Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33658386

RESUMO

V(D)J recombination generates mature B cells that express huge repertoires of primary antibodies as diverse immunoglobulin (Ig) heavy chain (IgH) and light chain (IgL) of their B cell antigen receptors (BCRs). Cognate antigen binding to BCR variable region domains activates B cells into the germinal center (GC) reaction in which somatic hypermutation (SHM) modifies primary variable region-encoding sequences, with subsequent selection for mutations that improve antigen-binding affinity, ultimately leading to antibody affinity maturation. Based on these principles, we developed a humanized mouse model approach to diversify an anti-PD1 therapeutic antibody and allow isolation of variants with novel properties. In this approach, component Ig gene segments of the anti-PD1 antibody underwent de novo V(D)J recombination to diversify the anti-PD1 antibody in the primary antibody repertoire in the mouse models. Immunization of these mouse models further modified the anti-PD1 antibodies through SHM. Known anti-PD1 antibodies block interaction of PD1 with its ligands to alleviate PD1-mediated T cell suppression, thereby boosting antitumor T cell responses. By diversifying one such anti-PD1 antibody, we derived many anti-PD1 antibodies, including anti-PD1 antibodies with the opposite activity of enhancing PD1/ligand interaction. Such antibodies theoretically might suppress deleterious T cell activities in autoimmune diseases. The approach we describe should be generally applicable for diversifying other therapeutic antibodies.


Assuntos
Afinidade de Anticorpos/genética , Cadeias Pesadas de Imunoglobulinas , Cadeias Leves de Imunoglobulina , Receptores de Antígenos de Linfócitos B , Hipermutação Somática de Imunoglobulina , Recombinação V(D)J/imunologia , Animais , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Leves de Imunoglobulina/genética , Cadeias Leves de Imunoglobulina/imunologia , Camundongos , Receptores de Antígenos de Linfócitos B/genética , Receptores de Antígenos de Linfócitos B/imunologia
2.
Cell Rep ; 5(3): 654-665, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24210820

RESUMO

Nonalcoholic fatty liver disease is the most common chronic liver disorder in developed countries. Its pathogenesis is poorly understood, and therapeutic options are limited. Here, we show that SIRT7, an NAD(+)-dependent H3K18Ac deacetylase, functions at chromatin to suppress ER stress and prevent the development of fatty liver disease. SIRT7 is induced upon ER stress and is stabilized at the promoters of ribosomal proteins through its interaction with the transcription factor Myc to silence gene expression and to relieve ER stress. SIRT7-deficient mice develop chronic hepatosteatosis resembling human fatty liver disease. Myc inactivation or pharmacological suppression of ER stress alleviates fatty liver caused by SIRT7 deficiency. Importantly, SIRT7 suppresses ER stress and reverts the fatty liver disease in diet-induced obese mice. Our study identifies SIRT7 as a cofactor of Myc for transcriptional repression and delineates a druggable regulatory branch of the ER stress response that prevents and reverts fatty liver disease.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Fígado Gorduroso/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Sirtuínas/metabolismo , Animais , Feminino , Genes myc , Células HEK293 , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/genética , Sirtuínas/deficiência , Sirtuínas/genética , Transfecção
3.
Nature ; 492(7428): 199-204, 2012 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-23201684

RESUMO

Although initially viewed as unregulated, increasing evidence suggests that cellular necrosis often proceeds through a specific molecular program. In particular, death ligands such as tumour necrosis factor (TNF)-α activate necrosis by stimulating the formation of a complex containing receptor-interacting protein 1 (RIP1) and receptor-interacting protein 3 (RIP3). Relatively little is known regarding how this complex formation is regulated. Here, we show that the NAD-dependent deacetylase SIRT2 binds constitutively to RIP3 and that deletion or knockdown of SIRT2 prevents formation of the RIP1-RIP3 complex in mice. Furthermore, genetic or pharmacological inhibition of SIRT2 blocks cellular necrosis induced by TNF-α. We further demonstrate that RIP1 is a critical target of SIRT2-dependent deacetylation. Using gain- and loss-of-function mutants, we demonstrate that acetylation of RIP1 lysine 530 modulates RIP1-RIP3 complex formation and TNF-α-stimulated necrosis. In the setting of ischaemia-reperfusion injury, RIP1 is deacetylated in a SIRT2-dependent fashion. Furthermore, the hearts of Sirt2(-/-) mice, or wild-type mice treated with a specific pharmacological inhibitor of SIRT2, show marked protection from ischaemic injury. Taken together, these results implicate SIRT2 as an important regulator of programmed necrosis and indicate that inhibitors of this deacetylase may constitute a novel approach to protect against necrotic injuries, including ischaemic stroke and myocardial infarction.


Assuntos
Necrose/enzimologia , Sirtuína 2/genética , Sirtuína 2/metabolismo , Acetilação , Animais , Linhagem Celular , Feminino , Células HEK293 , Células HeLa , Humanos , Células Jurkat , Masculino , Camundongos , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Ligação Proteica , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
4.
Mol Cell Biol ; 31(5): 1041-53, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21199920

RESUMO

Bright/Arid3a has been characterized both as an activator of immunoglobulin heavy-chain transcription and as a proto-oncogene. Although Bright expression is highly B lineage stage restricted in adult mice, its expression in the earliest identifiable hematopoietic stem cell (HSC) population suggests that Bright might have additional functions. We showed that >99% of Bright(-/-) embryos die at midgestation from failed hematopoiesis. Bright(-/-) embryonic day 12.5 (E12.5) fetal livers showed an increase in the expression of immature markers. Colony-forming assays indicated that the hematopoietic potential of Bright(-/-) mice is markedly reduced. Rare survivors of lethality, which were not compensated by the closely related paralogue Bright-derived protein (Bdp)/Arid3b, suffered HSC deficits in their bone marrow as well as B lineage-intrinsic developmental and functional deficiencies in their peripheries. These include a reduction in a natural antibody, B-1 responses to phosphocholine, and selective T-dependent impairment of IgG1 class switching. Our results place Bright/Arid3a on a select list of transcriptional regulators required to program both HSC and lineage-specific differentiation.


Assuntos
Linfócitos B/citologia , Proteínas de Ligação a DNA/metabolismo , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Linfopoese/genética , Fatores de Transcrição/metabolismo , Animais , Anticorpos/sangue , Linfócitos B/metabolismo , Linhagem da Célula , Proteínas de Ligação a DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes Letais , Células-Tronco Hematopoéticas/metabolismo , Switching de Imunoglobulina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilcolina/imunologia , Fosforilcolina/metabolismo , Fatores de Transcrição/genética
5.
Nature ; 469(7329): 250-4, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-21160472

RESUMO

Classical non-homologous DNA end-joining (NHEJ) is a major mammalian DNA double-strand-break (DSB) repair pathway. Deficiencies for classical NHEJ factors, such as XRCC4, abrogate lymphocyte development, owing to a strict requirement for classical NHEJ to join V(D)J recombination DSB intermediates. The XRCC4-like factor (XLF; also called NHEJ1) is mutated in certain immunodeficient human patients and has been implicated in classical NHEJ; however, XLF-deficient mice have relatively normal lymphocyte development and their lymphocytes support normal V(D)J recombination. The ataxia telangiectasia-mutated protein (ATM) detects DSBs and activates DSB responses by phosphorylating substrates including histone H2AX. However, ATM deficiency causes only modest V(D)J recombination and lymphocyte developmental defects, and H2AX deficiency does not have a measurable impact on these processes. Here we show that XLF, ATM and H2AX all have fundamental roles in processing and joining DNA ends during V(D)J recombination, but that these roles have been masked by unanticipated functional redundancies. Thus, combined deficiency of ATM and XLF nearly blocks mouse lymphocyte development due to an inability to process and join chromosomal V(D)J recombination DSB intermediates. Combined XLF and ATM deficiency also severely impairs classical NHEJ, but not alternative end-joining, during IgH class switch recombination. Redundant ATM and XLF functions in classical NHEJ are mediated by ATM kinase activity and are not required for extra-chromosomal V(D)J recombination, indicating a role for chromatin-associated ATM substrates. Correspondingly, conditional H2AX inactivation in XLF-deficient pro-B lines leads to V(D)J recombination defects associated with marked degradation of unjoined V(D)J ends, revealing that H2AX has a role in this process.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Rearranjo Gênico do Linfócito B , Histonas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Recombinação Genética , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Linhagem Celular Transformada , Cromatina/metabolismo , Cromossomos de Mamíferos/genética , Cromossomos de Mamíferos/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Rearranjo Gênico do Linfócito B/genética , Camundongos , Células Precursoras de Linfócitos B/citologia , Células Precursoras de Linfócitos B/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
6.
Proc Natl Acad Sci U S A ; 107(7): 3024-9, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20133615

RESUMO

DNA ligase IV (LIG4) is an essential component of the nonhomologous end-joining (NHEJ) repair pathway and plays a key role in V(D)J recombination. Hypomorphic LIG4 mutations in humans are associated with increased cellular radiosensitivity, microcephaly, facial dysmorphisms, growth retardation, developmental delay, and a variable degree of immunodeficiency. We have generated a knock-in mouse model with a homozygous Lig4 R278H mutation that corresponds to the first LIG4 mutation reported in humans. The phenotype of homozygous mutant mice Lig4(R278H/R278H) (Lig4(R/R)) includes growth retardation, a decreased life span, a severe cellular sensitivity to ionizing radiation, and a very severe, but incomplete block in T and B cell development. Peripheral T lymphocytes show an activated and anergic phenotype, reduced viability, and a restricted repertoire, reminiscent of human leaky SCID. Genomic instability is associated with a high rate of thymic tumor development. Finally, Lig4(R/R) mice spontaneously produce low-affinity antibodies that include autoreactive specificities, but are unable to mount high-affinity antibody responses. These findings highlight the importance of LIG4 in lymphocyte development and function, and in genomic stability maintenance, and provide a model for the complex phenotype of LIG4 syndrome in humans.


Assuntos
Anormalidades Múltiplas/genética , Formação de Anticorpos/genética , DNA Ligases/genética , Deficiências do Desenvolvimento/genética , Modelos Animais de Doenças , Mutação de Sentido Incorreto/genética , Imunodeficiência Combinada Severa/genética , Animais , Apoptose/imunologia , Southern Blotting , Criança , DNA Ligase Dependente de ATP , DNA Ligases/imunologia , Citometria de Fluxo , Humanos , Imunoglobulinas/sangue , Imunofenotipagem , Camundongos , Mutação de Sentido Incorreto/imunologia , Síndrome
7.
Blood ; 113(13): 2965-75, 2009 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-19126872

RESUMO

The RAG1/2 endonuclease initiates programmed DNA rearrangements in progenitor lymphocytes by generating double-strand breaks at specific recombination signal sequences. This process, known as V(D)J recombination, assembles the vastly diverse antigen receptor genes from numerous V, D, and J coding segments. In vitro biochemical and cellular transfection studies suggest that RAG1/2 may also play postcleavage roles by forming complexes with the recombining ends to facilitate DNA end processing and ligation. In the current study, we examine the in vivo consequences of a mutant form of RAG1, RAG1-S723C, that is proficient for DNA cleavage, yet exhibits defects in postcleavage complex formation and end joining in vitro. We generated a knockin mouse model harboring the RAG1-S723C hypomorphic mutation and examined the immune system in this fully in vivo setting. RAG1-S723C homozygous mice exhibit impaired lymphocyte development and decreased V(D)J rearrangements. Distinct from RAG nullizygosity, the RAG1-S723C hypomorph results in aberrant DNA double-strand breaks within rearranging loci. RAG1-S723C also predisposes to thymic lymphomas associated with chromosomal translocations in a p53 mutant background, and heterozygosity for the mutant allele accelerates age-associated immune system dysfunction. Thus, our study provides in vivo evidence that implicates aberrant RAG1/2 activity in lymphoid tumor development and premature immunosenescence.


Assuntos
Rearranjo Gênico/genética , Proteínas de Homeodomínio/genética , Mutação de Sentido Incorreto , Imunodeficiência Combinada Severa/genética , Envelhecimento/genética , Envelhecimento/imunologia , Substituição de Aminoácidos/fisiologia , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Técnicas de Introdução de Genes , Homozigoto , Linfoma/genética , Linfoma/imunologia , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto/fisiologia , Fenótipo , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/patologia , Linfócitos T/imunologia , Linfócitos T/patologia , Neoplasias do Timo/genética , Neoplasias do Timo/imunologia , Éxons VDJ
8.
Mol Cell ; 31(5): 631-40, 2008 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-18775323

RESUMO

Mutations in XLF/Cernunnos (XLF) cause lymphocytopenia in humans, and various studies suggest an XLF role in classical nonhomologous end joining (C-NHEJ). We now find that XLF-deficient mouse embryonic fibroblasts are ionizing radiation (IR) sensitive and severely impaired for ability to support V(D)J recombination. Yet mature lymphocyte numbers in XLF-deficient mice are only modestly decreased. Moreover, XLF-deficient pro-B lines, while IR-sensitive, perform V(D)J recombination at nearly wild-type levels. Correspondingly, XLF/p53-double-deficient mice are not markedly prone to the pro-B lymphomas that occur in previously characterized C-NHEJ/p53-deficient mice; however, like other C-NHEJ/p53-deficient mice, they still develop medulloblastomas. Despite nearly normal V(D)J recombination in developing B cells, XLF-deficient mature B cells are moderately defective for immunoglobulin heavy-chain class switch recombination. Together, our results implicate XLF as a C-NHEJ factor but also indicate that developing mouse lymphocytes harbor cell-type-specific factors/pathways that compensate for the absence of XLF function during V(D)J recombination.


Assuntos
Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Linfócitos/fisiologia , Recombinação Genética , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/fisiologia , Células Cultivadas , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/genética , Feminino , Fibroblastos/citologia , Fibroblastos/fisiologia , Rearranjo Gênico , Humanos , Switching de Imunoglobulina , Linfócitos/citologia , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
9.
Cell ; 124(2): 315-29, 2006 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-16439206

RESUMO

The Sir2 histone deacetylase functions as a chromatin silencer to regulate recombination, genomic stability, and aging in budding yeast. Seven mammalian Sir2 homologs have been identified (SIRT1-SIRT7), and it has been speculated that some may have similar functions to Sir2. Here, we demonstrate that SIRT6 is a nuclear, chromatin-associated protein that promotes resistance to DNA damage and suppresses genomic instability in mouse cells, in association with a role in base excision repair (BER). SIRT6-deficient mice are small and at 2-3 weeks of age develop abnormalities that include profound lymphopenia, loss of subcutaneous fat, lordokyphosis, and severe metabolic defects, eventually dying at about 4 weeks. We conclude that one function of SIRT6 is to promote normal DNA repair, and that SIRT6 loss leads to abnormalities in mice that overlap with aging-associated degenerative processes.


Assuntos
Envelhecimento/metabolismo , Doenças Genéticas Inatas/genética , Instabilidade Genômica , Sirtuínas/genética , Sirtuínas/fisiologia , Animais , Proliferação de Células , Cromatina/metabolismo , Dano ao DNA , Reparo do DNA , Doenças Genéticas Inatas/patologia , Humanos , Antígeno Ki-1/metabolismo , Linfócitos/imunologia , Camundongos , Camundongos Knockout , Fenótipo , Tolerância a Radiação , Transdução de Sinais , Sirtuínas/deficiência
10.
Cell Metab ; 2(1): 67-76, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16054100

RESUMO

The Saccharomyces cerevisiae chromatin silencing factor Sir2 suppresses genomic instability and extends replicative life span. In contrast, we find that mouse embryonic fibroblasts (MEFs) deficient for SIRT1, a mammalian Sir2 homolog, have dramatically increased resistance to replicative senescence. Extended replicative life span of SIRT1-deficient MEFs correlates with enhanced proliferative capacity under conditions of chronic, sublethal oxidative stress. In this context, SIRT1-deficient cells fail to normally upregulate either the p19(ARF) senescence regulator or its downstream target p53. However, upon acute DNA damage or oncogene expression, SIRT1-deficient cells show normal p19(ARF) induction and cell cycle arrest. Together, our findings demonstrate an unexpected SIRT1 function in promoting replicative senescence in response to chronic cellular stress and implicate p19(ARF) as a downstream effector in this pathway.


Assuntos
Senescência Celular , Dano ao DNA , Sirtuínas/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina , Dano ao DNA/efeitos dos fármacos , Doxorrubicina/farmacologia , Fibroblastos , Genes ras/genética , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Knockout , Células NIH 3T3 , Estresse Oxidativo/efeitos dos fármacos , Fase S/efeitos dos fármacos , Sirtuína 1 , Sirtuínas/deficiência , Sirtuínas/genética , Proteína Supressora de Tumor p14ARF/metabolismo
11.
Science ; 303(5666): 2011-5, 2004 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-14976264

RESUMO

The Sir2 deacetylase modulates organismal life-span in various species. However, the molecular mechanisms by which Sir2 increases longevity are largely unknown. We show that in mammalian cells, the Sir2 homolog SIRT1 appears to control the cellular response to stress by regulating the FOXO family of Forkhead transcription factors, a family of proteins that function as sensors of the insulin signaling pathway and as regulators of organismal longevity. SIRT1 and the FOXO transcription factor FOXO3 formed a complex in cells in response to oxidative stress, and SIRT1 deacetylated FOXO3 in vitro and within cells. SIRT1 had a dual effect on FOXO3 function: SIRT1 increased FOXO3's ability to induce cell cycle arrest and resistance to oxidative stress but inhibited FOXO3's ability to induce cell death. Thus, one way in which members of the Sir2 family of proteins may increase organismal longevity is by tipping FOXO-dependent responses away from apoptosis and toward stress resistance.


Assuntos
Histona Desacetilases/metabolismo , Estresse Oxidativo , Sirtuínas/metabolismo , Fatores de Transcrição/metabolismo , Acetilação , Animais , Apoptose , Ciclo Celular , Linhagem Celular , Núcleo Celular/metabolismo , Células Cultivadas , Cerebelo/citologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Histona Desacetilases/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Neurônios/citologia , Fosforilação , Proteínas/genética , Proteínas Recombinantes/metabolismo , Sirtuína 1 , Sirtuínas/genética , Fatores de Transcrição/genética , Transcrição Gênica , Proteínas GADD45
12.
Proc Natl Acad Sci U S A ; 100(19): 10794-9, 2003 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-12960381

RESUMO

SIRT1 is a mammalian homolog of the Saccharomyces cerevisiae chromatin silencing factor Sir2. Dominant-negative and overexpression studies have implicated a role for SIRT1 in deacetylating the p53 tumor suppressor protein to dampen apoptotic and cellular senescence pathways. To elucidate SIRT1 function in normal cells, we used gene-targeted mutation to generate mice that express either a mutant SIRT1 protein that lacks part of the catalytic domain or has no detectable SIRT1 protein at all. Both types of SIRT1 mutant mice and cells had essentially the same phenotypes. SIRT1 mutant mice were small, and exhibited notable developmental defects of the retina and heart, and only infrequently survived postnatally. Moreover, SIRT1-deficient cells exhibited p53 hyperacetylation after DNA damage and increased ionizing radiation-induced thymocyte apoptosis. In SIRT1-deficient embryonic fibroblasts, however, p53 hyperacetylation after DNA damage was not accompanied by increased p21 protein induction or DNA damage sensitivity. Together, our observations provide direct evidence that endogenous SIRT1 protein regulates p53 acetylation and p53-dependent apoptosis, and show that the function of this enzyme is required for specific developmental processes.


Assuntos
Anormalidades Congênitas/genética , Sirtuínas/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/fisiologia , Ciclo Celular/fisiologia , Dano ao DNA , Raios Infravermelhos , Camundongos , Camundongos Knockout , Sirtuína 2 , Sirtuínas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA