Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 187
Filtrar
1.
PLoS Pathog ; 17(11): e1010026, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34762705

RESUMEN

Salivarian trypanosomes are extracellular protozoan parasites causing infections in a wide range of mammalian hosts, with Trypanosoma evansi having the widest geographic distribution, reaching territories far outside Africa and occasionally even Europe. Besides causing the animal diseases, T. evansi can cause atypical Human Trypanosomosis. The success of this parasite is attributed to its capacity to evade and disable the mammalian defense response. To unravel the latter, we applied here for the first time a scRNA-seq analysis on splenocytes from trypanosome infected mice, at two time points during infection, i.e. just after control of the first parasitemia peak (day 14) and a late chronic time point during infection (day 42). This analysis was combined with flow cytometry and ELISA, revealing that T. evansi induces prompt activation of splenic IgM+CD1d+ Marginal Zone and IgMIntIgD+ Follicular B cells, coinciding with an increase in plasma IgG2c Ab levels. Despite the absence of follicles, a rapid accumulation of Aicda+ GC-like B cells followed first parasitemia peak clearance, accompanied by the occurrence of Xbp1+ expressing CD138+ plasma B cells and Tbx21+ atypical CD11c+ memory B cells. Ablation of immature CD93+ bone marrow and Vpreb3+Ly6d+Ighm+ expressing transitional spleen B cells prevented mature peripheral B cell replenishment. Interestingly, AID-/- mice that lack the capacity to mount anti-parasite IgG responses, exhibited a superior defense level against T. evansi infections. Here, elevated natural IgMs were able to exert in vivo and in vitro trypanocidal activity. Hence, we conclude that in immune competent mice, trypanosomosis associated B cell activation and switched IgG production is rapidly induced by T. evansi, facilitating an escape from the detrimental natural IgM killing activity, and resulting in increased host susceptibility. This unique role of IgM and its anti-trypanosome activity are discussed in the context of the dilemma this causes for the future development of anti-trypanosome vaccines.


Asunto(s)
Linfocitos B/inmunología , Citidina Desaminasa/fisiología , Cambio de Clase de Inmunoglobulina , Mutación , Análisis de la Célula Individual/métodos , Trypanosoma/genética , Tripanosomiasis/parasitología , Animales , Anticuerpos Antiprotozoarios/inmunología , Femenino , Isotipos de Inmunoglobulinas/inmunología , Activación de Linfocitos , Células B de Memoria/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transcriptoma , Trypanosoma/inmunología , Tripanosomiasis/genética , Tripanosomiasis/inmunología
2.
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
3.
Nucleic Acids Res ; 49(9): 5057-5073, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33950194

RESUMEN

Activation-induced deaminase (AID) initiates antibody diversification in germinal center B cells by deaminating cytosines, leading to somatic hypermutation and class-switch recombination. Loss-of-function mutations in AID lead to hyper-IgM syndrome type 2 (HIGM2), a rare human primary antibody deficiency. AID-mediated deamination has been proposed as leading to active demethylation of 5-methycytosines in the DNA, although evidence both supports and casts doubt on such a role. In this study, using whole-genome bisulfite sequencing of HIGM2 B cells, we investigated direct AID involvement in active DNA demethylation. HIGM2 naïve and memory B cells both display widespread DNA methylation alterations, of which ∼25% are attributable to active DNA demethylation. For genes that undergo active demethylation that is impaired in HIGM2 individuals, our analysis indicates that AID is not directly involved. We demonstrate that the widespread alterations in the DNA methylation and expression profiles of HIGM2 naïve B cells result from premature overstimulation of the B-cell receptor prior to the germinal center reaction. Our data support a role for AID in B cell central tolerance in preventing the expansion of autoreactive cell clones, affecting the correct establishment of DNA methylation patterns.


Asunto(s)
Linfocitos B/inmunología , Citidina Desaminasa/fisiología , Metilación de ADN , Síndrome de Inmunodeficiencia con Hiper-IgM/genética , Síndrome de Inmunodeficiencia con Hiper-IgM/inmunología , Autoinmunidad , Linfocitos B/metabolismo , Citidina Desaminasa/deficiencia , Citidina Desaminasa/genética , Centro Germinal/inmunología , Humanos , Síndrome de Inmunodeficiencia con Hiper-IgM/metabolismo , Tolerancia Inmunológica , Memoria Inmunológica , Receptores de Antígenos de Linfocitos B/genética , Transcriptoma , Secuenciación Completa del Genoma
4.
Front Immunol ; 11: 2129, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33072075

RESUMEN

Macrophages are key targets of human immunodeficiency virus type 1 (HIV-1) infection and main producers of the proinflammatory chemokine CC chemokine ligand 2 (CCL2), whose expression is induced by HIV-1 both in vitro and in vivo. We previously found that CCL2 neutralization in monocyte-derived macrophages (MDMs) strongly inhibited HIV-1 replication affecting post-entry steps of the viral life cycle. Here, we used RNA-sequencing to deeply characterize the cellular factors and pathways modulated by CCL2 blocking in MDMs and involved in HIV-1 replication restriction. We report that exposure to CCL2 neutralizing antibody profoundly affected the MDM transcriptome. Functional annotation clustering of up-regulated genes identified two clusters enriched for antiviral defense and immune response pathways, comprising several interferon-stimulated, and restriction factor coding genes. Transcripts in the clusters were enriched for RELA and NFKB1 targets, suggesting the activation of the canonical nuclear factor κB pathway as part of a regulatory network involving miR-155 up-regulation. Furthermore, while HIV-1 infection caused small changes to the MDM transcriptome, with no evidence of host defense gene expression and type I interferon signature, CCL2 blocking enabled the activation of a strong host innate response in infected macrophage cultures, and potently inhibited viral genes expression. Notably, an inverse correlation was found between levels of viral transcripts and of the restriction factors APOBEC3A (apolipoprotein B mRNA editing enzyme catalytic polypeptide-like 3 A), ISG15, and MX1. These findings highlight an association between activation of innate immune pathways and HIV-1 restriction upon CCL2 blocking and identify this chemokine as an endogenous factor contributing to the defective macrophage response to HIV-1. Therapeutic targeting of CCL2 may thus strengthen host innate immunity and restrict HIV-1 replication.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Quimiocina CCL2/farmacología , Perfilación de la Expresión Génica , Regulación Viral de la Expresión Génica/efectos de los fármacos , VIH-1/genética , Inmunidad Innata , Macrófagos/metabolismo , Anticuerpos Neutralizantes/inmunología , Especificidad de Anticuerpos , Células Cultivadas , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/inmunología , Citidina Desaminasa/fisiología , Conjuntos de Datos como Asunto , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/virología , MicroARNs/biosíntesis , MicroARNs/genética , Anotación de Secuencia Molecular , FN-kappa B/metabolismo , Proteínas/fisiología , ARN Viral/biosíntesis , ARN Viral/genética , RNA-Seq , Reacción en Cadena en Tiempo Real de la Polimerasa , Latencia del Virus , Replicación Viral
5.
Sci Rep ; 10(1): 14164, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32843697

RESUMEN

Activation-induced cytidine deaminase (AID) is one kind of the mutant enzymes, which target regulating the immunoglobulin (Ig) gene in Burkitt's lymphoma to initiate class switch recombination (CSR), resulting in c-Myc chromosomal translocation. However, it is not clear that whether AID induces c-Myc/IgH translocation in double-hit lymphoma (DHL) with c-Myc gene translocation. In this study, the AID in DHL tissues and classical diffuse large b-cell lymphoma (DLBCL) tissues were compared. The results suggested that AID is of important value in predicting DHL, stronger CSR of AID was observed in DHL patients, which exhibited AID overexpression and c-Myc gene translocation of DHL after CSR induction. It is concluded that AID directly induces CSR in DHL and may result in c-Myc gene translocation. Targeting AID may be a good treatment regimen for DHL.


Asunto(s)
Citidina Desaminasa/biosíntesis , Cambio de Clase de Inmunoglobulina/genética , Linfoma de Células B Grandes Difuso/enzimología , Terapia Molecular Dirigida , Proteínas de Neoplasias/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Citidina Desaminasa/genética , Citidina Desaminasa/fisiología , Inducción Enzimática/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes bcl-2 , Genes myc , Humanos , Isotipos de Inmunoglobulinas/biosíntesis , Isotipos de Inmunoglobulinas/sangre , Isotipos de Inmunoglobulinas/genética , Estimación de Kaplan-Meier , Antígeno Ki-67/genética , Lipopolisacáridos/farmacología , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/mortalidad , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Pronóstico , Proteínas Proto-Oncogénicas c-bcl-6/genética , Translocación Genética , Regulación hacia Arriba/efectos de los fármacos
6.
J Cancer Res Clin Oncol ; 146(11): 2721-2730, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32772231

RESUMEN

Activation Induced cytidine Deaminase (AID) is an essential enzyme of the adaptive immune system. Its canonical activity is restricted to B lymphocytes, playing an essential role in the diversification of antibodies by enhancing specificity and changing affinity. This is possible through its DNA deaminase function, leading to mutations in DNA. In the last decade, AID has been assigned an additional function: that of a powerful DNA demethylator. Adverse cellular conditions such as chronic inflammation can lead to its deregulation and overexpression. It is an important driver of B-cell lymphoma due to its natural ability to modify DNA through deamination, leading to mutations and epigenetic changes. However, the deregulation of AID is not restricted to lymphoid cells. Recent findings have provided new insights into the role that this protein plays in the development of non-lymphoid cancers, with some research shedding light on novel AID-driven mechanisms of cellular transformation. In this review, we provide an updated narrative of the normal physiological functions of AID. Additionally, we review and discuss the recent research studies that have implicated AID in carcinogenesis in varying tissue types including lymphoid and non-lymphoid cancers. We review the mechanisms, whereby AID promotes carcinogenesis and highlight important areas of future research.


Asunto(s)
Inmunidad Adaptativa/fisiología , Citidina Desaminasa/fisiología , Neoplasias/enzimología , Animales , Transformación Celular Neoplásica/inmunología , Humanos , Neoplasias/inmunología
7.
Int J Hematol ; 112(3): 395-408, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32533515

RESUMEN

The apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like (APOBEC) protein family members have cytidine deaminase activity and can induce cytosine to uracil transition in nucleic acid. The main function of APOBEC3 (A3) proteins is to trigger an innate immune response to viral infections. Recent reports have shown that several APOBEC family proteins such as A3B can induce somatic mutations into genomic DNA and thus promote cancer development. However, the role of A3D on somatic mutations is unclear. Here, we identified the alternative splicing of A3D, and investigated each splice variant's subcellular localization and role in DNA mutagenesis. We identified four A3D variants, which all have one or two cytidine deaminase domains. The full-length form of A3D (variant 1) and truncated forms of A3D (variant 2, 6, 7) showed the ability to induce C/G to T/A transitions in foreign DNA and genomic DNA and retained antiretroviral activity. Furthermore, we demonstrated that A3D and A3B could induce deletions that are possibly repaired by microhomology-mediated end joining (MMEJ). Taken together, our experiments illustrated that alternative splicing generates functional diversity of A3D, and some variants can act as DNA mutators in genomic DNA.


Asunto(s)
Empalme Alternativo/genética , Citidina Desaminasa/genética , Citidina Desaminasa/fisiología , ADN/genética , Mutación/genética , Antirretrovirales , Línea Celular Tumoral , Células Cultivadas , Citidina Desaminasa/química , Reparación del ADN por Unión de Extremidades/genética , Variación Genética , Humanos , Dominios Proteicos
8.
Front Immunol ; 11: 788, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32425948

RESUMEN

Somatic hypermutation (SHM) of the immunoglobulin variable (IgV) loci is a key process in antibody affinity maturation. The enzyme activation-induced deaminase (AID), initiates SHM by creating C → U mismatches on single-stranded DNA (ssDNA). AID has preferential hotspot motif targets in the context of WRC/GYW (W = A/T, R = A/G, Y = C/T) and particularly at WGCW overlapping hotspots where hotspots appear opposite each other on both strands. Subsequent recruitment of the low-fidelity DNA repair enzyme, Polymerase eta (Polη), during mismatch repair, creates additional mutations at WA/TW sites. Although there are more than 50 functional immunoglobulin heavy chain variable (IGHV) segments in humans, the fundamental differences between these genes and their ability to respond to all possible foreign antigens is still poorly understood. To better understand this, we generated profiles of WGCW hotspots in each of the human IGHV genes and found the expected high frequency in complementarity determining regions (CDRs) that encode the antigen binding sites but also an unexpectedly high frequency of WGCW in certain framework (FW) sub-regions. Principal Components Analysis (PCA) of these overlapping AID hotspot profiles revealed that one major difference between IGHV families is the presence or absence of WGCW in a sub-region of FW3 sometimes referred to as "CDR4." Further differences between members of each family (e.g., IGHV1) are primarily determined by their WGCW densities in CDR1. We previously suggested that the co-localization of AID overlapping and Polη hotspots was associated with high mutability of certain IGHV sub-regions, such as the CDRs. To evaluate the importance of this feature, we extended the WGCW profiles, combining them with local densities of Polη (WA) hotspots, thus describing the co-localization of both types of hotspots across all IGHV genes. We also verified that co-localization is associated with higher mutability. PCA of the co-localization profiles showed CDR1 and CDR2 as being the main contributors to variance among IGHV genes, consistent with the importance of these sub-regions in antigen binding. Our results suggest that AID overlapping (WGCW) hotspots alone or in conjunction with Polη (WA/TW) hotspots are key features of evolutionary variation between IGHV genes.


Asunto(s)
Citidina Desaminasa/fisiología , ADN Polimerasa Dirigida por ADN/fisiología , Evolución Molecular , Cadenas Pesadas de Inmunoglobulina/genética , Regiones Determinantes de Complementariedad , Humanos , Mutación
9.
Virologie (Montrouge) ; 24(6): 381-418, 2020 12 01.
Artículo en Francés | MEDLINE | ID: mdl-33441290

RESUMEN

The innate immune response is nonspecific and constitutes the first line of defense against infections by pathogens, mainly by enabling their elimination by phagocytosis or apoptosis. In immune cells, this response is characterized, amongst others, by the synthesis of restriction factors, a class of proteins whose role is to inhibit viral replication. Among them, the proteins of the APOBEC3 (Apolipoprotein B mRNA-editing Enzyme Catalytic polypeptide-like 3 or A3) family are major antiviral factors that target a wide range of viruses. One of their targets is the Human Immunodeficiency Virus Type 1 (HIV-1): the deaminase activity of some A3 proteins converts a fraction of cytidines of the viral genome into uridines, impairing its expression. Nevertheless, HIV-1 counteracts A3 proteins thanks to its Vif protein, which inhibits them by hijacking several cellular mechanisms. Besides, APOBEC3 proteins help maintaining the genome integrity by inhibiting retroelements but they also contribute to carcinogenesis, as it is the case for A3A and A3B, two major factors in this process. The large range of A3 activities, combined with recent studies showing their implication in the regulation of emerging viruses (Zika, SARS-CoV-2), allow A3 and their viral partners to be considered as therapeutic areas.


Asunto(s)
Desaminasas APOBEC/fisiología , COVID-19/inmunología , Inmunidad Innata , Adulto , Secuencias de Aminoácidos , Animales , Proteínas de Ciclo Celular/metabolismo , Citidina Desaminasa/fisiología , Reparación del ADN , ADN Viral/metabolismo , Desaminación , Humanos , Mamíferos/metabolismo , MicroARNs/genética , Modelos Moleculares , Terapia Molecular Dirigida , Mutagénesis , Neoplasias/enzimología , Neoplasias/etiología , Neoplasias/genética , Pronóstico , Conformación Proteica , Edición de ARN , Relación Estructura-Actividad , Transcripción Genética , Proteínas Virales/metabolismo , Virosis/tratamiento farmacológico , Virosis/enzimología , Virosis/inmunología , Replicación Viral
10.
Exp Dermatol ; 28(11): 1341-1347, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31400166

RESUMEN

Apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like (APOBEC) family consists of deaminases. Some isozymes of APOBEC3 are induced upon human papillomavirus infection or development of psoriasis skin lesions. However, the involvement of APOBEC3 in keratinocyte differentiation has not been addressed. We herein sought to evaluate the roles of APOBECs in mouse primary keratinocyte differentiation. We found that expression levels of APOBEC1 and APOBEC3 were increased during calcium-induced keratinocyte differentiation. Unexpectedly, however, the expression levels of keratinocyte differentiation markers keratin 1/10, involucrin, loricrin and filaggrin were higher in keratinocytes treated with APOBEC3 siRNAs than in those treated with control RNAs. In addition, the treatment of keratinocytes with APOBEC3 siRNAs increased the gene expression levels of Notch3, a master regulator of keratinocyte differentiation. Moreover, calcium-induced increase in Notch3 expression and keratinocyte differentiation were impaired by transfection with an APOBEC3 expression plasmid. Furthermore, co-treatment with Notch3 siRNAs reduced the APOBEC3 siRNA-mediated upregulation of Notch3 expression and in part attenuated the increased expression levels of keratinocyte differentiation markers. These results suggest that APOBEC3 is induced upon keratinocyte differentiation and negatively regulates the keratinocyte differentiation in part by its inhibitory role for Notch3 expression.


Asunto(s)
Diferenciación Celular , Citidina Desaminasa/fisiología , Queratinocitos/fisiología , Receptor Notch3/metabolismo , Animales , Animales Recién Nacidos , Proteínas Filagrina , Ratones , Cultivo Primario de Células
11.
J Am Soc Nephrol ; 30(7): 1238-1249, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31227634

RESUMEN

BACKGROUND: IgA nephropathy (IgAN) often follows infections and features IgA mesangial deposition. Polymeric IgA deposits in the mesangium seem to have varied pathogenic potential, but understanding their pathogenicity remains a challenge. Most mesangial IgA1 in human IgAN has a hypogalactosylated hinge region, but it is unclear whether this is required for IgA deposition. Another important question is the role of adaptive IgA responses and high-affinity mature IgA antibodies and whether low-affinity IgA produced by innate-like B cells might also yield mesangial deposits. METHODS: To explore the effects of specific qualitative variations in IgA and whether altered affinity maturation can influence IgA mesangial deposition and activate complement, we used several transgenic human IgA1-producing models with IgA deposition, including one lacking the DNA-editing enzyme activation-induced cytidine deaminase (AID), which is required in affinity maturation. Also, to explore the potential role of the IgA receptor CD89 in glomerular inflammation, we used a model that expresses CD89 in a pattern observed in humans. RESULTS: We found that human IgA induced glomerular damage independent of CD89. When comparing mice able to produce high-affinity IgA antibodies with mice lacking AID-enabled Ig affinity maturation, we found that IgA deposition and complement activation significantly increased and led to IgAN pathogenesis, although without significant proteinuria or hematuria. We also observed that hinge hypoglycosylation was not mandatory for IgA deposition. CONCLUSIONS: In a mouse model of IgAN, compared with high-affinity IgA, low-affinity innate-like IgA, formed in the absence of normal antigen-driven maturation, was more readily involved in IgA glomerular deposition with pathogenic effects.


Asunto(s)
Afinidad de Anticuerpos , Mesangio Glomerular/metabolismo , Glomerulonefritis por IGA/etiología , Inmunoglobulina A/metabolismo , Animales , Antígenos CD/fisiología , Activación de Complemento , Citidina Desaminasa/fisiología , Mesangio Glomerular/patología , Glomerulonefritis por IGA/inmunología , Glicosilación , Humanos , Inmunoglobulina A/toxicidad , Ratones , Receptores Fc/fisiología
12.
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
13.
Adv Exp Med Biol ; 1044: 49-64, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29956291

RESUMEN

The sources of genome instability can be attributed to many extra- and exo- cellular factors accompanying various biological processes. In leukemia and lymphomas, the collateral effect of programmed DNA alterations during immune diversification is the major source of genome instability. Cytidine deamination from cytidine (C) to uridine (U) at immunoglobulin (Ig) gene loci is required for initiation of antibody diversification, while the same process also contributes to recurrent translocation or mutations outside of Ig loci in lymphocyte-origin tumors. Furthermore, genome sequencing of cancer cells from many tissue origins revealed a significant enrichment of cytidine deaminase mutagenesis signature in human cancers. Thus, cytidine deamination, which can intensively happen in an enzyme-dependent fashion at specific genomic regions, is a widespread genome instability source across many tumor types. AID/APOBEC superfamily proteins are the main single-stranded DNA deaminases in eukaryotes, which play vital roles in adaptive and innate immunity. Their deamination products can be channeled into mutations, insertions and deletions (indels), clusters of mutations called kaetagis, or chromosomal rearrangements/translocations. Here, we review the generation of genome instability from AID/APOBEC-dependent cytidine deamination with emphasis on the most studied enzyme, AID.


Asunto(s)
Citidina Desaminasa/fisiología , Citidina/metabolismo , Inestabilidad Genómica , Desaminación , Humanos , Mutación
14.
Adv Exp Med Biol ; 1044: 65-87, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29956292

RESUMEN

In response to DNA double strand breaks (DSB), mammalian cells activate the DNA Damage Response (DDR), a network of factors that coordinate their detection, signaling and repair. Central to this network is the ATM kinase and its substrates at chromatin surrounding DSBs H2AX, MDC1 and 53BP1. In humans, germline inactivation of ATM causes Ataxia Telangiectasia (A-T), an autosomal recessive syndrome of increased proneness to hematological malignancies driven by clonal chromosomal translocations. Studies of cancers arising in A-T patients and in genetically engineered mouse models (GEMM) deficient for ATM and its substrates have revealed complex, multilayered roles for ATM in translocation suppression and identified functional redundancies between ATM and its substrates in this context. "Programmed" DSBs at antigen receptor loci in developing lymphocytes employ ubiquitous DDR factors for signaling and repair and have been particularly useful for mechanistic studies because they are region-specific and can be monitored in vitro and in vivo. In this context, murine thymocytes deficient for ATM recapitulate the molecular events that lead to transformation in T cells from A-T patients and provide a widely used model to study the mechanisms that suppress RAG recombinase-dependent translocations. Similarly, analyses of the fate of Activation induced Cytidine Deaminase (AID)-dependent DSBs during mature B cell Class Switch Recombination (CSR) have defined the genetic requirements for end-joining and translocation suppression in this setting. Moreover, a unique role for 53BP1 in the promotion of synapsis of distant DSBs has emerged from these studies.


Asunto(s)
Roturas del ADN de Doble Cadena , Translocación Genética , Animales , Ataxia Telangiectasia/genética , Citidina Desaminasa/fisiología , Reparación del ADN , Ingeniería Genética , Humanos , Cambio de Clase de Inmunoglobulina/genética , Ratones , Recombinación Genética
15.
Mol Biotechnol ; 60(6): 435-453, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29704159

RESUMEN

The disparate diversity in immunoglobulin (Ig) repertoire has been a subject of fascination since the emergence of prototypic adaptive immune system in vertebrates. The carboxy terminus region of activation-induced cytidine deaminase (AID) has been well established in tetrapod lineage and is crucial for its function in class switch recombination (CSR) event of Ig diversification. The absence of CSR in the paraphyletic group of fish is probably due to changes in catalytic domain of AID and lack of cis-elements in IgH locus. Therefore, understanding the arrangement of Ig genes in IgH locus and functional facets of fish AID opens up new realms of unravelling the alternative mechanisms of isotype switching and antibody diversity. Further, the teleost AID has been recently reported to have potential of catalyzing CSR in mammalian B cells by complementing AID deficiency in them. In that context, the present review focuses on the recent advances regarding the generation of diversity in Ig repertoire in the absence of AID-regulated class switching in teleosts and the possible role of T cell-independent pathway involving B cell activating factor and a proliferation-inducing ligand in activation of CSR machinery.


Asunto(s)
Citidina Desaminasa/fisiología , Peces/inmunología , Cambio de Clase de Inmunoglobulina , Isotipos de Inmunoglobulinas/genética , Animales , Diversidad de Anticuerpos , Factor Activador de Células B/inmunología , Evolución Molecular , Peces/genética , Genes de las Cadenas Pesadas de las Inmunoglobulinas , Humanos , Isotipos de Inmunoglobulinas/metabolismo , Ratones , Receptores de Antígenos/inmunología , Receptores de Reconocimiento de Patrones/inmunología
16.
Biosci Trends ; 11(3): 243-253, 2017 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-28484188

RESUMEN

Cytidine to uridine (C-to-U) editing is one type of substitutional RNA editing. It occurs in both mammals and plants. The molecular mechanism of C-to-U editing involves the hydrolytic deamination of a cytosine to a uracil base. C-to-U editing is mediated by RNA-specific cytidine deaminases and several complementation factors, which have not been completely identified. Here, we review recent findings related to the regulation and enzymatic basis of C-to-U RNA editing. More importantly, when C-to-U editing occurs in coding regions, it has the power to reprogram genetic information on the RNA level, therefore it has great potential for applications in transcript repair (diseases related to thymidine to cytidine (T>C) or adenosine to guanosine (A>G) point mutations). If it is possible to manipulate or mimic C-to-U editing, T>C or A>G genetic mutation-related diseases could be treated. Enzymatic and non-enzymatic site-directed RNA editing are two different approaches for mimicking C-to-U editing. For enzymatic site-directed RNA editing, C-to-U editing has not yet been successfully performed, and in theory, adenosine to inosine (A-to-I) editing involves the same strategy as C-to-U editing. Therefore, in this review, for applications in transcript repair, we will provide a detailed overview of enzymatic site-directed RNA editing, with a focus on A-to-I editing and non-enzymatic site-directed C-to-U editing.


Asunto(s)
Citidina/química , Mutación Puntual , Edición de ARN/fisiología , Uridina/química , Animales , Citidina Desaminasa/fisiología , Ingeniería Genética/métodos , Terapia Genética/métodos , Mamíferos/genética , Modelos Genéticos , Plantas/genética
17.
Blood ; 129(13): 1779-1790, 2017 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-28077417

RESUMEN

Recent studies have reported that activation-induced cytidine deaminase (AID) and ten-eleven-translocation (TET) family members regulate active DNA demethylation. Genetic alterations of TET2 occur in myeloid malignancies, and hematopoietic-specific loss of Tet2 induces aberrant hematopoietic stem cell (HSC) self-renewal/differentiation, implicating TET2 as a master regulator of normal and malignant hematopoiesis. Despite the functional link between AID and TET in epigenetic gene regulation, the role of AID loss in hematopoiesis and myeloid transformation remains to be investigated. Here, we show that Aid loss in mice leads to expansion of myeloid cells and reduced erythroid progenitors resulting in anemia, with dysregulated expression of Cebpa and Gata1, myeloid/erythroid lineage-specific transcription factors. Consistent with data in the murine context, silencing of AID in human bone marrow cells skews differentiation toward myelomonocytic lineage. However, in contrast to Tet2 loss, Aid loss does not contribute to enhanced HSC self-renewal or cooperate with Flt3-ITD to induce myeloid transformation. Genome-wide transcription and differential methylation analysis uncover the critical role of Aid as a key epigenetic regulator. These results indicate that AID and TET2 share common effects on myeloid and erythroid lineage differentiation, however, their role is nonredundant in regulating HSC self-renewal and in myeloid transformation.


Asunto(s)
Diferenciación Celular , Citidina Desaminasa/fisiología , Metilación de ADN , Células Madre Hematopoyéticas/metabolismo , Animales , Linaje de la Célula , Autorrenovación de las Células , Transformación Celular Neoplásica , Citidina Desaminasa/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Dioxigenasas , Células Eritroides/citología , Silenciador del Gen , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Células Mieloides/citología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología
18.
Nat Struct Mol Biol ; 24(2): 131-139, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27991903

RESUMEN

APOBEC-catalyzed cytosine-to-uracil deamination of single-stranded DNA (ssDNA) has beneficial functions in immunity and detrimental effects in cancer. APOBEC enzymes have intrinsic dinucleotide specificities that impart hallmark mutation signatures. Although numerous structures have been solved, mechanisms for global ssDNA recognition and local target-sequence selection remain unclear. Here we report crystal structures of human APOBEC3A and a chimera of human APOBEC3B and APOBEC3A bound to ssDNA at 3.1-Å and 1.7-Å resolution, respectively. These structures reveal a U-shaped DNA conformation, with the specificity-conferring -1 thymine flipped out and the target cytosine inserted deep into the zinc-coordinating active site pocket. The -1 thymine base fits into a groove between flexible loops and makes direct hydrogen bonds with the protein, accounting for the strong 5'-TC preference. These findings explain both conserved and unique properties among APOBEC family members, and they provide a basis for the rational design of inhibitors to impede the evolvability of viruses and tumors.


Asunto(s)
Citidina Desaminasa/química , Antígenos de Histocompatibilidad Menor/química , Proteínas/química , Aminación , Secuencia de Bases , Sitios de Unión , Dominio Catalítico , Secuencia de Consenso , Cristalografía por Rayos X , Citidina Desaminasa/fisiología , Citosina , ADN de Cadena Simple/química , Humanos , Enlace de Hidrógeno , Cinética , Antígenos de Histocompatibilidad Menor/fisiología , Modelos Moleculares , Mutagénesis , Unión Proteica , Conformación Proteica en Hélice alfa , Proteínas/fisiología , Especificidad por Sustrato
19.
J Exp Med ; 214(1): 183-196, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27994069

RESUMEN

Noncoding transcripts originating upstream of the immunoglobulin constant region (I transcripts) are required to direct activation-induced deaminase to initiate class switching in B cells. Differential regulation of Iε and Iγ1 transcription in response to interleukin 4 (IL-4), hence class switching to IgE and IgG1, is not fully understood. In this study, we combine novel mouse reporters and single-cell RNA sequencing to reveal the heterogeneity in IL-4-induced I transcription. We identify an early population of cells expressing Iε but not Iγ1 and demonstrate that early Iε transcription leads to switching to IgE and occurs at lower activation levels than Iγ1. Our results reveal how probabilistic transcription with a lower activation threshold for Iε directs the early choice of IgE versus IgG1, a key physiological response against parasitic infestations and a mediator of allergy and asthma.


Asunto(s)
Linfocitos B/metabolismo , Cambio de Clase de Inmunoglobulina , Inmunoglobulina E/genética , Transcripción Genética , Animales , Citidina Desaminasa/fisiología , Interleucina-4/sangre , Ratones , Ratones Endogámicos BALB C , Regiones Promotoras Genéticas
20.
J Exp Med ; 214(1): 197-208, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28011866

RESUMEN

Generation of cellular heterogeneity is an essential feature of the adaptive immune system. This is best exemplified during humoral immune response when an expanding B cell clone assumes multiple cell fates, including class-switched B cells, antibody-secreting plasma cells, and memory B cells. Although each cell type is essential for immunity, their generation must be exquisitely controlled because a class-switched B cell cannot revert back to the parent isotype, and a terminally differentiated plasma cell cannot contribute to the memory pool. In this study, we show that an environmental sensor, the aryl hydrocarbon receptor (AhR) is highly induced upon B cell activation and serves a critical role in regulating activation-induced cell fate outcomes. We find that AhR negatively regulates class-switch recombination ex vivo by altering activation-induced cytidine deaminase expression. We further demonstrate that AhR suppresses class switching in vivo after influenza virus infection and immunization with model antigens. In addition, by regulating Blimp-1 expression via Bach2, AhR represses differentiation of B cells into plasmablasts ex vivo and antibody-secreting plasma cells in vivo. These experiments suggest that AhR serves as a molecular rheostat in B cells to brake the effector response, possibly to facilitate optimal recall responses. Thus, AhR might represent a novel molecular target for manipulation of B cell responses during vaccination.


Asunto(s)
Linfocitos B/fisiología , Receptores de Hidrocarburo de Aril/fisiología , Animales , Diferenciación Celular , Citidina Desaminasa/fisiología , Femenino , Cambio de Clase de Inmunoglobulina , Subtipo H1N1 del Virus de la Influenza A/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Células Plasmáticas/citología , Dibenzodioxinas Policloradas/farmacología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Linfocitos T/fisiología , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA