export of both HIV-1 and HERV-K mRNAs is dependent on the interaction between a small viral protein (Rev in HIV-1 and Rec in HERV-K) and a region on the mRNA (RRE in HIV-1 and RcRE in HERV-K). HIV-1 Rev is able to mediate the nuclear export of RcRE-containing HERV-K mRNAs, which contributes to elevated production of HERV-K proteins in HIV-infected patients. We report the solution conformation of the RcRE RNA-the first three-dimensional topological structure for a HERV molecule-and find that the RcRE resembles the HIV-1 nuclear export signal, RRE. The finding reveals the structural basis for the increased HERV-K expression observed in HIV-infected patients. Elevated HERV expression, mediated by HIV infection or other stressors, can have various HERV-related biological consequences. The findings provide structural insight for regulation of HERV-K expression.


Asunto(s)
Retrovirus Endógenos/genética , Infecciones por VIH/genética , VIH-1/genética , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/genética , Transporte Activo de Núcleo Celular/genética , Sitios de Unión/genética , Núcleo Celular/genética , Núcleo Celular/ultraestructura , Retrovirus Endógenos/patogenicidad , Retrovirus Endógenos/ultraestructura , Regulación Viral de la Expresión Génica/genética , Infecciones por VIH/virología , VIH-1/patogenicidad , Humanos , ARN Viral/genética , Elementos de Respuesta/genética , Dispersión del Ángulo Pequeño , Difracción de Rayos X , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/ultraestructura
12.
Viruses ; 12(8)2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32759845

RESUMEN

Animal retroviruses are known for their transforming potential, and this is also true for the ones hosted by humans, which have gathered expanding attention as one of the potent causative agents in various disease, including specific cancer types. For instance, Human T Lymphotropic virus (HTLV) is a well-studied class of oncoviruses causing T cell leukemia, while human immunodeficiency virus (HIV) leads to acquired immunodeficiency syndrome (AIDS), which is linked to a series of defining cancers including Kaposi sarcoma, certain types of non-Hodgkin lymphoma, and cervical cancer. Of note, in addition to these "modern" exogenous retroviruses, our genome harbors a staggering number of human endogenous retroviruses (HERVs). HERVs are the genetic remnants of ancient retroviral germline infection of human ancestors and are typically silenced in normal tissues due to inactivating mutations and sequence loss. While some HERV elements have been appropriated and contribute to human physiological functions, others can be reactivated through epigenetic dysregulations to express retroviral elements and promote carcinogenesis. Conversely, HERV replication intermediates or protein products can also serve as intrinsic pathogen-associated molecular patterns that cause the immune system to interpret it as an exogenous infection, thereby stimulating immune responses against tumors. As such, HERVs have also been targeted as a potential internal strategy to sensitize tumor cells for promising immunotherapies. In this review, we discuss the dynamic role of human retroviruses in cancer development, focusing on HIV and HERVs contribution. We also describe potential treatment strategies, including immunotherapeutic targeting of HERVs, inhibiting DNA methylation to expose HERV signatures, and the use of antiretroviral drugs against HIV and HERVs, which can be employed as prospective anti-cancer modalities.


Asunto(s)
Retrovirus Endógenos/patogenicidad , Infecciones por VIH/virología , Inmunoterapia/métodos , Neoplasias/virología , Infecciones por Retroviridae/patología , Animales , Antirretrovirales/uso terapéutico , Metilación de ADN , Retrovirus Endógenos/genética , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Humanos , Ratones , Neoplasias/inmunología , Estudios Prospectivos
13.
Genes Genomics ; 42(10): 1145-1150, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32785870

RESUMEN

BACKGROUND: Human endogenous retroviruses (HERVs) constitute around 8% of the human genome and have important roles in human health and disease, including cancers. Previous studies showed that HERV envelope (Env) proteins are highly expressed in cancer tissues and co-related with cancer progression. KAP1 has been reported to play a key role in regulating retrotransposons, including HERV-K, through epigenetic silencing. OBJECTIVE: The relationship between KAP-1 and HERV Envs expressions was analyzed only in tumor cell lines and has not yet been studied in cancer tissues. In this study, we analyzed the expression patterns and relationship between KAP1 and HERV Env proteins in ovarian cancer tissues. METHOD: The expression patterns of KAP-1 and HERV Env proteins, including HERV-K and HERV-R, were analyzed in ovarian cancer tissue microarrays that contained 80 surgical specimens, including normal ovary and malignant ovarian cancers. RESULTS: The expression of HERV-R Env and KAP1 proteins is significantly higher in ovarian cancer compared with normal ovary tissues. However, the expression of HERV-K Env did not change significantly in cancer tissues. The expression patterns of HERV-K Env and HERV-R Env significantly increased in early stages of cancer and KAP1 expression was higher in certain stage and types of cancers. However, the expression of HERV-K Env, HERV-R Env, and KAP1 did not change in different age groups. The correlation between the expression of KAP1 and HERV-Env, including HERV-K and HERV-R, was not significantly correlated. CONCLUSIONS: The results of this study showed that there was no significant correlation between the expression of KAP1 and HERV Env proteins in ovarian cancer tissues, unlike studies with cell lines in vitro. These results suggest that the actual expression of HERV Env proteins in ovarian cancer tissues may be regulated through various complex factors as well as KAP1.


Asunto(s)
Productos del Gen env/genética , Neoplasias Ováricas/genética , Proteína 28 que Contiene Motivos Tripartito/genética , Anciano , Línea Celular Tumoral , Retrovirus Endógenos/genética , Retrovirus Endógenos/patogenicidad , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Regulación Viral de la Expresión Génica/genética , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/patología , Neoplasias Ováricas/virología , Análisis de Matrices Tisulares
14.
Cells ; 9(7)2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32629888

RESUMEN

Motor neuron degeneration and spinal cord demyelination are hallmark pathological events in Amyotrophic Lateral Sclerosis (ALS). Endogenous retrovirus-K (ERVK) expression has an established association with ALS neuropathology, with murine modeling pointing to a role for the ERVK envelope (env) gene in disease processes. Here, we describe a novel viral protein cryptically encoded within the ERVK env transcript, which resembles two distinct cysteine-rich neurotoxic proteins: conotoxin proteins found in marine snails and the Human Immunodeficiency Virus (HIV) Tat protein. Consistent with Nuclear factor-kappa B (NF-κB)-induced retrotransposon expression, the ERVK conotoxin-like protein (CTXLP) is induced by inflammatory signaling. CTXLP is found in the nucleus, impacting innate immune gene expression and NF-κB p65 activity. Using human autopsy specimens from patients with ALS, we further showcase CTXLP expression in degenerating motor cortex and spinal cord tissues, concomitant with inflammation linked pathways, including enhancement of necroptosis marker mixed lineage kinase domain-like (MLKL) protein and oligodendrocyte maturation/myelination inhibitor Nogo-A. These findings identify CTXLP as a novel ERVK protein product, which may act as an effector in ALS neuropathology.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Conotoxinas/genética , Conotoxinas/metabolismo , Retrovirus Endógenos/metabolismo , Retrovirus Endógenos/patogenicidad , Humanos , FN-kappa B/metabolismo , Necroptosis/genética , Necroptosis/fisiología , Retroviridae/genética , Retroviridae/patogenicidad
15.
Viruses ; 12(8)2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32718022

RESUMEN

Endogenous retroviruses (ERVs) in mammals are closely related to infectious retroviruses and utilize host tRNAs as a primer for reverse transcription and replication, a hallmark of long terminal repeat (LTR) retroelements. Their dependency on tRNA makes these elements vulnerable to targeting by small RNAs derived from the 3'-end of mature tRNAs (3'-tRFs), which are highly expressed during epigenetic reprogramming and potentially protect many tissues in eukaryotes. Here, we review some key functions of ERV reprogramming during mouse and human development and discuss how small RNA-mediated silencing maintains genome stability when ERVs are temporarily released from heterochromatin repression. In particular, we take a closer look at the tRNA primer binding sites (PBS) of two highly active ERV families in mice and their sequence variation that is shaped by the conflict of successful tRNA priming for replication versus evasion of silencing by 3'-tRFs.


Asunto(s)
Retrovirus Endógenos/genética , Retrovirus Endógenos/patogenicidad , Silenciador del Gen , Interacciones Microbiota-Huesped , ARN de Transferencia/genética , Animales , Sitios de Unión , VIH/genética , Humanos , Ratones , Retroelementos , Infecciones por Retroviridae/virología , Secuencias Repetidas Terminales
16.
Xenotransplantation ; 27(2): e12563, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31667881

RESUMEN

The infection of human transplant recipients by porcine endogenous retrovirus (PERV) is a safety issue for xenotransplantation (XTx). CRISPR/Cas9 technology has enabled the generation of pigs free of functional PERVs, and the susceptibility of these animals to reinfection by PERVs remains unclear. To assess virological safety, we characterized a cell line in which PERVs have been inactivated by CRISPR/Cas9 (PK15 clone 15) for its susceptibility to infectious PERV. First, basal expression of PERV pol, the porcine PERV-A receptor (POPAR), and reverse transcriptase (RT) activity of PERV were determined. PK15 clone 15 cells were inoculated with PERV and monitored post infection for virus expression and RT activity. Particles were visualized by electron microscopy. Our data show that PK15 clone 15 cells still produce viral proteins that assemble to produce impaired viral particles. These virions have an irregular morphology that diverges from that of mature wild type. The particles are no longer infectious when tested in a downstream infection assay using supernatants of PK15 clone 15 cells to infect susceptible swine testis-IOWA (ST-IOWA) cells. The expression of POPAR was quantified to exclude the possibility that lack of susceptibility to reinfection, for PERV-A, is caused by absence of viral host receptor(s). PK15 and PK15 clone 15 cells do, in fact, express POPAR equally. PERV RT inactivation mediated by CRISPR/Cas9 does not compromise virus assembly but affects virion structure and proviral integration. The constitutive virion production seems to maintain cellular resistance to superinfection and possibly indicates a protective side effect of this specific CRISPR/Cas9 mediated RT inactivation.


Asunto(s)
Sistemas CRISPR-Cas/fisiología , Retrovirus Endógenos/patogenicidad , Provirus/patogenicidad , Porcinos Enanos/virología , Animales , Línea Celular , Humanos , Porcinos , Trasplante Heterólogo/efectos adversos
17.
Xenotransplantation ; 27(2): e12565, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31692126

RESUMEN

BACKGROUND: Decellularized human pulmonary heart valve (dhHV) scaffolds have been shown to be the gold standard especially for younger, adolescent patients. However, human heart valves are limited in availability. Xenogeneic decellularized pig heart valves (dpHV) may serve as alternative. METHODS: The efficacy of DNA reduction processes upon decellularization of heart valves from German Landrace pigs was analyzed by measurements of remaining nucleic acids including proviral porcine endogenous retrovirus (PERV) sequences. Porcine pulmonary heart valves (pPHV) were decellularized by three different protocols and further treated with DNaseI or Benzonase, at varying incubation times. DNA isolated from valve associated muscle (m), valve cusp (c), and pulmonary artery (pa) was monitored by PCR and qRT-PCR using GAPDH and the PERV polymerase (pol) for read-out. RESULTS: Decellularization of pPHV led to a significant reduction of DNA (>99%) which could be further significantly increased for (m) and (pa) by nuclease treatment, reducing proviral PERV pol from approximately 5 × 107 to 5 × 103  copies/mg in nuclease treated tissues. CONCLUSIONS: Both nucleases demonstrated comparable activities. But DNaseI revealed to be less consistent for PERV, especially at muscular tissue. Noteworthy, remaining proviral sequences are still detectable by PCR; however, due to the absence of the cellular replication machinery the production of infectious particles is not expected. Decellularization and nuclease treatment of pPHV is an efficient procedure to reduce the DNA content including PERV, thus represents a valuable option to increase virus safety independently from the source animal background.


Asunto(s)
Retrovirus Endógenos/patogenicidad , Prótesis Valvulares Cardíacas/virología , Válvulas Cardíacas/patología , Ácidos Nucleicos/metabolismo , Provirus/patogenicidad , Animales , Bioprótesis/efectos adversos , Línea Celular , Porcinos , Trasplante Heterólogo/efectos adversos
18.
Int J Mol Sci ; 20(23)2019 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-31801288

RESUMEN

Human endogenous retroviruses (HERVs) are genetic elements resulting from relics of ancestral infection of germline cells, now recognized as cofactors in the etiology of several complex diseases. Here we present a review of findings supporting the role of the abnormal HERVs activity in neurodevelopmental disorders. The derailment of brain development underlies numerous neuropsychiatric conditions, likely starting during prenatal life and carrying on during subsequent maturation of the brain. Autism spectrum disorders, attention deficit hyperactivity disorders, and schizophrenia are neurodevelopmental disorders that arise clinically during early childhood or adolescence, currently attributed to the interplay among genetic vulnerability, environmental risk factors, and maternal immune activation. The role of HERVs in human embryogenesis, their intrinsic responsiveness to external stimuli, and the interaction with the immune system support the involvement of HERVs in the derailed neurodevelopmental process. Although definitive proofs that HERVs are involved in neurobehavioral alterations are still lacking, both preclinical models and human studies indicate that the abnormal expression of ERVs could represent a neurodevelopmental disorders-associated biological trait in affected individuals and their parents.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/virología , Trastorno del Espectro Autista/virología , Encéfalo/virología , Retrovirus Endógenos/genética , Efectos Tardíos de la Exposición Prenatal/virología , Esquizofrenia/virología , Adolescente , Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno por Déficit de Atención con Hiperactividad/metabolismo , Trastorno por Déficit de Atención con Hiperactividad/patología , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/patología , Encéfalo/inmunología , Encéfalo/patología , Niño , Embrión de Mamíferos , Desarrollo Embrionario/genética , Desarrollo Embrionario/inmunología , Retrovirus Endógenos/patogenicidad , Exposición a Riesgos Ambientales/efectos adversos , Femenino , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Humanos , Inmunidad Innata , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología , Esquizofrenia/genética , Esquizofrenia/metabolismo , Esquizofrenia/patología
19.
Curr Diab Rep ; 19(12): 141, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31754894

RESUMEN

PURPOSE OF THE REVIEW: The aim of this review is to discuss recent data pointing at an involvement of human endogenous retroviruses (HERVs) in type 1 diabetes (T1D) onset and progression. RECENT FINDINGS: The envelope protein of HERV-W family, named HERV-W-Env, was detected in pancreata from T1D patients and was shown to display pro-inflammatory properties and direct toxicity toward pancreatic beta cells. The etiopathogenesis of T1D remains elusive, even if conventional environmental viral infections have been recurrently involved. Nonetheless, a new category of pathogens may provide the missing link between genetic susceptibility and environmental factors long thought to contribute to T1D onset. A number of studies have now shown that HERV sequences, which are normally inactivated or repressed in the human genome, could be activated by environmental viruses. Thus, if similarly activated by viruses associated with T1D, disregarded HERV genes may underlie T1D genetic susceptibility. Moreover, once expressed, HERV elements may display broad pathogenic properties, which identify them as potential new therapeutic targets.


Asunto(s)
Diabetes Mellitus Tipo 1/etiología , Retrovirus Endógenos/fisiología , Productos del Gen env/aislamiento & purificación , Células Secretoras de Insulina/virología , Activación Viral/fisiología , Animales , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/fisiopatología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/virología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Retrovirus Endógenos/aislamiento & purificación , Retrovirus Endógenos/patogenicidad , Epigénesis Genética , Interacción Gen-Ambiente , Humanos , Ratones
20.
Xenotransplantation ; 26(6): e12541, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31342571

RESUMEN

BACKGROUND: Xenotransplantation of porcine tissues raises concerns, especially in the context of the potential interspecies transmission of porcine endogenous retroviruses (PERVs). To date, the possibility of PERV infections of various human cells has been confirmed in vitro. PERVs infect cells coupling viral Env protein with adequate functional receptor on the surface of the host cell. So far, two PERV-A receptors have been described in humans: HuPAR-1 and HuPAR-2. TFAP-2C was described as one of the transcription factors engaged in the expression of HuPAR-2. METHODS: Bacterial LPS, well known as a strong inflammation inducer, was used in this study to stimulate changes of the expression profile of inflammation-related genes in human cells in vitro. The aim of the study was to investigate the expression profile of HuPAR-1 and HuPAR-2 and TFAP-2C genes in human NHDF cells treated with LPS and/or infected with PERVs from PK15 cells. PERV infection and expression was confirmed by qPCR and RTqPCR. The expression of HuPAR-1, HuPAR-2, and TFAP-2C genes was studied using HGU 133A 2.0 microarrays and RTqPCR. RESULTS: NHDF cells expressed both HuPAR-1 and HuPAR-2 genes with a higher expression of HuPAR-1. LPS down-regulated the expression of HuPAR-1 and TFAP-2C in NHDF cells, but had no effect on HuPAR-2 expression. These changes induced by LPS were more pronounced in the presence of PERV infection. CONCLUSION: As reported previously, treatment of NHDF cells with LPS decreased PERV-A provirus integration and increased PERV-A mRNA expression. PERV infection alone did not modulate the expression of HuPAR-1, HuPAR-2, and TFAP-2C. This is the first study analyzing the expression profile of HuPAR-1, HuPAR-2, and TFAP-2C in NHDF cells treated by LPS and/or infected by PERVs.


Asunto(s)
Retrovirus Endógenos/patogenicidad , Fibroblastos/virología , Factores de Transcripción/metabolismo , Virosis/virología , Animales , Línea Celular , Humanos , Trasplante Heterólogo/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...