Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cell Res Ther ; 15(1): 10, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38167524

RESUMEN

BACKGROUND: Beyond the observed alterations in cellular structure and mitochondria, the mechanisms linking rare genetic mutations to the development of heart failure in patients affected by desmin mutations remain unclear due in part, to the lack of relevant human cardiomyocyte models. METHODS: To shed light on the role of mitochondria in these mechanisms, we investigated cardiomyocytes derived from human induced pluripotent stem cells carrying the heterozygous DESE439K mutation that were either isolated from a patient or generated by gene editing. To increase physiological relevance, cardiomyocytes were either cultured on an anisotropic micropatterned surface to obtain elongated and aligned cardiomyocytes, or as a cardiac spheroid to create a micro-tissue. Moreover, when applicable, results from cardiomyocytes were confirmed with heart biopsies of suddenly died patient of the same family harboring DESE439K mutation, and post-mortem heart samples from five control healthy donors. RESULTS: The heterozygous DESE439K mutation leads to dramatic changes in the overall cytoarchitecture of cardiomyocytes, including cell size and morphology. Most importantly, mutant cardiomyocytes display altered mitochondrial architecture, mitochondrial respiratory capacity and metabolic activity reminiscent of defects observed in patient's heart tissue. Finally, to challenge the pathological mechanism, we transferred normal mitochondria inside the mutant cardiomyocytes and demonstrated that this treatment was able to restore mitochondrial and contractile functions of cardiomyocytes. CONCLUSIONS: This work highlights the deleterious effects of DESE439K mutation, demonstrates the crucial role of mitochondrial abnormalities in the pathophysiology of desmin-related cardiomyopathy, and opens up new potential therapeutic perspectives for this disease.


Asunto(s)
Cardiomiopatías , Células Madre Pluripotentes Inducidas , Humanos , Desmina/genética , Desmina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Cardiomiopatías/metabolismo , Mutación/genética , Miocitos Cardíacos/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo
2.
J Infect Dis ; 228(10): 1421-1429, 2023 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-37224627

RESUMEN

BACKGROUND: On May 6, 2022, a powerful outbreak of monkeypox virus (MPXV) had been reported outside of Africa, with many continuing new cases being reported around the world. Analysis of mutations among the 2 different lineages present in the 2021 and 2022 outbreaks revealed the presence of G->A mutations occurring in the 5'GpA context, indicative of APOBEC3 cytidine deaminase activity. METHODS: By using a sensitive polymerase chain reaction (differential DNA denaturation PCR) method allowing differential amplification of AT-rich DNA, we analyzed the level of APOBEC3-induced MPXV editing in infected cells and in patients. RESULTS: We demonstrate that G->A hypermutated MPXV genomes can be recovered experimentally from APOBEC3 transfection followed by MPXV infection. Here, among the 7 human APOBEC3 cytidine deaminases (A3A-A3C, A3DE, A3F-A3H), only APOBEC3F was capable of extensively deaminating cytidine residues in MPXV genomes. Hyperedited genomes were also recovered in ∼42% of analyzed patients. Moreover, we demonstrate that substantial repair of these mutations occurs. Upon selection, corrected G->A mutations escaping drift loss contribute to the MPXV evolution observed in the current epidemic. CONCLUSIONS: Stochastic or transient overexpression of the APOBEC3F gene exposes the MPXV genome to a broad spectrum of mutations that may be modeling the mutational landscape after multiple cycles of viral replication.


Asunto(s)
Citidina Desaminasa , Monkeypox virus , Humanos , Monkeypox virus/genética , Citidina Desaminasa/genética , Mutación , Brotes de Enfermedades , Citidina , Citosina Desaminasa/química , Citosina Desaminasa/genética
3.
PLoS Pathog ; 19(2): e1011156, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36745676

RESUMEN

Human adenoviruses (HAdVs) are a large family of DNA viruses counting more than a hundred strains divided into seven species (A to G). HAdVs induce respiratory tract infections, gastroenteritis and conjunctivitis. APOBEC3B is a cytidine deaminase that restricts several DNA viruses. APOBEC3B is also implicated in numerous cancers where it is responsible for the introduction of clustered mutations into the cellular genome. In this study, we demonstrate that APOBEC3B is an adenovirus restriction factor acting through a deaminase-dependent mechanism. APOBEC3B introduces C-to-T clustered mutations into the adenovirus genome. APOBEC3B reduces the propagation of adenoviruses by limiting viral genome replication, progression to late phase, and production of infectious virions. APOBEC3B restriction efficiency varies between adenoviral strains, the A12 strain being more sensitive to APOBEC3B than the B3 or C2 strains. In A12-infected cells, APOBEC3B clusters in the viral replication centers. Importantly, we show that adenovirus infection leads to a reduction of the quantity and/or enzymatic activity of the APOBEC3B protein depending on the strains. The A12 strain seems less able to resist APOBEC3B than the B3 or C2 strains, a characteristic which could explain the strong depletion of the APOBEC3-targeted motifs in the A12 genome. These findings suggest that adenoviruses evolved different mechanisms to antagonize APOBEC3B. Elucidating these mechanisms could benefit the design of cancer treatments. This study also identifies adenoviruses as triggers of the APOBEC3B-mediated innate response. The involvement of certain adenoviral strains in the genesis of the APOBEC3 mutational signature observed in tumors deserves further study.


Asunto(s)
Infecciones por Adenoviridae , Neoplasias , Humanos , Adenoviridae/genética , Adenoviridae/metabolismo , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Proteínas/metabolismo , Neoplasias/patología , Antígenos de Histocompatibilidad Menor/genética
4.
PLoS Pathog ; 19(2): e1011170, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36802406

RESUMEN

Viruses have evolved countless mechanisms to subvert and impair the host innate immune response. Measles virus (MeV), an enveloped, non-segmented, negative-strand RNA virus, alters the interferon response through different mechanisms, yet no viral protein has been described as directly targeting mitochondria. Among the crucial mitochondrial enzymes, 5'-aminolevulinate synthase (ALAS) is an enzyme that catalyzes the first step in heme biosynthesis, generating 5'-aminolevulinate from glycine and succinyl-CoA. In this work, we demonstrate that MeV impairs the mitochondrial network through the V protein, which antagonizes the mitochondrial enzyme ALAS1 and sequesters it to the cytosol. This re-localization of ALAS1 leads to a decrease in mitochondrial volume and impairment of its metabolic potential, a phenomenon not observed in MeV deficient for the V gene. This perturbation of the mitochondrial dynamics demonstrated both in culture and in infected IFNAR-/- hCD46 transgenic mice, causes the release of mitochondrial double-stranded DNA (mtDNA) in the cytosol. By performing subcellular fractionation post infection, we demonstrate that the most significant source of DNA in the cytosol is of mitochondrial origin. Released mtDNA is then recognized and transcribed by the DNA-dependent RNA polymerase III. The resulting double-stranded RNA intermediates will be captured by RIG-I, ultimately initiating type I interferon production. Deep sequencing analysis of cytosolic mtDNA editing divulged an APOBEC3A signature, primarily analyzed in the 5'TpCpG context. Finally, in a negative feedback loop, APOBEC3A an interferon inducible enzyme will orchestrate the catabolism of mitochondrial DNA, decrease cellular inflammation, and dampen the innate immune response.


Asunto(s)
Interferones , Mitocondrias , Ratones , Animales , Mitocondrias/metabolismo , Virus del Sarampión , 5-Aminolevulinato Sintetasa/genética , 5-Aminolevulinato Sintetasa/metabolismo , ADN Mitocondrial
5.
J Infect Dis ; 226(5): 891-895, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-35022749

RESUMEN

Single-nucleotide polymorphism in APOBEC3C (resulting in a serine to isoleucine in position 188) is present in approximately 10% of African populations and greatly enhances restriction against human immunodeficiency virus-1 and simian immunodeficiency virus by improving dimerization and DNA processivity of the enzyme. In this study, we demonstrated in culture and in infected patients that hepatitis B virus (HBV) could be edited by APOBEC3CS188I. Using next-generation sequencing, we demonstrated that APOBEC3CS188I led to enhanced editing activity in 5'TpCpA→5'TpTpA context. This constitutes a new hallmark of this enzyme, which could be used to determine its impact on HBV or nuclear DNA.


Asunto(s)
Citidina Desaminasa , Genoma Viral , Virus de la Hepatitis B , Citidina Desaminasa/genética , Hepatitis B/genética , Virus de la Hepatitis B/genética , Humanos , Polimorfismo de Nucleótido Simple
6.
mBio ; 12(6): e0255721, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34809467

RESUMEN

Viruses have evolved a plethora of mechanisms to impair host innate immune responses. Herpes simplex virus type 1 (HSV-1), a double-stranded linear DNA virus, impairs the mitochondrial network and dynamics predominantly through the UL12.5 gene. We demonstrated that HSV-1 infection induced a remodeling of mitochondrial shape, resulting in a fragmentation of the mitochondria associated with a decrease in their volume and an increase in their sphericity. This damage leads to the release of mitochondrial DNA (mtDNA) to the cytosol. By generating a stable THP-1 cell line expressing the DNase I-mCherry fusion protein and a THP-1 cell line specifically depleted of mtDNA upon ethidium bromide treatment, we showed that cytosolic mtDNA contributes to type I interferon and APOBEC3A upregulation. This was confirmed by using an HSV-1 strain (KOS37 UL98-SPA) with a deletion of the UL12.5 gene that impaired its ability to induce mtDNA stress. Furthermore, by using an inhibitor of RNA polymerase III, we demonstrated that upon HSV-1 infection, cytosolic mtDNA enhanced type I interferon induction through the RNA polymerase III/RIG-I pathway. APOBEC3A was in turn induced by interferon. Deep sequencing analyses of cytosolic mtDNA mutations revealed an APOBEC3A signature predominantly in the 5'TpCpG context. These data demonstrate that upon HSV-1 infection, the mitochondrial network is disrupted, leading to the release of mtDNA and ultimately to its catabolism through APOBEC3-induced mutations. IMPORTANCE Herpes simplex virus 1 (HSV-1) impairs the mitochondrial network through the viral protein UL12.5. This leads to the fusion of mitochondria and simultaneous release of mitochondrial DNA (mtDNA) in a mouse model. We have shown that released mtDNA is recognized as a danger signal, capable of stimulating signaling pathways and inducing the production of proinflammatory cytokines. The expression of the human cytidine deaminase APOBEC3A is highly upregulated by interferon responses. This enzyme catalyzes the deamination of cytidine to uridine in single-stranded DNA substrates, resulting in the catabolism of edited DNA. Using human cell lines deprived of mtDNA and viral strains deficient in UL12, we demonstrated the implication of mtDNA in the production of interferon and APOBEC3A expression during viral infection. We have shown that HSV-1 induces mitochondrial network fragmentation in a human model and confirmed the implication of RNA polymerase III/RIG-I signaling in the capture of cytosolic mtDNA.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Herpes Simple/metabolismo , Herpesvirus Humano 1/fisiología , Interferón beta/metabolismo , Mitocondrias/virología , ARN Polimerasa III/metabolismo , Receptores Inmunológicos/metabolismo , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Proteína 58 DEAD Box/genética , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Desoxirribonucleasas/genética , Desoxirribonucleasas/metabolismo , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/genética , Interacciones Huésped-Patógeno , Humanos , Interferón beta/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas/genética , Proteínas/metabolismo , ARN Polimerasa III/genética , Receptores Inmunológicos/genética , Transducción de Señal , Proteínas Virales/genética , Proteínas Virales/metabolismo
7.
J Biol Chem ; 297(3): 101081, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34403699

RESUMEN

The human APOBEC3A (A3A) cytidine deaminase is a powerful DNA mutator enzyme recognized as a major source of somatic mutations in tumor cell genomes. However, there is a discrepancy between APOBEC3A mRNA levels after interferon stimulation in myeloid cells and A3A detection at the protein level. To understand this difference, we investigated the expression of two novel alternative "A3Alt" proteins encoded in the +1-shifted reading frame of the APOBEC3A gene. A3Alt-L and its shorter isoform A3Alt-S appear to be transmembrane proteins targeted to the mitochondrial compartment that induce membrane depolarization and apoptosis. Thus, the APOBEC3A gene represents a new example wherein a single gene encodes two proapoptotic proteins, A3A cytidine deaminases that target the genome and A3Alt proteins that target mitochondria.


Asunto(s)
Citidina Desaminasa/genética , Citidina Desaminasa/fisiología , Mitocondrias/genética , Proteínas/genética , Proteínas/fisiología , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Citidina Desaminasa/metabolismo , ADN/genética , Mutación del Sistema de Lectura/genética , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Genoma/genética , Humanos , Mitocondrias/metabolismo , Mutación/genética , Proteínas/metabolismo , ARN Mensajero/genética , Sistemas de Lectura/genética
8.
Sci Rep ; 11(1): 7893, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33846459

RESUMEN

APOBEC3 (A3) enzymes are best known for their role as antiviral restriction factors and as mutagens in cancer. Although four of them, A3A, A3B, A3F and A3G, are induced by type-1-interferon (IFN-I), their role in inflammatory conditions is unknown. We thus investigated the expression of A3, and particularly A3A and A3B because of their ability to edit cellular DNA, in Systemic Lupus Erythematosus (SLE), a chronic inflammatory disease characterized by high IFN-α serum levels. In a cohort of 57 SLE patients, A3A and A3B, but also A3C and A3G, were upregulated ~ 10 to 15-fold (> 1000-fold for A3B) compared to healthy controls, particularly in patients with flares and elevated serum IFN-α levels. Hydroxychloroquine, corticosteroids and immunosuppressive treatment did not reverse A3 levels. The A3AΔ3B polymorphism, which potentiates A3A, was detected in 14.9% of patients and in 10% of controls, and was associated with higher A3A mRNA expression. A3A and A3B mRNA levels, but not A3C or A3G, were correlated positively with dsDNA breaks and negatively with lymphopenia. Exposure of SLE PBMCs to IFN-α in culture induced massive and sustained A3A levels by 4 h and led to massive cell death. Furthermore, the rs2853669 A > G polymorphism in the telomerase reverse transcriptase (TERT) promoter, which disrupts an Ets-TCF-binding site and influences certain cancers, was highly prevalent in SLE patients, possibly contributing to lymphopenia. Taken together, these findings suggest that high baseline A3A and A3B levels may contribute to cell frailty, lymphopenia and to the generation of neoantigens in SLE patients. Targeting A3 expression could be a strategy to reverse cell death and the generation of neoantigens.


Asunto(s)
Desaminasas APOBEC/metabolismo , Lupus Eritematoso Sistémico/enzimología , Desaminasas APOBEC/genética , Adulto , Muerte Celular/efectos de los fármacos , Estudios de Cohortes , Femenino , Regulación Enzimológica de la Expresión Génica , Mutación de Línea Germinal/genética , Humanos , Interferón-alfa/farmacología , Lupus Eritematoso Sistémico/genética , Masculino , Polimorfismo de Nucleótido Simple/genética , Telomerasa/genética , Regulación hacia Arriba
9.
BMC Genomics ; 20(1): 858, 2019 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-31726973

RESUMEN

BACKGROUND: APOBEC1 (A1) enzymes are cytidine deaminases involved in RNA editing. In addition to this activity, a few A1 enzymes have been shown to be active on single stranded DNA. As two human ssDNA cytidine deaminases APOBEC3A (A3A), APOBEC3B (A3B) and related enzymes across the spectrum of placental mammals have been shown to introduce somatic mutations into nuclear DNA of cancer genomes, we explored the mutagenic threat of A1 cytidine deaminases to chromosomal DNA. RESULTS: Molecular cloning and expression of various A1 enzymes reveal that the cow, pig, dog, rabbit and mouse A1 have an intracellular ssDNA substrate specificity. However, among all the enzymes studied, mouse A1 appears to be singular, being able to introduce somatic mutations into nuclear DNA with a clear 5'TpC editing context, and to deaminate 5-methylcytidine substituted DNA which are characteristic features of the cancer related mammalian A3A and A3B enzymes. However, mouse A1 activity fails to elicit formation of double stranded DNA breaks, suggesting that mouse A1 possess an attenuated nuclear DNA mutator phenotype reminiscent of human A3B. CONCLUSIONS: At an experimental level mouse APOBEC1 is remarkable among 12 mammalian A1 enzymes in that it represents a source of somatic mutations in mouse genome, potentially fueling oncogenesis. While the order Rodentia is bereft of A3A and A3B like enzymes it seems that APOBEC1 may well substitute for it, albeit remaining much less active. This modifies the paradigm that APOBEC3 and AID enzymes are the sole endogenous mutator enzymes giving rise to off-target editing of mammalian genomes.


Asunto(s)
Desaminasas APOBEC-1/metabolismo , Cromosomas de los Mamíferos/genética , Mutación , Desaminasas APOBEC-1/química , Desaminasas APOBEC-1/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Roturas del ADN de Doble Cadena , ADN de Cadena Simple , Activación Enzimática , Expresión Génica , Ratones , Filogenia , Edición de ARN , Especificidad por Sustrato
10.
Nat Microbiol ; 4(7): 1196-1207, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30936483

RESUMEN

Numerous human APOBEC3 cytidine deaminases have proven to be, inter alia, host cell restriction factors for retroviruses and hepadnaviruses. Although they can bind to genomic RNA and become encapsidated, they are only catalytically active on single-stranded DNA. As there are many cellular deoxyribonucleases (DNases), we hypothesized that a parallel could be struck between APOBEC3 and DNases. For human hepatitis B virus (HBV), we show that DNase I can considerably reduce the virion genome copy number from a variety of transfected or infected cells. DNASE1 is overexpressed and encapsidated in HBV particles in vitro in hypoxic environments and in vivo in cirrhotic patient livers as well as in the serum of infected patients. The use of CoCl2 and dimethyloxalylglycine, mimetic agents used to induce hypoxia by inhibiting prolyl hydroxylase enzymes that stabilize hypoxia-inducible factor (HIF)-1α, showed that the formation of HIF-1α/HIF-1ß heterodimers results in the induction of DNASE1. Indeed, transfection with HIF-1α and HIF-1ß expression constructs upregulated DNASE1. These findings suggest that human DNase I can impact HBV replication through the catabolism of the DNA genome within the capsid. The activity of DNases in general may explain in part the high frequency of empty or 'light' hepatitis B virions observed in vivo.


Asunto(s)
Desoxirribonucleasa I/metabolismo , Virus de la Hepatitis B/fisiología , Hipoxia , Replicación Viral , Línea Celular , Cobalto/farmacología , ADN Viral/metabolismo , Desoxirribonucleasa I/genética , Expresión Génica , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hepatitis B/enzimología , Antígenos del Núcleo de la Hepatitis B/metabolismo , Humanos , Hipoxia/inducido químicamente , Factor 1 Inducible por Hipoxia/metabolismo , Cirrosis Hepática/enzimología , Mutación , Virión/metabolismo , Replicación Viral/efectos de los fármacos
11.
Sci Rep ; 9(1): 3109, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30816165

RESUMEN

Human cells are stressed by numerous mechanisms that can lead to leakage of mitochondrial DNA (mtDNA) to the cytoplasm and ultimately apoptosis. This agonist DNA constitutes a danger to the cell and is counteracted by cytoplasmic DNases and APOBEC3 cytidine deamination of DNA. To investigate APOBEC3 editing of leaked mtDNA to the cytoplasm, we performed a PCR analysis of APOBEC3 edited cytoplasmic mtDNA (cymtDNA) at the single cell level for primary CD4+ T cells and the established P2 EBV blast cell line. Up to 17% of primary CD4+ T cells showed signs of APOBEC3 edited cymtDNA with ~50% of all mtDNA sequences showing signs of APOBEC3 editing - between 1500-5000 molecules. Although the P2 cell line showed a much lower frequency of stressed cells, the number of edited mtDNA molecules in such cells was of the same order. Addition of the genotoxic molecules, etoposide or actinomycin D increased the number of cells showing APOBEC3 edited cymtDNA to around 40%. These findings reveal a very dynamic image of the mitochondrial network, which changes considerably under stress. APOBEC3 deaminases are involved in the catabolism of mitochondrial DNA to circumvent chronic immune stimulation triggered by released mitochondrial DNA from damaged cells.


Asunto(s)
Citidina Desaminasa/fisiología , Daño del ADN , ADN Mitocondrial/metabolismo , Mitocondrias/metabolismo , Desaminasas APOBEC , Animales , Linfocitos T CD4-Positivos , Células Cultivadas , Citosol/metabolismo , Dactinomicina/toxicidad , Etopósido/toxicidad , Fibroblastos , Humanos , Codorniz , Análisis de la Célula Individual/métodos
12.
Oncotarget ; 9(45): 27809-27822, 2018 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-29963239

RESUMEN

APOBEC3 are cytidine deaminases that convert cytidine to uridine residues. APOBEC3A and APOBEC3B enzymes able to target genomic DNA are involved in oncogenesis of a sizeable proportion of human cancers. While the APOBEC3 locus is conserved in mammals, it encodes from 1-7 genes. APOBEC3A is conserved in most mammals, although absent in pigs, cats and throughout Rodentia whereas APOBEC3B is restricted to the Primate order. Here we show that the rabbit APOBEC3 locus encodes two genes of which APOBEC3A enzyme is strictly orthologous to human APOBEC3A. The rabbit enzyme is expressed in the nucleus and the cytoplasm, it can deaminate cytidine, 5-methcytidine residues, nuclear DNA and induce double-strand DNA breaks. The rabbit APOBEC3A enzyme is negatively regulated by the rabbit TRIB3 pseudokinase protein which is guardian of genome integrity, just like its human counterpart. This indicates that the APOBEC3A/TRIB3 pair is conserved over approximately 100 million years. The rabbit APOBEC3A gene is widely expressed in rabbit tissues, unlike human APOBEC3A. These data demonstrate that rabbit could be used as a small animal model for studying APOBEC3 driven oncogenesis.

13.
Nucleic Acids Res ; 45(6): 3231-3241, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28100701

RESUMEN

Foreign and self-cytoplasmic DNA are recognized by numerous DNA sensor molecules leading to the production of type I interferons. Such DNA agonists should be degraded otherwise cells would be chronically stressed. Most human APOBEC3 cytidine deaminases can initiate catabolism of cytoplasmic mitochondrial DNA. Using the human myeloid cell line THP-1 with an interferon inducible APOBEC3A gene, we show that cytoplasmic DNA triggers interferon α and ß production through the RNA polymerase III transcription/RIG-I pathway leading to massive upregulation of APOBEC3A. By catalyzing C→U editing in single stranded DNA fragments, the enzyme prevents them from re-annealing so attenuating the danger signal. The price to pay is chromosomal DNA damage in the form of CG→TA mutations and double stranded DNA breaks which, in the context of chronic inflammation, could drive cells down the path toward cancer.


Asunto(s)
Citidina Desaminasa/biosíntesis , Roturas del ADN de Doble Cadena , ADN Mitocondrial/metabolismo , Línea Celular Tumoral , Cromosomas Humanos , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Citosol/metabolismo , Proteína 58 DEAD Box , ADN Mitocondrial/química , Humanos , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Interferón beta/fisiología , Proteínas/genética , Proteínas/metabolismo , ARN Polimerasa III/metabolismo , Receptores Inmunológicos , Transcripción Genética , Regulación hacia Arriba , Uracilo/metabolismo
14.
Retrovirology ; 13(1): 84, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27998270

RESUMEN

BACKGROUND: The replication of HBV involves the production of covalently closed circular DNA (cccDNA) from the HBV genome through the repair of virion relaxed circular DNA (rcDNA) in the virion. As cccDNA is the transcription template for HBV genomes, it needs to be eliminated from hepatocytes if the eradication of chronic HBV infection is to be achieved. PCR quantitation of cccDNA copy number is the technique of choice for evaluating the efficiency of treatment regimens. The PCR target commonly used to identify cccDNA spans the gapped region of rcDNA and is considered to accurately distinguish between cccDNA and rcDNA. There is however, a potentially confounding issue in that PCR can generate larger targets from collections of small DNA fragments, a phenomenon known as PCR recombination. RESULTS: The impact of PCR recombination towards the amplification of this cccDNA specific target was explored by mixing three marked, yet overlapping HBV DNA fragments. Thirteen of sixteen possible recombinants were identified by sequencing with frequencies ranging from 0.6 to 23%. To confirm this finding in vivo, HBV positive sera were treated with DNase I and submitted to quantitative real-time PCR. Under these conditions, it was possible to amplify the cccDNA specific segment without difficulty. As the virion contains uniquely rcDNA, amplification of the cccDNA target resulted from PCR recombination. CONCLUSIONS: PCR quantitation of cccDNA may be more difficult than hitherto thought. Current detection protocols need to be investigated so as to help in the management of chronic HBV infection.


Asunto(s)
ADN Circular/análisis , ADN Viral/análisis , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/aislamiento & purificación , Hepatitis B/virología , Reacción en Cadena en Tiempo Real de la Polimerasa , ADN Circular/sangre , ADN Viral/sangre , ADN Viral/genética , Virus de la Hepatitis B/fisiología , Hepatitis B Crónica/virología , Humanos , Virión/genética , Replicación Viral
15.
J Mol Biol ; 428(17): 3514-28, 2016 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-27289067

RESUMEN

The APOBEC3 locus consists of seven genes (A3A-A3C, A3DE, A3F-A3H) that encode DNA cytidine deaminases. These enzymes deaminate single-stranded DNA, the result being DNA peppered with CG →TA mutations preferentially in the context of 5'TpC with the exception of APOBEC3G (A3G), which prefers 5'CpC dinucleotides. Hepatitis B virus (HBV) DNA is vulnerable to genetic editing by APOBEC3 cytidine deaminases, A3G being a major restriction factor. APOBEC3DE (A3DE) stands out in that it is catalytically inactive due to a fixed Tyr320Cys substitution in the C-terminal domain. As A3DE is closely related to A3F and A3G, which can form homo- and heterodimers and multimers, the impact of A3DE on HBV replication via modulation of other APOBEC3 restriction factors was investigated. A3DE binds to itself, A3F, and A3G and antagonizes A3F and, to a lesser extent, A3G restriction of HBV replication. A3DE suppresses A3F and A3G from HBV particles, leading to enhanced HBV replication. Ironically, while being part of a cluster of innate restriction factors, the A3DE phenotype is proviral. As the gorilla genome encodes the same Tyr320Cys substitution, this proviral phenotype seems to have been selected for.


Asunto(s)
Desaminasa APOBEC-3G/antagonistas & inhibidores , Citidina Desaminasa/metabolismo , Citosina Desaminasa/antagonistas & inhibidores , Virus de la Hepatitis B/inmunología , Virus de la Hepatitis B/fisiología , Replicación Viral , Desaminasa APOBEC-3G/metabolismo , Animales , Línea Celular , Citosina Desaminasa/metabolismo , Gorilla gorilla , Humanos , Unión Proteica
16.
Nucleic Acids Res ; 43(19): 9340-9, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26384561

RESUMEN

The human APOBEC3A and APOBEC3B genes (A3A and A3B) encode DNA mutator enzymes that deaminate cytidine and 5-methylcytidine residues in single-stranded DNA (ssDNA). They are important sources of mutations in many cancer genomes which show a preponderance of CG->TA transitions. Although both enzymes can hypermutate chromosomal DNA in an experimental setting, only A3A can induce double strand DNA breaks, even though the catalytic domains of A3B and A3A differ by only 9% at the protein level. Accordingly we sought the molecular basis underlying A3B attenuation through the generation of A3A-A3B chimeras and mutants. It transpires that the N-terminal domain facilitates A3B activity while a handful of substitutions in the catalytic C-terminal domain impacting ssDNA binding serve to attenuate A3B compared to A3A. Interestingly, functional attenuation is also observed for the rhesus monkey rhA3B enzyme compared to rhA3A indicating that this genotoxic dichotomy has been selected for and maintained for some 38 million years. Expression of all human ssDNA cytidine deaminase genes is absent in mature sperm indicating they contribute to somatic mutation and cancer but not human diversity.


Asunto(s)
Citidina Desaminasa/genética , Roturas del ADN de Doble Cadena , Animales , Línea Celular , Citidina Desaminasa/química , Citidina Desaminasa/metabolismo , Células HeLa , Humanos , Macaca mulatta , Antígenos de Histocompatibilidad Menor , Mutación , Fenotipo , Estructura Terciaria de Proteína , Proteínas/química , Proteínas/genética , Codorniz , Edición de ARN
17.
Retrovirology ; 11: 93, 2014 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-25389016

RESUMEN

BACKGROUND: The role of innate immunity in general and of type I interferon (IFN-I) in particular in HTLV-1 pathogenesis is still a matter of debate. ADAR1-p150 is an Interferon Stimulated Gene (ISG) induced by IFN-I that can edit viral RNAs. We therefore investigated whether it could play the role of an anti-HTLV factor. RESULTS: We demonstrate here that ADAR1 is also expressed in the absence of IFN stimulation in activated primary T-lymphocytes that are the natural target of this virus and in HTLV-1 or HTLV-2 chronically infected T-cells. ADAR1 expression is also increased in primary lymphocytes obtained from HTLV-1 infected individuals. We show that ADAR1 enhances HTLV-1 and HTLV-2 infection in T-lymphocytes and that this proviral effect is independent from its editing activity. ADAR1 expression suppresses IFN-α inhibitory effect on HTLV-1 and HTLV-2 and acts through the repression of PKR phosphorylation. DISCUSSION: This study demonstrates that two interferon stimulated genes, i.e. PKR and ADAR1 have opposite effects on HTLV replication in vivo. The balanced expression of those proteins could determine the fate of the viral cycle in the course of infection.


Asunto(s)
Adenosina Desaminasa/metabolismo , Interacciones Huésped-Patógeno , Virus Linfotrópico T Tipo 1 Humano/fisiología , Virus Linfotrópico T Tipo 2 Humano/fisiología , Proteínas de Unión al ARN/metabolismo , Replicación Viral , eIF-2 Quinasa/antagonistas & inhibidores , Células Cultivadas , Humanos , Inhibición Psicológica , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Linfocitos T/inmunología , Linfocitos T/virología
18.
Nat Commun ; 5: 5129, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25298230

RESUMEN

Human APOBEC3A (A3A) cytidine deaminase is a host enzyme that can introduce mutations into chromosomal DNA. As APOBEC3B (A3B) encodes a C-terminal catalytic domain ~91% identical to A3A, we examined its genotoxic potential as well as that of a highly prevalent chimaeric A3A-A3B deletion allele (ΔA3B), which is linked to a higher odds ratio of developing breast, ovarian and liver cancer. Interestingly, breast cancer genomes from ΔA3B(-/-) patients show a higher overall mutation burden. Here it is shown that germline A3B can hypermutate nuclear DNA, albeit less efficiently than A3A. Chimaeric A3A mRNA resulting from ΔA3B was more stable, resulting in higher intracellular A3A levels and greater DNA damage. The cancer burden implied by the higher A3A levels could be considerable given the high penetration of the ΔA3B allele in South East Asia.


Asunto(s)
Citidina Desaminasa/genética , Daño del ADN/genética , Neoplasias/genética , Proteínas/genética , Regiones no Traducidas 3' , Alelos , Línea Celular Tumoral , Predisposición Genética a la Enfermedad , Células HEK293 , Células HeLa , Humanos , Antígenos de Histocompatibilidad Menor , Mutación
19.
Mol Biol Evol ; 31(2): 330-40, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24162735

RESUMEN

The human APOBEC3 gene cluster locus encodes polynucleotide cytidine deaminases. Although many act as viral restriction factors through mutation of single-stranded DNA, recent reports have shown that human APOBEC3A was capable of efficiently hypermutating nuclear DNA and inducing DNA breaks in genomic DNA. In addition, the enzyme was unique in efficiently deaminating 5-methylcytidine in single-stranded DNA. To appreciate the evolutionary relevance of these activities, we analyzed A3A-related enzymes from the rhesus and tamarin monkey, horse, sheep, dog, and panda. All proved to be orthologous to the human enzyme in all these activities revealing strong conservation more than 148 My. Hence, their singular role in DNA catabolism is a well-established mechanism probably outweighing any deleterious or pathological roles such as genomic instability and cancer formation.


Asunto(s)
Citidina Desaminasa/metabolismo , Citidina/análogos & derivados , ADN/genética , Mamíferos/metabolismo , Secuencia de Aminoácidos , Animales , Citidina/metabolismo , Citidina Desaminasa/química , Citidina Desaminasa/genética , Roturas del ADN de Doble Cadena , Desaminación , Perros , Inestabilidad Genómica , Células HEK293 , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Mamíferos/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Filogenia , Estructura Secundaria de Proteína , Proteínas/química , Proteínas/genética , Proteínas/metabolismo , Alineación de Secuencia
20.
PLoS One ; 8(8): e73641, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977391

RESUMEN

Human APOBEC3 enzymes deaminate single stranded DNA. At least five can deaminate mitochondrial DNA in the cytoplasm, while three can deaminate viral DNA in the nucleus. However, only one, APOBEC3A, can hypermutate genomic DNA. We analysed the distribution and function of the two APOBEC3A isoforms p1 and p2 in transfected cell lines. Both can translocate to the nucleus and hypermutate CMYC DNA and induce DNA double strand breaks as visualized by the detection of ©H2AX or Chk2. APOBEC3A induced G1 phase cell cycle arrest and triggered several members of the intrinsic apoptosis pathway. Activation of purified human CD4+ T lymphocytes with PHA, IL2 and interferon α resulted in C->T hypermutation of genomic DNA and double stranded breaks suggesting a role for APOBEC3A in pro-inflammatory conditions. As chronic inflammation underlies many diseases including numerous cancers, it is possible that APOBEC3A induction may generate many of the lesions typical of a cancer genome.


Asunto(s)
Núcleo Celular/metabolismo , Citidina Desaminasa/metabolismo , Roturas del ADN de Doble Cadena , Proteínas/metabolismo , Estrés Fisiológico , Secuencia de Bases , Linfocitos T CD4-Positivos/inmunología , Puntos de Control del Ciclo Celular , Muerte Celular , Citidina Desaminasa/genética , Células HeLa , Histonas/metabolismo , Humanos , Activación de Linfocitos , Datos de Secuencia Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , Proteínas/genética , Edición de ARN/genética , Transducción de Señal/genética , Uracil-ADN Glicosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...