Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Immunol ; 6(66): eabj4026, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34919442

RESUMO

Despite the enormous promise of T cell therapies, the isolation and study of human T cell receptors (TCRs) of dedicated specificity remains a major challenge. To overcome this limitation, we generated mice with a genetically humanized system of T cell immunity. We used VelociGene technology to replace the murine TCRαß variable regions, along with regions encoding the extracellular domains of co-receptors CD4 and CD8, and major histocompatibility complex (MHC) class I and II, with corresponding human sequences. The resulting "VelociT" mice have normal myeloid and lymphoid immune cell populations, including thymic and peripheral αß T cell subsets comparable with wild-type mice. VelociT mice expressed a diverse TCR repertoire, mounted functional T cell responses to lymphocytic choriomeningitis virus infection, and could develop experimental autoimmune encephalomyelitis. Immunization of VelociT mice with human tumor-associated peptide antigens generated robust, antigen-specific responses and led to identification of a TCR against tumor antigen New York esophageal squamous cell carcinoma-1 with potent antitumor activity. These studies demonstrate that VelociT mice mount clinically relevant T cell responses to both MHC-I­ and MHC-II­restricted antigens, providing a powerful new model for analyzing T cell function in human disease. Moreover, VelociT mice are a new platform for de novo discovery of therapeutic human TCRs.


Assuntos
Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Linfócitos T/imunologia , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T alfa-beta/genética
2.
J Vis Exp ; (167)2021 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-33554965

RESUMO

Extensive studies have characterized the development and differentiation of murine B cells in secondary lymphoid organs. Antibodies secreted by B cells have been isolated and developed into well-established therapeutics. Validation of murine B cell development, in the context of autoimmune prone mice, or in mice with modified immune systems, is a crucial component of developing or testing therapeutic agents in mice and is an appropriate use of flow cytometry. Well established B cell flow cytometric parameters can be used to evaluate B cell development in the murine peritoneum, bone marrow, and spleen, but a number of best practices must be adhered to. In addition, flow cytometric analysis of B cell compartments should also complement additional readouts of B cell development. Data generated using this technique can further our understanding of wild type, autoimmune prone mouse models as well as humanized mice that can be used to generate antibody or antibody-like molecules as therapeutics.


Assuntos
Linfócitos B/citologia , Citometria de Fluxo/métodos , Animais , Linfócitos B/imunologia , Células da Medula Óssea/citologia , Contagem de Células , Diferenciação Celular , Separação Celular , Análise de Dados , Feminino , Cadeias lambda de Imunoglobulina/metabolismo , Imunoglobulinas/metabolismo , Ativação Linfocitária , Subpopulações de Linfócitos/citologia , Camundongos Endogâmicos C57BL , Peritônio/citologia , Baço/citologia , Coloração e Rotulagem
3.
Skelet Muscle ; 5: 34, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26457176

RESUMO

BACKGROUND: Loss of skeletal muscle mass and function in humans is associated with significant morbidity and mortality. The role of myostatin as a key negative regulator of skeletal muscle mass and function has supported the concept that inactivation of myostatin could be a useful approach for treating muscle wasting diseases. METHODS: We generated a myostatin monoclonal blocking antibody (REGN1033) and characterized its effects in vitro using surface plasmon resonance biacore and cell-based Smad2/3 signaling assays. REGN1033 was tested in mice for the ability to induce skeletal muscle hypertrophy and prevent atrophy induced by immobilization, hindlimb suspension, or dexamethasone. The effect of REGN1033 on exercise training was tested in aged mice. Messenger RNA sequencing, immunohistochemistry, and ex vivo force measurements were performed on skeletal muscle samples from REGN1033-treated mice. RESULTS: The human monoclonal antibody REGN1033 is a specific and potent myostatin antagonist. Chronic treatment of mice with REGN1033 increased muscle fiber size, muscle mass, and force production. REGN1033 prevented the loss of muscle mass induced by immobilization, glucocorticoid treatment, or hindlimb unweighting and increased the gain of muscle mass during recovery from pre-existing atrophy. In aged mice, REGN1033 increased muscle mass and strength and improved physical performance during treadmill exercise. CONCLUSIONS: We show that specific myostatin antagonism with the human antibody REGN1033 enhanced muscle mass and function in young and aged mice and had beneficial effects in models of skeletal muscle atrophy.

4.
Endocrinology ; 156(12): 4502-10, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26406932

RESUMO

Secreted frizzled-related protein 4 (SFRP4) is an extracellular regulator of the wingless-type mouse mammary tumor virus integration site family (WNT) pathway. SFRP4 has been implicated in adipocyte dysfunction, obesity, insulin resistance, and impaired insulin secretion in patients with type 2 diabetes. However, the exact role of SFRP4 in regulating whole-body metabolism and glucose homeostasis is unknown. We show here that male Sfrp4(-/-) mice have increased spine length and gain more weight when fed a high-fat diet. The body composition and body mass per spine length of diet-induced obese Sfrp4(-/-) mice is similar to wild-type littermates, suggesting that the increase in body weight can be accounted for by their longer body size. The diet-induced obese Sfrp4(-/-) mice have reduced energy expenditure, food intake, and bone mineral density. Sfrp4(-/-) mice have normal glucose and insulin tolerance and ß-cell mass. Diet-induced obese Sfrp4(-/-) and control mice show similar impairments of glucose tolerance and a 5-fold compensatory expansion of their ß-cell mass. In summary, our data suggest that loss of SFRP4 alters body length and bone mineral density as well as energy expenditure and food intake. However, SFRP4 does not control glucose homeostasis and ß-cell mass in mice.


Assuntos
Tamanho Corporal/genética , Densidade Óssea/genética , Dieta Hiperlipídica , Ingestão de Alimentos/genética , Metabolismo Energético/genética , Células Secretoras de Insulina/metabolismo , Obesidade , Proteínas Proto-Oncogênicas/genética , Animais , Glicemia/metabolismo , Composição Corporal/genética , Comportamento Alimentar , Técnicas de Introdução de Genes , Teste de Tolerância a Glucose , Células HEK293 , Homeostase/genética , Humanos , Insulina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Via de Sinalização Wnt , Microtomografia por Raio-X
5.
PLoS Genet ; 8(9): e1002948, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23028355

RESUMO

FoxO transcription factors and sirtuin family deacetylases regulate diverse biological processes, including stress responses and longevity. Here we show that the Caenorhabditis elegans sirtuin SIR-2.4--homolog of mammalian SIRT6 and SIRT7 proteins--promotes DAF-16-dependent transcription and stress-induced DAF-16 nuclear localization. SIR-2.4 is required for resistance to multiple stressors: heat shock, oxidative insult, and proteotoxicity. By contrast, SIR-2.4 is largely dispensable for DAF-16 nuclear localization and function in response to reduced insulin/IGF-1-like signaling. Although acetylation is known to regulate localization and activity of mammalian FoxO proteins, this modification has not been previously described on DAF-16. We find that DAF-16 is hyperacetylated in sir-2.4 mutants. Conversely, DAF-16 is acetylated by the acetyltransferase CBP-1, and DAF-16 is hypoacetylated and constitutively nuclear in response to cbp-1 inhibition. Surprisingly, a SIR-2.4 catalytic mutant efficiently rescues the DAF-16 localization defect in sir-2.4 null animals. Acetylation of DAF-16 by CBP-1 in vitro is inhibited by either wild-type or mutant SIR-2.4, suggesting that SIR-2.4 regulates DAF-16 acetylation indirectly, by preventing CBP-1-mediated acetylation under stress conditions. Taken together, our results identify SIR-2.4 as a critical regulator of DAF-16 specifically in the context of stress responses. Furthermore, they reveal a novel role for acetylation, modulated by the antagonistic activities of CBP-1 and SIR-2.4, in modulating DAF-16 localization and function.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Sirtuínas , Fatores de Transcrição , Acetilação , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/fisiologia , Núcleo Celular/genética , Núcleo Celular/metabolismo , Fatores de Transcrição Forkhead , Resposta ao Choque Térmico/genética , Resposta ao Choque Térmico/fisiologia , Histona Acetiltransferases/metabolismo , Longevidade/genética , Longevidade/fisiologia , Estresse Oxidativo/genética , Estresse Oxidativo/fisiologia , Transdução de Sinais , Sirtuínas/genética , Sirtuínas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia
6.
Aging Cell ; 8(5): 604-6, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19594485

RESUMO

Calorie restriction (CR) increases lifespan in organisms ranging from budding yeast through mammals. Mitochondrial adaptation represents a key component of the response to CR. Molecular mechanisms underlying this adaptation are largely unknown. Here we show that lysine acetylation of mitochondrial proteins is altered during CR in a tissue-specific fashion. Via large-scale mass spectrometry screening, we identify 72 candidate proteins involved in a variety of metabolic pathways with altered acetylation during CR. Mitochondrial acetylation changes may play an important role in the pro-longevity CR response.


Assuntos
Restrição Calórica/estatística & dados numéricos , Longevidade/fisiologia , Mamíferos/fisiologia , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias/metabolismo , Proteínas/metabolismo , Saccharomycetales/fisiologia , Acetilação , Animais , Humanos , Expectativa de Vida , Mamíferos/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Saccharomycetales/crescimento & desenvolvimento
8.
Cell ; 135(1): 85-96, 2008 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-18854157

RESUMO

The Mre11/Rad50/NBS1 (MRN) complex maintains genomic stability by bridging DNA ends and initiating DNA damage signaling through activation of the ATM kinase. Mre11 possesses DNA nuclease activities that are highly conserved in evolution but play unknown roles in mammals. To define the functions of Mre11, we engineered targeted mouse alleles that either abrogate nuclease activities or inactivate the entire MRN complex. Mre11 nuclease deficiency causes a striking array of phenotypes indistinguishable from the absence of MRN, including early embryonic lethality and dramatic genomic instability. We identify a crucial role for the nuclease activities in homology-directed double-strand-break repair and a contributing role in activating the ATR kinase. However, the nuclease activities are not required to activate ATM after DNA damage or telomere deprotection. Therefore, nucleolytic processing by Mre11 is an essential function of fundamental importance in DNA repair, distinct from MRN control of ATM signaling.


Assuntos
Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Instabilidade Genômica , Sequência de Aminoácidos , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Transformada , Proliferação de Células , Quebras de DNA de Cadeia Dupla , Dano ao DNA , Enzimas Reparadoras do DNA/química , Proteínas de Ligação a DNA/química , Fibroblastos/metabolismo , Proteína Homóloga a MRE11 , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Recombinação Genética , Telômero/metabolismo , Proteínas Supressoras de Tumor/metabolismo
9.
Hum Mol Genet ; 14(18): 2685-93, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16087684

RESUMO

The Mre11/Rad50/NBS1 (MRN) complex is mutated in inherited genomic instability syndromes featuring cancer predisposition, mental retardation and immunodeficiency. It functions both in DNA double-strand break repair and in controlling the ataxia telangiectasia mutated (ATM) kinase during the response to these lesions. Patients inheriting homozygosity for an NBS1 hypomorphic allele display reduced phosphorylation of signaling factors such as Chk1, but not of chromatin-associated factor H2AX, after stresses that activate the ATM-related kinase, ATR. Therefore, we tested whether MRN has a global controlling role over the ATR kinase through the study of MRN deficiencies generated via RNA interference. We show for the first time that MRN is required for ATR-dependent phosphorylation of structural maintenance of chromosomes 1 (Smc1), which acts within chromatin to ensure sister chromatid cohesion and to effect several DNA damage responses. We have uncovered novel phenotypes caused by MRN deficiency that support a functional link between this complex, ATR and Smc1, including hypersensitivity to UV exposure, a defective UV responsive intra-S phase checkpoint and a specific pattern of genomic instability. In addition, certain ATR-dependent responses do not require MRN. These studies demonstrate that there is indeed a controlling role for MRN over the ATR kinase and have established that the downstream events under this control are broad, including both chromatin-associated and diffuse signaling factors, but may not be universal. These studies contribute to our understanding of the central role that MRN plays in damage detection and signaling, which serve to maintain genomic stability and resist neoplastic transformation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Instabilidade Genômica/genética , Complexos Multiproteicos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Hidrolases Anidrido Ácido , Proteínas Mutadas de Ataxia Telangiectasia , Análise Citogenética , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/genética , Citometria de Fluxo , Raios gama , Instabilidade Genômica/efeitos da radiação , Células HCT116 , Humanos , Immunoblotting , Complexos Multiproteicos/genética , Fenótipo , Fosforilação , Interferência de RNA , Raios Ultravioleta
10.
Mol Cell Biol ; 25(2): 661-70, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15632067

RESUMO

Ataxia-telangiectasia (A-T) mutated (ATM) kinase signals all three cell cycle checkpoints after DNA double-stranded break (DSB) damage. H2AX, NBS1, and p53 are substrates of ATM kinase and are involved in ATM-dependent DNA damage responses. We show here that H2AX is dispensable for the activation of ATM and p53 responses after DNA DSB damage. Therefore, H2AX functions primarily as a downstream mediator of ATM functions in the parallel pathway of p53. NBS1 appears to function both as an activator of ATM and as an adapter to mediate ATM activities after DNA DSB damage. Phosphorylation of ATM and H2AX induced by DNA DSB damage is normal in NBS1 mutant/mutant (NBS1m/m) mice that express an N-terminally truncated NBS1 at lower levels. Therefore, the pleiotropic A-T-related systemic and cellular defects observed in NBS1m/m mice are due to the disruption of the adapter function of NBS1 in mediating ATM activities. While H2AX is required for the irradiation-induced focus formation of NBS1, our findings indicate that NBS1 and H2AX have distinct roles in DNA damage responses. ATM-dependent phosphorylation of p53 and p53 responses are largely normal in NBS1m/m mice after DNA DSB damage, and p53 deficiency greatly facilitates tumorigenesis in NBS1m/m mice. Therefore, NBS1, H2AX, and p53 play synergistic roles in ATM-dependent DNA damage responses and tumor suppression.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiologia , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Células Cultivadas , Quinase do Ponto de Checagem 2 , Proteínas de Ligação a DNA/genética , Fibroblastos/citologia , Fibroblastos/fisiologia , Fibroblastos/efeitos da radiação , Genes Supressores de Tumor , Histonas/genética , Humanos , Cariotipagem , Linfoma/genética , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Radiação Ionizante , Serina/metabolismo , Transcrição Gênica , Translocação Genética , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética
11.
Genes Dev ; 18(11): 1283-92, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15175260

RESUMO

Nonhomologous end joining (NHEJ) and homologous recombination (HR) represent the two major pathways of DNA double-strand break (DSB) repair in eukaryotic cells. NHEJ repairs DSBs by ligation of cognate broken ends irrespective of homologous flanking sequences, whereas HR repairs DSBs using an undamaged homologous template. Although both NHEJ and HR have been clearly implicated in the maintenance of genome stability, how these apparently independent and mechanistically distinct pathways are coordinated remains largely unexplored. To investigate the relationship between HR and NHEJ modes of DSB repair, we generated cells doubly deficient for the NHEJ factor DNA Ligase IV (Lig4) and the HR factor Rad54. We show that Lig4 and Rad54 cooperate to support cellular proliferation, repair spontaneous DSBs, and prevent chromosome and single chromatid aberrations. These findings demonstrate a role for NHEJ in the repair of DSBs that occur spontaneously during or after DNA replication, and reveal overlapping functions for NHEJ and Rad54-dependent HR in the repair of such DSBs.


Assuntos
Cromátides/genética , DNA Ligases/metabolismo , Proteínas Nucleares/metabolismo , Animais , Encéfalo/patologia , Divisão Celular/genética , Células Cultivadas , Aberrações Cromossômicas , DNA/genética , DNA/metabolismo , DNA Helicases , DNA Ligase Dependente de ATP , DNA Ligases/genética , Reparo do DNA/fisiologia , Fertilidade/genética , Fibroblastos , Mamíferos/genética , Camundongos , Camundongos Mutantes , Neurônios/patologia , Proteínas Nucleares/genética , Recombinação Genética , Troca de Cromátide Irmã
12.
Proc Natl Acad Sci U S A ; 101(8): 2410-5, 2004 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-14983023

RESUMO

The nonhomologous DNA end-joining (NHEJ) pathway contains six known components, including Artemis, a nuclease mutated in a subset of human severe combined immunodeficient patients. Mice doubly deficient for the five previously analyzed NHEJ factors and p53 inevitably develop progenitor B lymphomas harboring der(12)t(12;15) translocations and immunoglobin heavy chain (IgH)/c-myc coamplification mediated by a breakage-fusion-bridge mechanism. In this report, we show that Artemis/p53-deficient mice also succumb reproducibly to progenitor B cell tumors, demonstrating that Artemis is a tumor suppressor in mice. However, the majority of Artemis/p53-deficient tumors lacked der(12)t(12;15) translocations and c-myc amplification and instead coamplified IgH and N-myc through an intra- or interchromosome 12 breakage-fusion-bridge mechanism. We discuss this finding in the context of potential implications for mechanisms that may target IgH locus translocations to particular oncogenes.


Assuntos
Linfócitos B/fisiologia , Genes p53 , Neoplasias Experimentais/genética , Proteínas Nucleares/genética , Supressão Genética/genética , Proteína Supressora de Tumor p53/genética , Animais , Linfócitos B/citologia , Sequência de Bases , Mapeamento Cromossômico , Reparo do DNA , Endonucleases , Genes myc , Cadeias Pesadas de Imunoglobulinas/genética , Leucemia de Células B/genética , Linfoma de Células B/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Proteínas Nucleares/deficiência , Taxa de Sobrevida , Translocação Genética , Proteína Supressora de Tumor p53/deficiência
13.
Cell ; 114(3): 359-70, 2003 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-12914700

RESUMO

We employed gene targeting to study H2AX, a histone variant phosphorylated in chromatin surrounding DNA double-strand breaks. Mice deficient for both H2AX and p53 (H(delta/delta)P(-/-)) rapidly developed immature T and B lymphomas and solid tumors. Moreover, H2AX haploinsufficiency caused genomic instability in normal cells and, on a p53-deficient background, early onset of various tumors including more mature B lymphomas. Most H2AX(delta/delta)p53(-/-) or H2AX(+/delta)p53(-/-) B lineage lymphomas harbored chromosome 12 (IgH)/15 (c-myc) translocations with hallmarks of either aberrant V(D)J or class switch recombination. In contrast, H2AX(delta/delta)p53(-/-) thymic lymphomas had clonal translocations that did not involve antigen receptor loci and which likely occurred during cellular expansion. Thus, H2AX helps prevent aberrant repair of both programmed and general DNA breakage and, thereby, functions as a dosage-dependent suppressor of genomic instability and tumors in mice. Notably, H2AX maps to a cytogenetic region frequently altered in human cancers, possibly implicating similar functions in man.


Assuntos
Histonas/metabolismo , Neoplasias/genética , Oncogenes , Translocação Genética , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/metabolismo , Animais , Sequência de Bases , Cromatina , Dano ao DNA , Reparo do DNA , Marcação de Genes , Histonas/genética , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/metabolismo , Alinhamento de Sequência , Taxa de Sobrevida , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética
14.
J Exp Med ; 197(5): 553-65, 2003 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-12615897

RESUMO

In developing lymphocytes, the recombination activating gene endonuclease cleaves DNA between V, D, or J coding and recombination signal (RS) sequences to form hairpin coding and blunt RS ends, which are fused to form coding and RS joins. Nonhomologous end joining (NHEJ) factors repair DNA double strand breaks including those induced during VDJ recombination. Human radiosensitive severe combined immunodeficiency results from lack of Artemis function, an NHEJ factor with in vitro endonuclease/exonuclease activities. We inactivated Artemis in murine embryonic stem (ES) cells by targeted mutation. Artemis deficiency results in impaired VDJ coding, but not RS, end joining. In addition, Artemis-deficient ES cells are sensitive to a radiomimetic drug, but less sensitive to ionizing radiation. VDJ coding joins from Artemis-deficient ES cells, which surprisingly are distinct from the highly deleted joins consistently obtained from DNA-dependent protein kinase catalytic subunit-deficient ES cells, frequently lack deletions and often display large junctional palindromes, consistent with a hairpin coding end opening defect. Strikingly, Artemis-deficient ES cells have increased chromosomal instability including telomeric fusions. Thus, Artemis appears to be required for a subset of NHEJ reactions that require end processing. Moreover, Artemis functions as a genomic caretaker, most notably in prevention of translocations and telomeric fusions. As Artemis deficiency is compatible with human life, Artemis may also suppress genomic instability in humans.


Assuntos
Reparo do DNA , Proteínas Nucleares/metabolismo , Recombinação Genética , Células-Tronco/fisiologia , Animais , Antineoplásicos/farmacologia , Bleomicina/farmacologia , Linhagem Celular , Aberrações Cromossômicas , DNA/efeitos dos fármacos , DNA/efeitos da radiação , Dano ao DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Endonucleases , Marcação de Genes , Genoma , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Hibridização in Situ Fluorescente , Camundongos , Mutação , Proteínas Nucleares/genética , Radiação Ionizante , Análise de Sequência de DNA , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/metabolismo , Telômero/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...