Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 276
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 16(7): 766-774, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25985233

RESUMEN

Childhood acute lymphoblastic leukemia (ALL) can often be traced to a pre-leukemic clone carrying a prenatal genetic lesion. Postnatally acquired mutations then drive clonal evolution toward overt leukemia. The enzymes RAG1-RAG2 and AID, which diversify immunoglobulin-encoding genes, are strictly segregated in developing cells during B lymphopoiesis and peripheral mature B cells, respectively. Here we identified small pre-BII cells as a natural subset with increased genetic vulnerability owing to concurrent activation of these enzymes. Consistent with epidemiological findings on childhood ALL etiology, susceptibility to genetic lesions during B lymphopoiesis at the transition from the large pre-BII cell stage to the small pre-BII cell stage was exacerbated by abnormal cytokine signaling and repetitive inflammatory stimuli. We demonstrated that AID and RAG1-RAG2 drove leukemic clonal evolution with repeated exposure to inflammatory stimuli, paralleling chronic infections in childhood.


Asunto(s)
Linfocitos B/inmunología , Evolución Clonal/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Células Precursoras de Linfocitos B/inmunología , Adolescente , Animales , Diversidad de Anticuerpos/genética , Diversidad de Anticuerpos/inmunología , Linfocitos B/metabolismo , Niño , Preescolar , Evolución Clonal/genética , Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , Citidina Desaminasa/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Femenino , Citometría de Flujo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Proteínas de Homeodominio/metabolismo , Humanos , Immunoblotting , Lactante , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Microscopía Fluorescente , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
2.
Nat Immunol ; 14(7): 749-55, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23708250

RESUMEN

Beyond its well-characterized functions in antibody diversification, the cytidine deaminase AID can catalyze off-target DNA damage and has been hypothesized to edit RNA and mediate DNA demethylation. To comprehensively examine the effects of AID on the transcriptome and the pattern of DNA methylation ('methylome'), we analyzed AID-deficient (Aicda(-/-)), wild-type and AID-overexpressing activated B cells by high-throughput RNA sequencing (RNA-Seq) and reduced-representation bisulfite sequencing (RRBS). These analyses confirmed the known role of AID in immunoglobulin isotype switching and also demonstrated few other effects of AID on gene expression. Additionally, we detected no evidence of AID-dependent editing of mRNA or microRNA. Finally, the RRBS data did not support the proposed role for AID in regulating DNA methylation. Thus, despite evidence of its additional activities in other systems, antibody diversification seems to be the sole physiological function of AID in activated B cells.


Asunto(s)
Linfocitos B/inmunología , Citidina Desaminasa/inmunología , Isotipos de Inmunoglobulinas/inmunología , Hipermutación Somática de Inmunoglobulina/inmunología , Transcriptoma/inmunología , Animales , Femenino , Masculino , Ratones , Ratones Noqueados , MicroARNs/química , MicroARNs/genética , ARN Mensajero/química , ARN Mensajero/genética , Análisis de Secuencia de ADN
3.
Nat Immunol ; 14(11): 1183-1189, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24097111

RESUMEN

The ability of activation-induced cytidine deaminase (AID) to efficiently mediate class-switch recombination (CSR) is dependent on its phosphorylation at Ser38; however, the trigger that induces AID phosphorylation and the mechanism by which phosphorylated AID drives CSR have not been elucidated. Here we found that phosphorylation of AID at Ser38 was induced by DNA breaks. Conversely, in the absence of AID phosphorylation, DNA breaks were not efficiently generated at switch (S) regions in the immunoglobulin heavy-chain locus (Igh), consistent with a failure of AID to interact with the endonuclease APE1. Additionally, deficiency in the DNA-damage sensor ATM impaired the phosphorylation of AID at Ser38 and the interaction of AID with APE1. Our results identify a positive feedback loop for the amplification of DNA breaks at S regions through the phosphorylation- and ATM-dependent interaction of AID with APE1.


Asunto(s)
Linfocitos B/inmunología , Citidina Desaminasa/inmunología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/inmunología , Retroalimentación Fisiológica , Cambio de Clase de Inmunoglobulina , Cadenas Pesadas de Inmunoglobulina/inmunología , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/inmunología , Linfocitos B/citología , Citidina Desaminasa/genética , Roturas del ADN de Doble Cadena , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Regulación de la Expresión Génica , Cadenas Pesadas de Inmunoglobulina/genética , Ratones , Fosforilación , Unión Proteica , Serina/inmunología , Serina/metabolismo , Transducción de Señal
4.
Immunity ; 43(6): 1075-86, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26620760

RESUMEN

Phosphatidylinositol 3' OH kinase (PI3K) signaling and FOXO transcription factors play opposing roles at several B cell developmental stages. We show here abundant nuclear FOXO1 expression in the proliferative compartment of the germinal center (GC), its dark zone (DZ), and PI3K activity, downregulating FOXO1, in the light zone (LZ), where cells are selected for further differentiation. In the LZ, however, FOXO1 was expressed in a fraction of cells destined for DZ reentry. Upon FOXO1 ablation or induction of PI3K activity, GCs lost their DZ, owing at least partly to downregulation of the chemokine receptor CXCR4. Although this prevented proper cyclic selection of cells in GCs, somatic hypermutation and proliferation were maintained. Class switch recombination was partly lost due to a failure of switch region targeting by activation-induced deaminase (AID).


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/inmunología , Centro Germinal/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Animales , Linfocitos B/citología , Separación Celular , Cromatografía Liquida , Citidina Desaminasa/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteína Forkhead Box O1 , Regulación de la Expresión Génica/inmunología , Centro Germinal/citología , Cambio de Clase de Inmunoglobulina/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Mutantes , Reacción en Cadena de la Polimerasa , Hipermutación Somática de Inmunoglobulina/inmunología , Espectrometría de Masas en Tándem
5.
Nat Immunol ; 12(3): 264-70, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21258321

RESUMEN

To elucidate the specific role of somatic hypermutation (SHM) in mucosal immunity, we generated mice carrying a knock-in point mutation in Aicda, which encodes activation-induced cytidine deaminase (AID), an enzyme essential to SHM and class-switch recombination (CSR). These mutant AID(G23S) mice had much less SHM but had normal amounts of immunoglobulin in both serum and intestinal secretions. AID(G23S) mice developed hyperplasia of germinal center B cells in gut-associated lymphoid tissues, accompanied by expansion of microflora in the small intestine. Moreover, AID(G23S) mice had more translocation of Yersinia enterocolitica into mesenteric lymph nodes and were more susceptible than wild-type mice to oral challenge with cholera toxin. Together our results indicate that SHM is critical in maintaining intestinal homeostasis and efficient mucosal defense.


Asunto(s)
Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , Homeostasis/inmunología , Mucosa Intestinal/inmunología , Intestinos/inmunología , Mutación , Animales , Técnicas de Sustitución del Gen , Genotipo , Homeostasis/genética , Ratones , Hipermutación Somática de Inmunoglobulina/genética , Hipermutación Somática de Inmunoglobulina/inmunología
6.
Nat Immunol ; 12(2): 160-6, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21186367

RESUMEN

During immunoglobulin class-switch recombination (CSR), the cytidine deaminase AID induces double-strand breaks into transcribed, repetitive DNA elements called switch sequences. The mechanism that promotes the binding of AID specifically to switch regions remains to be elucidated. Here we used a proteomic screen with in vivo biotinylation of AID to identify the splicing regulator PTBP2 as a protein that interacts with AID. Knockdown of PTBP2 mediated by short hairpin RNA in B cells led to a decrease in binding of AID to transcribed switch regions, which resulted in considerable impairment of CSR. PTBP2 is thus an effector of CSR that promotes the binding of AID to switch-region DNA.


Asunto(s)
Citidina Desaminasa/metabolismo , ADN/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , ADN/genética , Cambio de Clase de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/inmunología , Unión Proteica/genética , ARN Interferente Pequeño/genética , Activación Transcripcional/genética , Activación Transcripcional/inmunología , Transgenes/genética
7.
Nat Immunol ; 11(2): 148-54, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19966806

RESUMEN

Activation-induced cytidine deaminase (AID) is essential for the generation of antibody memory but also targets oncogenes, among other genes. We investigated the transcriptional regulation of Aicda (which encodes AID) in class switch-inducible CH12F3-2 cells and found that Aicda regulation involved derepression by several layers of positive regulatory elements in addition to the 5' promoter region. The 5' upstream region contained functional motifs for the response to signaling by cytokines, the ligand for the costimulatory molecule CD40 or stimuli that activated the transcription factor NF-kappaB. The first intron contained functional binding elements for the ubiquitous silencers c-Myb and E2f and for the B cell-specific activator Pax5 and E-box-binding proteins. Our results show that Aicda is regulated by the balance between B cell-specific and stimulation-responsive elements and ubiquitous silencers.


Asunto(s)
Linfocitos B/inmunología , Citidina Desaminasa/genética , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica/inmunología , Genes de Inmunoglobulinas/genética , Elementos Silenciadores Transcripcionales/genética , Animales , Citidina Desaminasa/inmunología , Elementos de Facilitación Genéticos/inmunología , Expresión Génica , Perfilación de la Expresión Génica , Genes de Inmunoglobulinas/inmunología , Humanos , Cambio de Clase de Inmunoglobulina/genética , Cambio de Clase de Inmunoglobulina/inmunología , Memoria Inmunológica/genética , Memoria Inmunológica/inmunología , Ratones , Mutagénesis Sitio-Dirigida , Análisis de Secuencia por Matrices de Oligonucleótidos , Oncogenes/genética , Oncogenes/inmunología , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Elementos Silenciadores Transcripcionales/inmunología , Hipermutación Somática de Inmunoglobulina/genética , Hipermutación Somática de Inmunoglobulina/inmunología
8.
J Immunol ; 204(5): 1091-1100, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31988182

RESUMEN

Almost a decade has passed since the approval of belimumab, an mAb directed against B lymphocyte stimulation and the first targeted therapy approved for systemic lupus erythematous (SLE) in over 50 y. Although well tolerated, the efficacy of belimumab remains limited and is not labeled for patients suffering from nephritis, the leading cause of patient mortality. We sought to explore alternative targets of autoreactive B lymphocytes through manipulation of affinity maturation. The BXSB/MpJ mouse, a well-established model of human SLE, develops elevated antinuclear Abs and immune complex-mediated nephritis along with other manifestations of SLE-like disease. To limit interfering with critical background genetics, we used CRISPR-Cas9 to disrupt activation-induced cytidine deaminase (AID; Aicda) directly in BXSB zygotes. Homozygous null mice demonstrated significantly prolonged survival compared with wild-type. Although mice continued to develop plasma cells, splenic follicular structure was restored, and renal pathology was reduced. Mice developed expanded germinal center B lymphocyte populations as in other models of AID deficiency as well as increased populations of CD73+ B lymphocytes. Treatment with the small molecule inhibitor of RAD51, 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid, resulted in minimal changes in disease markers in BXSB mice. The prolonged survival in AID-deficient BXSB mice appears attributed primarily to the reduced renal pathology, warranting further exploration, as current therapeutics targeting lupus nephritis are limited and, thus, in great demand.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Citidina Desaminasa/inmunología , Lupus Eritematoso Sistémico/inmunología , Animales , Subgrupos de Linfocitos B/patología , Sistemas CRISPR-Cas , Citidina Desaminasa/genética , Modelos Animales de Enfermedad , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Noqueados
9.
PLoS Genet ; 15(6): e1007721, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31199803

RESUMEN

B-cell activation yields abundant cell death in parallel to clonal amplification and remodeling of immunoglobulin (Ig) genes by activation-induced deaminase (AID). AID promotes affinity maturation of Ig variable regions and class switch recombination (CSR) in mature B lymphocytes. In the IgH locus, these processes are under control of the 3' regulatory region (3'RR) super-enhancer, a region demonstrated in the mouse to be both transcribed and itself targeted by AID-mediated recombination. Alternatively to CSR, IgH deletions joining Sµ to "like-switch" DNA repeats that flank the 3' super-enhancer can thus accomplish so-called "locus suicide recombination" (LSR) in mouse B-cells. Using an optimized LSR-seq high throughput method, we now show that AID-mediated LSR is evolutionarily conserved and also actively occurs in humans, providing an activation-induced cell death pathway in multiple conditions of B-cell activation. LSR either focuses on the functional IgH allele or is bi-allelic, and its signature is mainly detected when LSR is ongoing while it vanishes from fully differentiated plasma cells or from "resting" blood memory B-cells. Highly diversified breakpoints are distributed either within the upstream (3'RR1) or downstream (3'RR2) copies of the IgH 3' super-enhancer and all conditions activating CSR in vitro also seem to trigger LSR although TLR ligation appeared the most efficient. Molecular analysis of breakpoints and junctions confirms that LSR is AID-dependent and reveals junctional sequences somehow similar to CSR junctions but with increased usage of microhomologies.


Asunto(s)
Linfocitos B/inmunología , Citidina Desaminasa/genética , Región de Cambio de la Inmunoglobulina/genética , Inmunoglobulinas/inmunología , Alelos , Animales , Diferenciación Celular/genética , Citidina Desaminasa/inmunología , Marcación de Gen , Humanos , Región de Cambio de la Inmunoglobulina/inmunología , Tejido Linfoide/inmunología , Ratones , Tonsila Palatina/inmunología , Tonsila Palatina/metabolismo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Secuencias Reguladoras de Ácidos Nucleicos
10.
Eur J Immunol ; 50(9): 1307-1320, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32346855

RESUMEN

In addition to canonical TCR and BCR, cartilaginous fish assemble noncanonical TCR that employ various B-cell components. For example, shark T cells associate alpha (TCR-α) or delta (TCR-δ) constant (C) regions with Ig heavy chain (H) variable (V) segments or TCR-associated Ig-like V (TAILV) segments to form chimeric IgV-TCR, and combine TCRδC with both Ig-like and TCR-like V segments to form the doubly rearranging NAR-TCR. Activation-induced (cytidine) deaminase-catalyzed somatic hypermutation (SHM), typically used for B-cell affinity maturation, also is used by TCR-α during selection in the shark thymus presumably to salvage failing receptors. Here, we found that the use of SHM by nurse shark TCR varies depending on the particular V segment or C region used. First, SHM significantly alters alpha/delta V (TCRαδV) segments using TCR αC but not δC. Second, mutation to IgHV segments associated with TCR δC was reduced compared to mutation to TCR αδV associated with TCR αC. Mutation was present but limited in V segments of all other TCR chains including NAR-TCR. Unexpectedly, we found preferential rearrangement of the noncanonical IgHV-TCRδC over canonical TCR αδV-TCRδC receptors. The differential use of SHM may reveal how activation-induced (cytidine) deaminase targets V regions.


Asunto(s)
Citidina Desaminasa/inmunología , Reordenamiento Génico de la Cadena alfa de los Receptores de Antígenos de los Linfocitos T/genética , Reordenamiento Génico de la Cadena delta de los Receptores de Antígenos de los Linfocitos T/genética , Cadenas Pesadas de Inmunoglobulina/genética , Tiburones/inmunología , Hipermutación Somática de Inmunoglobulina/genética , Animales , Citidina Desaminasa/genética , Tiburones/genética
11.
Immunogenetics ; 73(1): 17-33, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33449123

RESUMEN

Immunoglobulins and T cell receptors (TCR) have obvious structural similarities as well as similar immunogenetic diversification and selection mechanisms. Nevertheless, the two receptor systems and the loci that encode them are distinct in humans and classical murine models, and the gene segments comprising each repertoire are mutually exclusive. Additionally, while both B and T cells employ recombination-activating genes (RAG) for primary diversification, immunoglobulins are afforded a supplementary set of activation-induced cytidine deaminase (AID)-mediated diversification tools. As the oldest-emerging vertebrates sharing the same adaptive B and T cell receptor systems as humans, extant cartilaginous fishes allow a potential view of the ancestral immune system. In this review, we discuss breakthroughs we have made in studies of nurse shark (Ginglymostoma cirratum) T cell receptors demonstrating substantial integration of loci and diversification mechanisms in primordial B and T cell repertoires. We survey these findings in this shark model where they were first described, while noting corroborating examples in other vertebrate groups. We also consider other examples where the gnathostome common ancestry of the B and T cell receptor systems have allowed dovetailing of genomic elements and AID-based diversification approaches for the TCR. The cartilaginous fish seem to have retained this T/B cell plasticity to a greater extent than more derived vertebrate groups, but representatives in all vertebrate taxa except bony fish and placental mammals show such plasticity.


Asunto(s)
Inmunoglobulinas/genética , Mamíferos/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos/genética , Tiburones/inmunología , Inmunidad Adaptativa , Animales , Citidina Desaminasa/inmunología , Evolución Molecular , Humanos , Mamíferos/genética , Tiburones/genética
12.
Immunogenetics ; 73(1): 5-16, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33159554

RESUMEN

Jawless vertebrates diverged from an ancestor of jawed vertebrates approximately 550 million years ago. They mount adaptive immune responses to repetitive antigenic challenges, despite lacking major histocompatibility complex molecules, immunoglobulins, T cell receptors, and recombination-activating genes. Instead of B cell and T cell receptors, agnathan lymphocytes express unique antigen receptors named variable lymphocyte receptors (VLRs), which generate diversity through a gene conversion-like mechanism. Although gnathostome antigen receptors and VLRs are structurally unrelated, jawed and jawless vertebrates share essential features of lymphocyte-based adaptive immunity, including the expression of a single type of receptor on each lymphocyte, clonal expansion of antigen-stimulated lymphocytes, and the dichotomy of cellular and humoral immunity, indicating that the backbone of the adaptive immune system was established in a common ancestor of all vertebrates. Furthermore, recent evidence indicates that, unlike previously thought, agnathans have a unique classical pathway of complement activation where VLRB molecules act as antibodies instead of immunoglobulins. It seems likely that the last common ancestor of all vertebrates had an adaptive immune system resembling that of jawless vertebrates, suggesting that, as opposed to jawed vertebrates, agnathans have retained the prototype of vertebrate adaptive immunity.


Asunto(s)
Inmunidad Adaptativa/genética , Inmunidad Adaptativa/inmunología , Vertebrados/inmunología , Animales , Anticuerpos/genética , Anticuerpos/inmunología , Evolución Biológica , Vía Clásica del Complemento , Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , Citocinas/genética , Citocinas/inmunología , Inmunidad Innata , Linfocitos/citología , Linfocitos/inmunología , Receptores de Antígenos/genética , Receptores de Antígenos/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Vertebrados/genética
13.
Cancer Immunol Immunother ; 70(8): 2275-2289, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33507341

RESUMEN

B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is the most common childhood malignancy. The two-step BCP-ALL pathogenesis requires in utero-induced chromosomal aberrations and additional mutagenic events for overt leukemia. In mouse models, activation-induced cytidine deaminase (AID/AICDA) was suggested to contribute to BCP-ALL pathogenesis by off-target mutagenic activity. The role of AID in patients, however, remains unclear. Moreover, AID is usually not expressed in precursor B-cells but in germinal center B-cells, where it is induced upon T-helper (Th) cell stimulation. We have previously demonstrated that autologous Th-cells supportively interacted with BCP-ALL-cells. Here, we hypothesize that this interaction additionally induces AID expression in BCP-ALL-cells, leading to off-target mutagenic activity. We show that co-culture with autologous bone marrow Th-cells induced high AICDA expression in primary BCP-ALL-cells. This induction was mediated by a mechanism similar to the induction in mature B-cells involving IL-13/Stat6, CD40L/NF-κB and TGFß/Smad2/3 signaling. Even though Th-cell-induced AID seemed to be active in vitro in a BCP-ALL reporter cell line, extensive mutational signature analysis revealed no major contribution of AID activity to the mutational landscape in BCP-ALL patients. AID activity was neither detected in mutation clusters nor in known AID targets. Moreover, no recurrently mutated gene showed a relevant enrichment of mutations in the AID motif. Together, the lack of AID-induced mutational consequences argues towards a Th-cell-promoted yet AID-independent BCP-ALL pathogenesis and favors therapeutic research focusing on Th-cell-derived support of BCP-ALL-cells rather than AID-induced effects.


Asunto(s)
Médula Ósea/inmunología , Citidina Desaminasa/inmunología , Linfoma de Células B/inmunología , Mutagénesis/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Adolescente , Adulto , Linfocitos B/inmunología , Línea Celular Tumoral , Células Cultivadas , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Mutación/inmunología , Transducción de Señal/inmunología , Adulto Joven
14.
J Virol ; 95(1)2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33028711

RESUMEN

Gammaherpesviruses (GHVs) are DNA tumor viruses that establish lifelong, chronic infections in lymphocytes of humans and other mammals. GHV infections are associated with numerous cancers, especially in immunocompromised hosts. While it is known that GHVs utilize host germinal center (GC) B cell responses during latency establishment, an understanding of how viral gene products function in specific B cell subsets to regulate this process is incomplete. Using murine gammaherpesvirus 68 (MHV68) as a small-animal model to define mechanisms of GHV pathogenesis in vivo, we generated a virus in which the M2 gene was flanked by loxP sites (M2.loxP), enabling the use of Cre-lox technology to define M2 function in specific cell types in infection and disease. The M2 gene encodes a protein that is highly expressed in GC B cells that promotes plasma cell differentiation and viral reactivation. M2 was efficiently deleted in Cre-expressing cells, and the presence of loxP sites flanking M2 did not alter viral replication or latency in mice that do not express Cre. In contrast, M2.loxP MHV68 exhibited a deficit in latency establishment and reactivation that resembled M2-null virus, following intranasal (IN) infection of mice that express Cre in all B cells (CD19-Cre). Nearly identical phenotypes were observed for M2.loxP MHV68 in mice that express Cre in germinal center (GC) B cells (AID-Cre). However, colonization of neither draining lymph nodes after IN infection nor the spleen after intraperitoneal (IP) infection required M2, although the reactivation defect was retained. Together, these data confirm that M2 function is B cell-specific and demonstrate that M2 primarily functions in AID-expressing cells to facilitate MHV68 dissemination to distal latency reservoirs within the host and reactivation from latency. Our study reveals that a viral latency gene functions within a distinct subset of cells to facilitate host colonization.IMPORTANCE Gammaherpesviruses establish lifelong chronic infections in cells of the immune system that can lead to lymphomas and other diseases. To facilitate colonization of a host, gammaherpesviruses encode gene products that manipulate processes involved in cellular proliferation and differentiation. Whether and how these viral gene products function in specific cells of the immune system is poorly defined. We report here the use of a viral genetic system that allows for deletion of specific viral genes in discrete populations of cells. We employ this system in an in vivo model to demonstrate cell-type-specific requirements for a particular viral gene. Our findings reveal that a viral gene product can function in distinct cellular subsets to direct gammaherpesvirus pathogenesis.


Asunto(s)
Linfocitos B/inmunología , Citidina Desaminasa/inmunología , Infecciones por Herpesviridae/virología , Rhadinovirus/fisiología , Proteínas Virales/inmunología , Activación Viral , Animales , Antígenos CD19/metabolismo , Linfocitos B/virología , Diferenciación Celular , Proliferación Celular , Centro Germinal/inmunología , Centro Germinal/virología , Infecciones por Herpesviridae/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/virología , Ratones , Rhadinovirus/genética , Rhadinovirus/metabolismo , Proteínas Virales/genética , Latencia del Virus
15.
PLoS Pathog ; 15(6): e1007880, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31211814

RESUMEN

The largest ever recorded epidemic of the Chikungunya virus (CHIKV) broke out in 2004 and affected four continents. Acute symptomatic infections are typically associated with the onset of fever and often debilitating polyarthralgia/polyarthritis. In this study, a systems biology approach was adopted to analyze the blood transcriptomes of adults acutely infected with the CHIKV. Gene signatures that were associated with viral RNA levels and the onset of symptoms were identified. Among these genes, the putative role of the Eukaryotic Initiation Factor (eIF) family genes and apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC3A) in the CHIKV replication process were displayed. We further compared these signatures with signatures induced by the Dengue virus infection and rheumatoid arthritis. Finally, we demonstrated that the CHIKV in vitro infection of murine bone marrow-derived macrophages induced IL-1 beta production in a mechanism that is significantly dependent on the inflammasome NLRP3 activation. The observations provided valuable insights into virus-host interactions during the acute phase and can be instrumental in the investigation of new and effective therapeutic interventions.


Asunto(s)
Artritis/inmunología , Fiebre Chikungunya/inmunología , Virus Chikungunya/fisiología , Citidina Desaminasa/inmunología , Proteínas/inmunología , Replicación Viral/inmunología , Adulto , Animales , Artritis/patología , Artritis/virología , Fiebre Chikungunya/patología , Virus del Dengue/inmunología , Virus del Dengue/patogenicidad , Femenino , Fiebre/inmunología , Fiebre/patología , Fiebre/virología , Estudios de Seguimiento , Humanos , Interleucina-1beta/inmunología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología
16.
Nat Immunol ; 10(4): 420-6, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19234474

RESUMEN

Immunoglobulin class-switch recombination (CSR) requires activation-induced cytidine deaminase (AID). Deamination of DNA by AID in transcribed switch (S) regions leads to double-stranded breaks in DNA that serve as obligatory CSR intermediates. Here we demonstrate that the catalytic and regulatory subunits of protein kinase A (PKA) were specifically recruited to S regions to promote the localized phosphorylation of AID, which led to binding of replication protein A and subsequent propagation of the CSR cascade. Accordingly, inactivation of PKA resulted in considerable disruption of CSR because of decreased AID phosphorylation and recruitment of replication protein A to S regions. We propose that PKA nucleates the formation of active AID complexes specifically on S regions to generate the high density of DNA lesions required for CSR.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/inmunología , Citidina Desaminasa/inmunología , Cambio de Clase de Inmunoglobulina , Recombinación Genética/inmunología , Proteína de Replicación A/inmunología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Roturas del ADN de Doble Cadena , Ratones , Ratones Mutantes , Fosforilación , Unión Proteica , Infecciones por Retroviridae/inmunología
17.
Nat Immunol ; 10(11): 1147-53, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19841648

RESUMEN

The discovery and characterization of activation-induced cytidine deaminase (AID) 10 years ago provided the basis for a mechanistic understanding of secondary antibody diversification and the subsequent generation and maintenance of cellular memory in B lymphocytes, which signified a major advance in the field of B cell immunology. Here we celebrate and review the triumphs in the mission to understand the mechanisms through which AID influences antibody diversification, as well as the implications of AID function on human physiology. We also take time to point out important ongoing controversies and outstanding questions in the field and highlight key experiments and techniques that hold the potential to elucidate the remaining mysteries surrounding this vital protein.


Asunto(s)
Diversidad de Anticuerpos , Citidina Desaminasa/genética , Procesamiento Proteico-Postraduccional , Procesamiento Postranscripcional del ARN , Linfocitos B/inmunología , Linfocitos B/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citidina Desaminasa/inmunología , Citidina Desaminasa/metabolismo , Regulación de la Expresión Génica , Transcripción Genética
18.
Immunity ; 37(3): 399-411, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22999946

RESUMEN

Viral infections are often detrimental to host survival and reproduction. Consequently, hosts have evolved a variety of mechanisms to defend themselves against viruses. A component of this arsenal is a set of proteins, termed restriction factors, which exhibit direct antiviral activity. Among these are several classes of proteins (APOBEC3, TRIM5, Tetherin, and SAMHD1) that inhibit the replication of human and simian immunodeficiency viruses. Here, we outline the features, mechanisms, and evolution of these defense mechanisms. We also speculate on how restriction factors arose, how they might interact with the conventional innate and adaptive immune systems, and how an understanding of these intrinsic cellular defenses might be usefully exploited.


Asunto(s)
Resistencia a la Enfermedad/inmunología , Infecciones por VIH/inmunología , VIH/inmunología , Interacciones Huésped-Patógeno/inmunología , Desaminasa APOBEC-3G , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos CD/inmunología , Antígenos CD/metabolismo , Citidina Desaminasa/inmunología , Citidina Desaminasa/metabolismo , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , VIH/fisiología , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Humanos , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Modelos Inmunológicos , Proteínas de Unión al GTP Monoméricas/inmunología , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteína 1 que Contiene Dominios SAM y HD
19.
Int J Med Sci ; 18(16): 3788-3793, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790054

RESUMEN

As the world is racing to develop perpetual immunity to the SARS-CoV-2 virus. The emergence of new viral strains, together with vaccination and reinfections, are all contributing to a long-term immunity against the deadly virus that has taken over the world since its introduction to humans in late December 2019. The discovery that more than 95 percent of people who recovered from COVID-19 had long-lasting immunity and that asymptomatic people have a different immune response to SARS-CoV-2 than symptomatic people has shifted attention to how our immune system initiates such diverse responses. These findings have provided reason to believe that SARS-CoV-2 days are numbered. Hundreds of research papers have been published on the causes of long-lasting immune responses and variations in the numbers of different immune cell types in COVID 19 survivors, but the main reason of these differences has still not been adequately identified. In this article, we focus on the activation-induced cytidine deaminase (AID), which initiates molecular processes that allow our immune system to generate antibodies against SARS-CoV-2. To establish lasting immunity to SARS-CoV-2, we suggest that AID could be the key to unlocking it.


Asunto(s)
COVID-19/inmunología , Citidina Desaminasa/genética , Inmunidad/genética , SARS-CoV-2/inmunología , COVID-19/virología , Citidina/genética , Citidina/inmunología , Citidina Desaminasa/inmunología , Desaminación/inmunología , Humanos , SARS-CoV-2/patogenicidad , Vacunación
20.
Proc Natl Acad Sci U S A ; 115(19): 4921-4926, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29669924

RESUMEN

Activation-induced deaminase (AID) initiates hypermutation of Ig genes in activated B cells by converting C:G into U:G base pairs. G1-phase variants of uracil base excision repair (BER) and mismatch repair (MMR) then deploy translesion polymerases including REV1 and Pol η, which exacerbates mutation. dNTP paucity may contribute to hypermutation, because dNTP levels are reduced in G1 phase to inhibit viral replication. To derestrict G1-phase dNTP supply, we CRISPR-inactivated SAMHD1 (which degrades dNTPs) in germinal center B cells. Samhd1 inactivation increased B cell virus susceptibility, increased transition mutations at C:G base pairs, and substantially decreased transversion mutations at A:T and C:G base pairs in both strands. We conclude that SAMHD1's restriction of dNTP supply enhances AID's mutagenicity and that the evolution of Ig hypermutation included the repurposing of antiviral mechanisms based on dNTP starvation.


Asunto(s)
Linfocitos B/inmunología , Fase G1/inmunología , Activación de Linfocitos , Mutación , Proteína 1 que Contiene Dominios SAM y HD , Hipermutación Somática de Inmunoglobulina/inmunología , Animales , Linfocitos B/citología , Citidina Desaminasa/inmunología , ADN Polimerasa Dirigida por ADN , Fase G1/genética , Masculino , Ratones , Ratones Transgénicos , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/inmunología , Proteína 1 que Contiene Dominios SAM y HD/genética , Proteína 1 que Contiene Dominios SAM y HD/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA