Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Exp Med ; 219(9)2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35796685

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to be a global problem in part because of the emergence of variants of concern that evade neutralization by antibodies elicited by prior infection or vaccination. Here we report on human neutralizing antibody and memory responses to the Gamma variant in a cohort of hospitalized individuals. Plasma from infected individuals potently neutralized viruses pseudotyped with Gamma SARS-CoV-2 spike protein, but neutralizing activity against Wuhan-Hu-1-1, Beta, Delta, or Omicron was significantly lower. Monoclonal antibodies from memory B cells also neutralized Gamma and Beta pseudoviruses more effectively than Wuhan-Hu-1. 69% and 34% of Gamma-neutralizing antibodies failed to neutralize Delta or Wuhan-Hu-1. Although Class 1 and 2 antibodies dominate the response to Wuhan-Hu-1 or Beta, 54% of antibodies elicited by Gamma infection recognized Class 3 epitopes. The results have implications for variant-specific vaccines and infections, suggesting that exposure to variants generally provides more limited protection to other variants.


Assuntos
COVID-19 , SARS-CoV-2 , Anticorpos Neutralizantes , Anticorpos Antivirais , Formação de Anticorpos , Humanos , Glicoproteínas de Membrana/metabolismo , Testes de Neutralização , Glicoproteína da Espícula de Coronavírus , Proteínas do Envelope Viral
2.
J Exp Med ; 218(5)2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33831141

RESUMO

Tick-borne encephalitis virus (TBEV) is an emerging human pathogen that causes potentially fatal disease with no specific treatment. Mouse monoclonal antibodies are protective against TBEV, but little is known about the human antibody response to infection. Here, we report on the human neutralizing antibody response to TBEV in a cohort of infected and vaccinated individuals. Expanded clones of memory B cells expressed closely related anti-envelope domain III (EDIII) antibodies in both groups of volunteers. However, the most potent neutralizing antibodies, with IC50s below 1 ng/ml, were found only in individuals who recovered from natural infection. These antibodies also neutralized other tick-borne flaviviruses, including Langat, louping ill, Omsk hemorrhagic fever, Kyasanur forest disease, and Powassan viruses. Structural analysis revealed a conserved epitope near the lateral ridge of EDIII adjoining the EDI-EDIII hinge region. Prophylactic or early therapeutic antibody administration was effective at low doses in mice that were lethally infected with TBEV.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Encefalite Transmitida por Carrapatos/imunologia , Imunoglobulina G/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/genética , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/genética , Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/genética , Células Cultivadas , Estudos de Coortes , Reações Cruzadas/imunologia , Vírus da Encefalite Transmitidos por Carrapatos/efeitos dos fármacos , Vírus da Encefalite Transmitidos por Carrapatos/fisiologia , Encefalite Transmitida por Carrapatos/prevenção & controle , Encefalite Transmitida por Carrapatos/virologia , Epitopos/imunologia , Feminino , Humanos , Imunoglobulina G/administração & dosagem , Camundongos Endogâmicos BALB C , Homologia de Sequência de Aminoácidos , Análise de Sobrevida , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
3.
Nature ; 591(7851): 639-644, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33461210

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected 78 million individuals and is responsible for over 1.7 million deaths to date. Infection is associated with the development of variable levels of antibodies with neutralizing activity, which can protect against infection in animal models1,2. Antibody levels decrease with time, but, to our knowledge, the nature and quality of the memory B cells that would be required to produce antibodies upon reinfection has not been examined. Here we report on the humoral memory response in a cohort of 87 individuals assessed at 1.3 and 6.2 months after infection with SARS-CoV-2. We find that titres of IgM and IgG antibodies against the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 decrease significantly over this time period, with IgA being less affected. Concurrently, neutralizing activity in plasma decreases by fivefold in pseudotype virus assays. By contrast, the number of RBD-specific memory B cells remains unchanged at 6.2 months after infection. Memory B cells display clonal turnover after 6.2 months, and the antibodies that they express have greater somatic hypermutation, resistance to RBD mutations and increased potency, indicative of continued evolution of the humoral response. Immunofluorescence and PCR analyses of intestinal biopsies obtained from asymptomatic individuals at 4 months after the onset of coronavirus disease 2019 (COVID-19) revealed the persistence of SARS-CoV-2 nucleic acids and immunoreactivity in the small bowel of 7 out of 14 individuals. We conclude that the memory B cell response to SARS-CoV-2 evolves between 1.3 and 6.2 months after infection in a manner that is consistent with antigen persistence.


Assuntos
Anticorpos Antivirais/imunologia , COVID-19/imunologia , Imunidade Humoral/imunologia , SARS-CoV-2/imunologia , Adolescente , Adulto , Idoso , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/genética , Antígenos Virais/química , Antígenos Virais/genética , Antígenos Virais/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Biópsia , COVID-19/sangue , Estudos de Coortes , Imunofluorescência , Humanos , Imunidade Humoral/genética , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Memória Imunológica/imunologia , Intestinos/imunologia , Pessoa de Meia-Idade , Mutação , Hipermutação Somática de Imunoglobulina , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Fatores de Tempo , Adulto Jovem
4.
Sci Transl Med ; 13(577)2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33288661

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19), primarily infects cells at mucosal surfaces. Serum neutralizing antibody responses are variable and generally low in individuals that suffer mild forms of COVID-19. Although potent immunoglobulin G (IgG) antibodies can neutralize the virus, less is known about secretory antibodies such as IgA that might affect the initial viral spread and transmissibility from the mucosa. Here, we characterize the IgA response to SARS-CoV-2 in a cohort of 149 convalescent individuals after diagnosis with COVID-19. IgA responses in plasma generally correlated with IgG responses. Furthermore, clones of IgM-, IgG-, and IgA-producing B cells were derived from common progenitor cells. Plasma IgA monomers specific to SARS-CoV-2 proteins were demonstrated to be twofold less potent than IgG equivalents. However, IgA dimers, the primary form of antibody in the nasopharynx, were, on average, 15 times more potent than IgA monomers against the same target. Thus, dimeric IgA responses may be particularly valuable for protection against SARS-CoV-2 and for vaccine efficacy.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , COVID-19/diagnóstico , Imunoglobulina A/sangue , SARS-CoV-2/imunologia , Animais , Biomarcadores/sangue , COVID-19/sangue , COVID-19/imunologia , COVID-19/virologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Convalescença , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Multimerização Proteica , Células Vero
5.
Cell Host Microbe ; 28(2): 335-349.e6, 2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32504577

RESUMO

Although there is no effective cure for chronic hepatitis B virus (HBV) infection, antibodies are protective and correlate with recovery from infection. To examine the human antibody response to HBV, we screened 124 vaccinated and 20 infected, spontaneously recovered individuals. The selected individuals produced shared clones of broadly neutralizing antibodies (bNAbs) that targeted 3 non-overlapping epitopes on the HBV S antigen (HBsAg). Single bNAbs protected humanized mice against infection but selected for resistance mutations in mice with prior established infection. In contrast, infection was controlled by a combination of bNAbs targeting non-overlapping epitopes with complementary sensitivity to mutations that commonly emerge during human infection. The co-crystal structure of one of the bNAbs with an HBsAg peptide epitope revealed a stabilized hairpin loop. This structure, which contains residues frequently mutated in clinical immune escape variants, provides a molecular explanation for why immunotherapy for HBV infection may require combinations of complementary bNAbs.


Assuntos
Anticorpos Amplamente Neutralizantes/imunologia , Anticorpos Anti-Hepatite B/imunologia , Antígenos de Superfície da Hepatite B/imunologia , Vírus da Hepatite B/imunologia , Animais , Anticorpos Monoclonais/imunologia , Linhagem Celular Tumoral , Pré-Escolar , Modelos Animais de Doenças , Epitopos/imunologia , Feminino , Células HEK293 , Células Hep G2 , Hepatite B Crônica/tratamento farmacológico , Hepatite B Crônica/imunologia , Humanos , Lactente , Camundongos , Camundongos Knockout , Conformação Proteica
6.
Mol Cell ; 72(4): 636-649.e8, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30293785

RESUMO

Class switch recombination (CSR) is a DNA recombination reaction that diversifies the effector component of antibody responses. CSR is initiated by activation-induced cytidine deaminase (AID), which targets transcriptionally active immunoglobulin heavy chain (Igh) switch donor and acceptor DNA. The 3' Igh super-enhancer, 3' regulatory region (3'RR), is essential for acceptor region transcription, but how this function is regulated is unknown. Here, we identify the chromatin reader ZMYND8 as an essential regulator of the 3'RR. In B cells, ZMYND8 binds promoters and super-enhancers, including the Igh enhancers. ZMYND8 controls the 3'RR activity by modulating the enhancer transcriptional status. In its absence, there is increased 3'RR polymerase loading and decreased acceptor region transcription and CSR. In addition to CSR, ZMYND8 deficiency impairs somatic hypermutation (SHM) of Igh, which is also dependent on the 3'RR. Thus, ZMYND8 controls Igh diversification in mature B lymphocytes by regulating the activity of the 3' Igh super-enhancer.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Switching de Imunoglobulina/genética , Cadeias Pesadas de Imunoglobulinas/genética , Proteínas Supressoras de Tumor/genética , Animais , Linfócitos B , Linhagem Celular , Cromatina/genética , Cromatina/metabolismo , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , DNA/genética , Elementos Facilitadores Genéticos , Rearranjo Gênico , Humanos , Domínios MYND , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico , Hipermutação Somática de Imunoglobulina/genética , Proteínas Supressoras de Tumor/metabolismo
7.
Nature ; 543(7646): 559-563, 2017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-28289286

RESUMO

Highly potent and broadly neutralizing anti-HIV-1 antibodies (bNAbs) have been used to prevent and treat lentivirus infections in humanized mice, macaques, and humans. In immunotherapy experiments, administration of bNAbs to chronically infected animals transiently suppresses virus replication, which invariably returns to pre-treatment levels and results in progression to clinical disease. Here we show that early administration of bNAbs in a macaque simian/human immunodeficiency virus (SHIV) model is associated with very low levels of persistent viraemia, which leads to the establishment of T-cell immunity and resultant long-term infection control. Animals challenged with SHIVAD8-EO by mucosal or intravenous routes received a single 2-week course of two potent passively transferred bNAbs (3BNC117 and 10-1074 (refs 13, 14)). Viraemia remained undetectable for 56-177 days, depending on bNAb half-life in vivo. Moreover, in the 13 treated monkeys, plasma virus loads subsequently declined to undetectable levels in 6 controller macaques. Four additional animals maintained their counts of T cells carrying the CD4 antigen (CD4+) and very low levels of viraemia persisted for over 2 years. The frequency of cells carrying replication-competent virus was less than 1 per 106 circulating CD4+ T cells in the six controller macaques. Infusion of a T-cell-depleting anti-CD8ß monoclonal antibody to the controller animals led to a specific decline in levels of CD8+ T cells and the rapid reappearance of plasma viraemia. In contrast, macaques treated for 15 weeks with combination anti-retroviral therapy, beginning on day 3 after infection, experienced sustained rebound plasma viraemia when treatment was interrupted. Our results show that passive immunotherapy during acute SHIV infection differs from combination anti-retroviral therapy in that it facilitates the emergence of potent CD8+ T-cell immunity able to durably suppress virus replication.


Assuntos
Infecções por HIV/imunologia , Infecções por HIV/terapia , HIV/imunologia , Imunização Passiva , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/terapia , Vírus da Imunodeficiência Símia/imunologia , Animais , Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/uso terapêutico , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Terapia Combinada , Modelos Animais de Doenças , Feminino , HIV/efeitos dos fármacos , HIV/isolamento & purificação , Anticorpos Anti-HIV/administração & dosagem , Anticorpos Anti-HIV/imunologia , Anticorpos Anti-HIV/uso terapêutico , Infecções por HIV/virologia , Meia-Vida , Macaca mulatta , Masculino , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Vírus da Imunodeficiência Símia/isolamento & purificação , Carga Viral/efeitos dos fármacos , Carga Viral/imunologia , Viremia/imunologia , Viremia/terapia , Replicação Viral/efeitos dos fármacos , Replicação Viral/imunologia
8.
Proc Natl Acad Sci U S A ; 111(52): 18667-72, 2014 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-25512519

RESUMO

Activation-induced cytidine deaminase (AID) initiates class switch recombination (CSR) and somatic hypermutation (SHM) by deaminating cytosine residues in immunoglobulin genes (Igh, Igκ, and Igλ). At a lower frequency, AID also causes collateral DNA damage at non-Ig loci, including genes that are rearranged or mutated in B-cell lymphoma. Precisely how AID is recruited to these off-target sites is not entirely understood. To gain further insight into how AID selects its targets, we compared AID-mediated translocations in two different cell types, B cells and mouse embryonic fibroblasts (MEFs). AID targets a distinct set of hotspots in the two cell types. In both cases, hotspots are concentrated in highly transcribed but stalled genes. However, transcription alone is insufficient to recruit AID activity. Comparison of genes similarly transcribed in B cells and MEFs but targeted in only one of the two cell types reveals a common set of epigenetic features associated with AID recruitment in both cells. AID target genes are enriched in chromatin modifications associated with active enhancers (such as H3K27Ac) and marks of active transcription (such as H3K36me3) in both fibroblasts and B cells, indicating that these features are universal mediators of AID recruitment.


Assuntos
Linfócitos B/enzimologia , Citidina Desaminase , Embrião de Mamíferos/enzimologia , Epigênese Genética , Marcação de Genes , Transcrição Gênica/fisiologia , Animais , Linfócitos B/citologia , Linhagem Celular , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Fibroblastos/enzimologia , Histonas/genética , Histonas/metabolismo , Imunoglobulinas/biossíntese , Imunoglobulinas/genética , Camundongos , Camundongos Knockout
9.
Cell ; 159(7): 1524-37, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25483777

RESUMO

The antibody gene mutator activation-induced cytidine deaminase (AID) promiscuously damages oncogenes, leading to chromosomal translocations and tumorigenesis. Why nonimmunoglobulin loci are susceptible to AID activity is unknown. Here, we study AID-mediated lesions in the context of nuclear architecture and the B cell regulome. We show that AID targets are not randomly distributed across the genome but are predominantly grouped within super-enhancers and regulatory clusters. Unexpectedly, in these domains, AID deaminates active promoters and eRNA(+) enhancers interconnected in some instances over megabases of linear chromatin. Using genome editing, we demonstrate that 3D-linked targets cooperate to recruit AID-mediated breaks. Furthermore, a comparison of hypermutation in mouse B cells, AID-induced kataegis in human lymphomas, and translocations in MEFs reveals that AID damages different genes in different cell types. Yet, in all cases, the targets are predominantly associated with topological complex, highly transcribed super-enhancers, demonstrating that these compartments are key mediators of AID recruitment.


Assuntos
Linfócitos B/metabolismo , Carcinogênese , Citidina Desaminase/genética , Elementos Facilitadores Genéticos , Animais , Dano ao DNA , Humanos , Linfoma/metabolismo , Camundongos
10.
J Exp Med ; 211(12): 2361-72, 2014 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-25385756

RESUMO

Antibody-mediated immunotherapy is effective in humanized mice when combinations of broadly neutralizing antibodies (bNAbs) are used that target nonoverlapping sites on the human immunodeficiency virus type 1 (HIV-1) envelope. In contrast, single bNAbs can control simian-human immunodeficiency virus (SHIV) infection in immune-competent macaques, suggesting that the host immune response might also contribute to the control of viremia. Here, we investigate how the autologous antibody response in intact hosts can contribute to the success of immunotherapy. We find that frequently arising antibodies that normally fail to control HIV-1 infection can synergize with passively administered bNAbs by preventing the emergence of bNAb viral escape variants.


Assuntos
Anticorpos Neutralizantes/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Imunoterapia/métodos , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Anticorpos Neutralizantes/uso terapêutico , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Infecções por HIV/terapia , Infecções por HIV/virologia , HIV-1/genética , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macaca mulatta , Camundongos Endogâmicos NOD , Camundongos Knockout , Mutação/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/terapia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/fisiologia , Proteínas do Envelope Viral/imunologia , Carga Viral/imunologia
11.
J Exp Med ; 211(10): 2061-74, 2014 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-25155019

RESUMO

It is widely appreciated that effective human vaccines directed against viral pathogens elicit neutralizing antibodies (NAbs). The passive transfer of anti-HIV-1 NAbs conferring sterilizing immunity to macaques has been used to determine the plasma neutralization titers, which must be present at the time of exposure, to prevent acquisition of SIV/HIV chimeric virus (SHIV) infections. We administered five recently isolated potent and broadly acting anti-HIV neutralizing monoclonal antibodies (mAbs) to rhesus macaques and challenged them intrarectally 24 h later with either of two different R5-tropic SHIVs. By combining the results obtained from 60 challenged animals, we determined that the protective neutralization titer in plasma preventing virus infection in 50% of the exposed monkeys was relatively modest (∼1:100) and potentially achievable by vaccination.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Anti-HIV/imunologia , HIV/imunologia , Infecções por Lentivirus/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Neutralizantes/administração & dosagem , Primers do DNA/genética , Anticorpos Anti-HIV/administração & dosagem , Humanos , Macaca mulatta , Mutagênese , Testes de Neutralização , Análise de Regressão , Vacinação/métodos
12.
J Exp Med ; 210(1): 115-23, 2013 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-23254285

RESUMO

DNA double-strand breaks (DSBs) are byproducts of normal cellular metabolism and obligate intermediates in antigen receptor diversification reactions. These lesions are potentially dangerous because they can lead to deletion of genetic material or chromosome translocation. The chromatin-binding protein 53BP1 and the histone variant H2AX are required for efficient class switch (CSR) and V(D)J recombination in part because they protect DNA ends from resection and thereby favor nonhomologous end joining (NHEJ). Here, we examine the mechanism of DNA end resection in primary B cells. We find that resection depends on both CtBP-interacting protein (CtIP, Rbbp8) and exonuclease 1 (Exo1). Inhibition of CtIP partially rescues the CSR defect in 53BP1- and H2AX-deficient lymphocytes, as does interference with the RecQ helicases Bloom (Blm) and Werner (Wrn). We conclude that CtIP, Exo1, and RecQ helicases contribute to the metabolism of DNA ends during DSB repair in B lymphocytes and that minimizing resection favors efficient CSR.


Assuntos
Linfócitos B/fisiologia , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Exodesoxirribonucleases/metabolismo , Isotipos de Imunoglobulinas/genética , Recombinação Genética , Animais , Sequência de Bases , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases , Endonucleases , Exodesoxirribonucleases/genética , Histonas/genética , Histonas/metabolismo , Isotipos de Imunoglobulinas/metabolismo , Proteína Homóloga a MRE11 , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , RecQ Helicases/genética , RecQ Helicases/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Recombinação V(D)J , Helicase da Síndrome de Werner
13.
Adv Cancer Res ; 113: 167-90, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22429855

RESUMO

DNA damage, rearrangement, and mutation of the human genome are the basis of carcinogenesis and thought to be avoided at all costs. An exception is the adaptive immune system where lymphocytes utilize programmed DNA damage to effect antigen receptor diversification. Both B and T lymphocytes diversify their antigen receptors through RAG1/2 mediated recombination, but B cells undergo two additional processes--somatic hypermutation (SHM) and class-switch recombination (CSR), both initiated by activation-induced cytidine deaminase (AID). AID deaminates cytidines in DNA resulting in U:G mismatches that are processed into point mutations in SHM or double-strand breaks in CSR. Although AID activity is focused at Immunoglobulin (Ig) gene loci, it also targets a wide array of non-Ig genes including oncogenes associated with lymphomas. Here, we review the molecular basis of AID regulation, targeting, and initiation of CSR and SHM, as well as AID's role in generating chromosome translocations that contribute to lymphomagenesis.


Assuntos
Diversidade de Anticorpos/genética , Transformação Celular Neoplásica/genética , Citidina Desaminase/genética , Switching de Imunoglobulina/genética , Hipermutação Somática de Imunoglobulina/genética , Animais , Anticorpos/genética , Citidina Desaminase/imunologia , Citidina Desaminase/metabolismo , Dano ao DNA/genética , Reparo do DNA , Genes de Imunoglobulinas , Humanos , Switching de Imunoglobulina/imunologia , Camundongos , Hipermutação Somática de Imunoglobulina/imunologia , Transcrição Gênica , Translocação Genética
14.
Mol Cell Biol ; 31(3): 442-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21135131

RESUMO

Activation-induced cytidine deaminase (AID) is a mutator enzyme that initiates class switch recombination and somatic hypermutation of immunoglobulin genes (Ig) in B lymphocytes. However, AID also produces off-target DNA damage, including mutations in oncogenes and double-stranded breaks that can serve as substrates for oncogenic chromosomal translocations. AID is strictly regulated by a number of mechanisms, including phosphorylation at serine 38 and threonine 140, which increase activity. Here we show that phosphorylation can also suppress AID activity in vivo. Serine 3 is a novel phospho-acceptor which, when mutated to alanine, leads to increased class switching and c-myc/IgH translocations without affecting AID levels or catalytic activity. Conversely, increasing AID phosphorylation specifically on serine 3 by interfering with serine/threonine protein phosphatase 2A (PP2A) leads to decreased class switching. We conclude that AID activity and its oncogenic potential can be downregulated by phosphorylation of serine 3 and that this process is controlled by PP2A.


Assuntos
Citidina Desaminase/química , Citidina Desaminase/metabolismo , Cadeias Pesadas de Imunoglobulinas/genética , Proteínas Proto-Oncogênicas c-myc/genética , Translocação Genética , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Switching de Imunoglobulina/efeitos dos fármacos , Camundongos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Proteína Fosfatase 2/antagonistas & inibidores , Proteína Fosfatase 2/metabolismo , Recombinação Genética/efeitos dos fármacos , Recombinação Genética/genética , Hipermutação Somática de Imunoglobulina/efeitos dos fármacos , Hipermutação Somática de Imunoglobulina/genética , Translocação Genética/efeitos dos fármacos
15.
Cell ; 143(1): 122-33, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20887897

RESUMO

Activation-induced cytidine deaminase (AID) initiates antibody gene diversification by creating U:G mismatches. However, AID is not specific for antibody genes; Off-target lesions can activate oncogenes or cause chromosome translocations. Despite its importance in these transactions little is known about how AID finds its targets. We performed an shRNA screen to identify factors required for class switch recombination (CSR) of antibody loci. We found that Spt5, a factor associated with stalled RNA polymerase II (Pol II) and single stranded DNA (ssDNA), is required for CSR. Spt5 interacts with AID, it facilitates association between AID and Pol II, and AID recruitment to its Ig and non-Ig targets. ChIP-seq experiments reveal that Spt5 colocalizes with AID and stalled Pol II. Further, Spt5 accumulation at sites of Pol II stalling is predictive of AID-induced mutation. We propose that AID is targeted to sites of Pol II stalling in part via its association with Spt5.


Assuntos
Linfócitos B/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Citidina Desaminase/metabolismo , Switching de Imunoglobulina , RNA Polimerase II/metabolismo , Fatores de Elongação da Transcrição/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Fibroblastos/metabolismo , Humanos , Imunoglobulinas/genética , Camundongos
16.
Science ; 329(5994): 917-23, 2010 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-20671152

RESUMO

Programmed genetic rearrangements in lymphocytes require transcription at antigen receptor genes to promote accessibility for initiating double-strand break (DSB) formation critical for DNA recombination and repair. Here, we showed that activated B cells deficient in the PTIP component of the MLL3 (mixed-lineage leukemia 3)-MLL4 complex display impaired trimethylation of histone 3 at lysine 4 (H3K4me3) and transcription initiation of downstream switch regions at the immunoglobulin heavy-chain (Igh) locus, leading to defective immunoglobulin class switching. We also showed that PTIP accumulation at DSBs contributes to class switch recombination (CSR) and genome stability independently of Igh switch transcription. These results demonstrate that PTIP promotes specific chromatin changes that control the accessibility of the Igh locus to CSR and suggest a nonredundant role for the MLL3-MLL4 complex in altering antibody effector function.


Assuntos
Proteínas de Transporte/fisiologia , Switching de Imunoglobulina/fisiologia , Proteínas Nucleares/fisiologia , Animais , Especificidade de Anticorpos/genética , Proteínas de Transporte/genética , Citidina Desaminase/metabolismo , DNA , Proteínas de Ligação a DNA , Histonas/metabolismo , Switching de Imunoglobulina/genética , Região de Troca de Imunoglobulinas , Metilação , Camundongos , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Recombinação Genética , Ativação Transcricional
17.
J Exp Med ; 207(4): 855-65, 2010 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-20368578

RESUMO

Class switch recombination (CSR) diversifies antibodies by joining highly repetitive DNA elements, which are separated by 60-200 kbp. CSR is initiated by activation-induced cytidine deaminase, an enzyme that produces multiple DNA double-strand breaks (DSBs) in switch regions. Switch regions are joined by a mechanism that requires an intact DNA damage response and classical or alternative nonhomologous end joining (A-NHEJ). Among the DNA damage response factors, 53BP1 has the most profound effect on CSR. We explore the role of 53BP1 in intrachromosomal DNA repair using I-SceI to introduce paired DSBs in the IgH locus. We find that the absence of 53BP1 results in an ataxia telangiectasia mutated-dependent increase in DNA end resection and that resected DNA is preferentially repaired by microhomology-mediated A-NHEJ. We propose that 53BP1 favors long-range CSR in part by protecting DNA ends against resection, which prevents A-NHEJ-dependent short-range rejoining of intra-switch region DSBs.


Assuntos
Reparo do DNA/fisiologia , Switching de Imunoglobulina/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Recombinação Genética/fisiologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona , Pareamento Cromossômico/genética , Citidina Desaminase/genética , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Desoxirribonucleases de Sítio Específico do Tipo II/genética , Cadeias Pesadas de Imunoglobulinas/genética , Integrases/genética , Interleucina-4/farmacologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
18.
J Exp Med ; 205(11): 2585-94, 2008 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-18838546

RESUMO

Activation-induced cytidine deaminase (AID) is a mutator enzyme that initiates somatic mutation and class switch recombination in B lymphocytes by introducing uracil:guanine mismatches into DNA. Repair pathways process these mismatches to produce point mutations in the Ig variable region or double-stranded DNA breaks in the switch region DNA. However, AID can also produce off-target DNA damage, including mutations in oncogenes. Therefore, stringent regulation of AID is required for maintaining genomic stability during maturation of the antibody response. It has been proposed that AID phosphorylation at serine 38 (S38) regulates its activity, but this has not been tested in vivo. Using a combination of mass spectrometry and immunochemical approaches, we found that in addition to S38, AID is also phosphorylated at position threonine 140 (T140). Mutation of either S38 or T140 to alanine does not impact catalytic activity, but interferes with class switching and somatic hypermutation in vivo. This effect is particularly pronounced in haploinsufficient mice where AID levels are limited. Although S38 is equally important for both processes, T140 phosphorylation preferentially affects somatic mutation, suggesting that posttranslational modification might contribute to the choice between hypermutation and class switching.


Assuntos
Linfócitos B/fisiologia , Citidina Desaminase/metabolismo , Switching de Imunoglobulina/fisiologia , Mutação Puntual/genética , Animais , Linfócitos B/metabolismo , Imuno-Histoquímica , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Fosforilação
19.
Immunity ; 28(5): 630-8, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18455451

RESUMO

MicroRNAs (miRNAs) are small noncoding RNAs that regulate vast networks of genes that share miRNA target sequences. To examine the physiologic effects of an individual miRNA-mRNA interaction in vivo, we generated mice that carry a mutation in the putative microRNA-155 (miR-155) binding site in the 3'-untranslated region of activation-induced cytidine deaminase (AID), designated Aicda(155) mice. AID is required for immunoglobulin gene diversification in B lymphocytes, but it also promotes chromosomal translocations. Aicda(155) caused an increase in steady-state Aicda mRNA and protein amounts by increasing the half-life of the mRNA, resulting in a high degree of Myc-Igh translocations. A similar but more pronounced translocation phenotype was also found in miR-155-deficient mice. Our experiments indicate that miR-155 can act as a tumor suppressor by reducing potentially oncogenic translocations generated by AID.


Assuntos
Linfócitos B/enzimologia , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Genes myc , Cadeias Pesadas de Imunoglobulinas/genética , MicroRNAs/metabolismo , Translocação Genética , Regiões 3' não Traduzidas , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Genes de Imunoglobulinas , Switching de Imunoglobulina , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Mutantes , MicroRNAs/genética , Mutação , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Hipermutação Somática de Imunoglobulina
20.
Nat Immunol ; 5(4): 443-50, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15021880

RESUMO

We have identified a silencer and an antisilencing element that interact at a distance of 85 kilobases to regulate expression of the recombination activating genes Rag1 and Rag2 in thymocytes. Transgenic experiments showed that Rag promoter-proximal cis elements directed tissue-specific expression and that a Runx-dependent intergenic silencer suppressed expression in developing T cells. Deletion of the antisilencing element from the genomic Rag locus unmasked the intergenic silencer and abrogated Rag expression in developing CD4(+)CD8(+) T cells. We speculate that the Rag antisilencing element belongs to a class of cis elements that might be useful for genome diversification by activating genes encoded by otherwise silent transposable elements.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Genes RAG-1 , Elementos Silenciadores Transcricionais , Timo/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , Deleção de Sequência , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA