Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Bone Marrow Transplant ; 48(3): 376-82, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23064038

RESUMO

Some clinical characteristics of cord blood transplantation (CBT) might be explained by specificities in the reconstitution of immune subsets differing by their maturation stage or their implication in GVHD, tolerance or immune responses against tumor or infectious agents. Here, we compare the immune reconstitution of several of these subsets after CBT and BMT. B-cell count recovery was faster after CBT. There was no difference in the recovery of CD4(+) and CD8(+) cell counts. There was no difference either in the frequency of several subsets: CD45RO(+) memory, and CD45RA(+) naïve cells within the CD4(+) T-cell compartment, CD27(+) among B cells, CD56(bright), NKG2A(+), and KIR(+) cells among natural killer (NK) cells, CD25(+)FOXP3(+) regulatory T cells and invariant NKT cells. The proportion of the thymic naïve CD31(+)CD45RA(+)CD4(+) T cells was lower after CBT at 6 months post-transplant, and was still below normal at 1 year in both groups. NK-cell expansion was more sustained after CBT, with fewer double-negative NKG2A(-)KIR(-) hyporesponsive cells and more double-positive NKG2A(+)KIR(+) hyper-responsive NK cells. These results, therefore, indicate that further research to improve CBT outcome should try to improve thymopoieisis and take advantage of the sustained NK-cell reconstitution.


Assuntos
Transplante de Medula Óssea/métodos , Sangue Fetal/transplante , Leucemia/cirurgia , Subpopulações de Linfócitos/citologia , Linfócitos/citologia , Linfócitos T Reguladores/citologia , Adolescente , Criança , Estudos de Coortes , Feminino , Sangue Fetal/imunologia , Humanos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Leucemia/sangue , Leucemia/patologia , Linfócitos/imunologia , Masculino , Linfócitos T Reguladores/imunologia
2.
Br J Haematol ; 130(3): 404-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16042690

RESUMO

Familial haemophagocytic lymphohistiocytosis (FHLH) is a genetic disorder caused by defective lymphocyte cytotoxicity, resulting in impaired lymphocyte homeostasis and macrophage infiltration of solid tissues and bone marrow, with extensive haemophagocytosis. It is invariably fatal unless treated by allogeneic haematopoietic stem cell transplantation (HSCT). In a retrospective analysis of 11 cases of FHLH, transplanted in one centre between January 1999 and December 2003, it was found that host T cell expansion occurred early after HSCT in a setting of a viral infection (cytomegalovirus and Epstein-Barr virus respectively) in two cases who received T cell-depleted HSCT. Transient recurrence of clinical and biological manifestations of FHLH was observed, despite evidence for donor cell engraftment. Secondary development of donor T cells led to stable mixed chimaerism and sustained remission of FHLH. Detection of host-derived T cells soon after HSCT in a patient with FHLH should thus not mistakenly be taken as a manifestation of graft rejection.


Assuntos
Antígenos CD34 , Transplante de Células-Tronco Hematopoéticas , Histiocitose de Células não Langerhans/imunologia , Proliferação de Células , Humanos , Lactente , Masculino , Período Pós-Operatório , Recidiva , Estudos Retrospectivos , Linfócitos T/patologia , Quimeras de Transplante , Transplante Homólogo
4.
Bone Marrow Transplant ; 33(11): 1089-95, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15077132

RESUMO

SUMMARY: Hematological inherited diseases can be cured by hematopoietic stem cell transplantation (HSCT) from an human leukocyte antigen (HLA)-identical sibling donor (MSD), but the outcome of unrelated donors (URD) or haploidentical donors (HMD) has been a cause of concern. In all, 94 children affected with inherited diseases underwent HSCT at a single center using MSD (group A, n=31), URD (group B, n=23) or HMD (group C, n=40). There was no difference in the rate of engraftment or in the incidence of grades III-IV acute graft-versus-host disease (GVHD) between the groups. Survival rate was 80.6% in group A, 62.5% in group B and 47.5% in group C (P=0.023). In group B, survival rate was 73.7% in the subgroup with zero or one class I mismatch, and 25% in the subgroup with two or more class I mismatches (P=0.04). In group C, survival rate was 83.3% in the 9/10-identical subgroup, 64.3% in the seven or 8/10 subgroup, and 25% in the five or 6/10 subgroup (P=0.0007). Thus, engraftment, incidence of GVHD and survival are similar in recipients of grafts from MSD, URD with 0-1 class I-mismatch, or HMD with at least 7/10 HLA matches. The low success of HSCT using more disparate donors suggests reserving them for patients with very poor prognosis.


Assuntos
Doenças Genéticas Inatas/terapia , Transplante de Células-Tronco Hematopoéticas , Teste de Histocompatibilidade , Histocompatibilidade/genética , Adolescente , Criança , Pré-Escolar , Doenças Genéticas Inatas/complicações , Doenças Genéticas Inatas/mortalidade , Genótipo , Sobrevivência de Enxerto/imunologia , Doença Enxerto-Hospedeiro/imunologia , Haplótipos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/mortalidade , Transplante de Células-Tronco Hematopoéticas/estatística & dados numéricos , Humanos , Lactente , Infecções Oportunistas/imunologia , Análise de Sobrevida , Doadores de Tecidos , Transplante Homólogo , Transplante Isogênico , Resultado do Tratamento
5.
Science ; 302(5644): 415-9, 2003 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-14564000

RESUMO

We have previously shown correction of X-linked severe combined immunodeficiency [SCID-X1, also known as gamma chain (gamma(c)) deficiency] in 9 out of 10 patients by retrovirus-mediated gamma(c) gene transfer into autologous CD34 bone marrow cells. However, almost 3 years after gene therapy, uncontrolled exponential clonal proliferation of mature T cells (with gammadelta+ or alphabeta+ T cell receptors) has occurred in the two youngest patients. Both patients' clones showed retrovirus vector integration in proximity to the LMO2 proto-oncogene promoter, leading to aberrant transcription and expression of LMO2. Thus, retrovirus vector insertion can trigger deregulated premalignant cell proliferation with unexpected frequency, most likely driven by retrovirus enhancer activity on the LMO2 gene promoter.


Assuntos
Proteínas de Ligação a DNA/genética , Terapia Genética/efeitos adversos , Vetores Genéticos , Leucemia-Linfoma de Células T do Adulto/etiologia , Metaloproteínas/genética , Retroviridae/genética , Imunodeficiência Combinada Severa/terapia , Linfócitos T/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Ensaios Clínicos como Assunto , Células Clonais/fisiologia , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Humanos , Lactente , Proteínas com Domínio LIM , Mutagênese Insercional , Regiões Promotoras Genéticas , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas , Proto-Oncogenes , Receptores de Interleucina-2/genética , Retroviridae/fisiologia , Transcrição Gênica , Integração Viral , Replicação Viral
7.
Cell Death Differ ; 10(1): 124-33, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12655301

RESUMO

Human and mouse natural mutants presenting with lymphoproliferative syndrome and autoimmunity (ALPS) have enlightened the role of the Fas and FasL in lymphocyte cell death and peripheral tolerance. Further study of the genetic basis of the human pathology led to the identification of apoptosis signaling defect, and pointed out to the crucial role of caspase-10 in the process of apoptosis induction. In contrast, the absence of lymproliferation in engineered mutants of 'death pathways' suggests that additional events are necessary to recapitulate the overt phenotype of ALPS patients or MRL/lpr mice. Moreover, these models highlight the roles of Fas and associated molecules, such as FADD and caspase-8, in lymphocyte development or activation. This review will discuss the main findings provided by the study of mouse models and human conditions.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose/genética , Doenças Autoimunes/genética , Linfócitos/imunologia , Transtornos Linfoproliferativos/genética , Animais , Apoptose/imunologia , Doenças Autoimunes/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Caspases/genética , Caspases/imunologia , Modelos Animais de Doenças , Proteína de Domínio de Morte Associada a Fas , Humanos , Linfócitos/metabolismo , Transtornos Linfoproliferativos/imunologia , Camundongos , Receptor fas/genética , Receptor fas/imunologia
9.
Genes Immun ; 3 Suppl 1: S66-70, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12215906

RESUMO

Mutations in the Fas (apo-1, CD95) gene result in autoimmune lymphoproliferative syndrome (ALPS). These mutations are dominated by small deletions and point mutations that result in splicing errors or missense changes. We report here a novel mutation caused by retrotransposon insertion, which results in loss of exon 8 and ALPS. A father and son suffering from recurrent lymphadenopathy were examined for resistance to Fas-mediated apoptosis. A functional defect was detected and RT-PCR analysis revealed two different copies of Fas mRNA, one normal and a second shorter version lacking exon 8. DNA analysis of the genomic region between exons seven and nine in the longer copy revealed two PCR products, one being 331 base pairs (bp) longer than expected. Sequencing revealed that intron 7 had undergone an insertion event with an Alu element (99.31% homology with Alu-Sb1) of 331 bp. This element included a 34-bp Poly A tract that was flanked on each side by a perfect 17 bp direct duplication of the target site. Both patients were heterozygous for the mutated allele that produced Fas mRNA lacking exon 8, although not due to loss of a splice junction. The structure of the insertion suggests that the Alu element may have integrated by retrotransposition, and represents the first report of a retrotransposon causing ALPS.


Assuntos
Elementos Alu/genética , Elementos Alu/fisiologia , Autoimunidade/genética , Inativação Gênica , Transtornos Linfoproliferativos/genética , Receptor fas/genética , Receptor fas/fisiologia , Apoptose/genética , Apoptose/imunologia , Autoimunidade/imunologia , Sequência de Bases , Humanos , Lactente , Íntrons , Transtornos Linfoproliferativos/imunologia , Masculino , Dados de Sequência Molecular , RNA Mensageiro , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Lancet ; 358(9292): 1511-3, 2001 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-11705566

RESUMO

Autoimmune haemolytic anaemia commonly has a severe course in young children, thus requiring multiple immunosuppressive treatments. Five children with refractory idiopathic autoimmune haemolytic anaemia, and one child with the disease after bone-marrow transplantation, were treated with rituximab-a monoclonal antibody against CD20. Tolerance of the treatment was good. However, circulating Bcells were absent and hypogammaglobulinaemia was seen for 9 months after treatment. All patients remained in complete remission 15-22 months after the start of rituximab therapy. Corticosteroids and immunosuppressive drugs were stopped or their dose markedly reduced. We suggest that rituximab could be a valuable treatment for autoimmune haemolytic anaemia, although a long-lasting but transient B-cell deficiency develops.


Assuntos
Anemia Hemolítica Autoimune/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Anticorpos Monoclonais Murinos , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Prednisona/uso terapêutico , Rituximab , Resultado do Tratamento
11.
J Soc Biol ; 195(1): 65-8, 2001.
Artigo em Francês | MEDLINE | ID: mdl-11530503

RESUMO

The success of HSCT from HLA partially disparate donors depends on the development of new strategies able to efficiently prevent GVHD and to protect patients from infections and relapse. Using an immunotoxin (IT) directed against the alpha-chain (p55) of the human IL-2r (RFT5-SMPT-dgA), we have previously shown that it is possible to kill mature T cells activated towards a specific HLA complex by a one-way MLR. We designed a clinical trial assessing the effect of infusing increasing doses of T lymphocytes in the setting of children recipients of non HLA genetically identical HSCT. Thirteen patients have been enrolled from September 1998 to April 2000 and fourteen HSCT have been realized in 13 patients (pts). Donors were MUD in 3 cases and familial HLA partially disparate in the remaining cases. Allodepleted donor T cells were injected between day +14 and day +30 provided that ATG was undetectable in the serum and blood PMN counts was > 500/microliter. The mean age of these patients was 17 months (range 1 to 42). Diagnosis included immune deficient and malignant hemopathies. Three patients received 1 x 10(5) allodepleted T cell/kg, 7 patients received 4 x 10(5)/kg and 4 patients received 6 x 10(5)/kg allodepleted T cells. Full inhibition of MLR was achieved in 12 out of 14 cases. In two cases, a residual T cell reactivity to the recipient was observed (4 to 5%) and patients developed grade II aGVHD. aGVHD occurred in 4 out of 11 grafted patients (all grade II). No chronic GVHD has developed, so far. Three patients died from severe VOD or PHT at day +34, day 51 and day +166, while one infected patient by VZV, CMV and EBV before HSCT died 6 months after transplantation from meningoencephalitis and another patient died from relapse at day +291. The patient for which there was no engraftment died at day +48 from staphylococcus infection. Overall survival is 54%, with a median follow up of 8 months; the mean time to reach a blood lymphocyte count > 500 was 41 days, to reach a CD3 count > 300 microliters 63 days (20-111), CD4 > 200 microliters 97 days and positive mitogen-induced proliferation 90 days. In three patients, a tetanus-toxoid positive proliferation was detected before immunization. From this intermediate analysis, we conclude that 1) specific allodepletion is an effective approach to prevent aGVHD in a haploincompatible setting, 2) data on immunological reconstitution suggest that infused T cells do survive and expand. A higher number of patients must be enrolled to determine the optimal number of T cells to infuse.


Assuntos
Anticorpos Monoclonais/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Transplante de Células-Tronco Hematopoéticas/métodos , Imunotoxinas/farmacologia , Depleção Linfocítica/métodos , Receptores de Interleucina-2/imunologia , Subpopulações de Linfócitos T/transplante , Doença Aguda , Criança , Pré-Escolar , Doença Enxerto-Hospedeiro/epidemiologia , Neoplasias Hematológicas/terapia , Histocompatibilidade , Humanos , Síndromes de Imunodeficiência/terapia , Imunotoxinas/imunologia , Lactente , Recém-Nascido , Infecções/etiologia , Infecções/mortalidade , Contagem de Linfócitos , Teste de Cultura Mista de Linfócitos , Subpopulações de Linfócitos T/imunologia , Transplante Homólogo , Resultado do Tratamento
13.
J Biol Chem ; 276(40): 36902-8, 2001 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-11463783

RESUMO

The protein tyrosine kinase ZAP-70 plays a pivotal role involved in signal transduction through the T cell receptor and CD2. Defects in ZAP-70 result in severe combined immunodeficiency. We report that Herpesvirus saimiri, which does not code for a ZAP-70 homologue, can replace this tyrosine kinase. H. saimiri is an oncogenic virus that transforms human T cells to stable growth based on mutual CD2-mediated activation. Although CD2-mediated proliferation of ZAP-70-deficient uninfected T cells was absent, we could establish H. saimiri-transformed T cell lines from two unrelated patients presenting with ZAP-70 deficiencies. In these cell lines, CD2 and CD3 activation were restored in terms of [Ca(2+)](i), MAPK activation, cytokine production, and proliferation. Activation-induced tyrosine phosphorylation of zeta remained defective. The transformed cells expressed very high levels of the ZAP-70-related kinase Syk. This increased expression was not observed in the primary T cells from the patients and was not due to the transformation by the virus because transformed cell lines established from control T cells did not present this particularity. In conclusion, wild type H. saimiri can restore CD2- and CD3-mediated activation in signaling-deficient human T cells. It extends our understanding of interactions between the oncogenic H. saimiri and the infected host cells.


Assuntos
Transformação Celular Viral/fisiologia , Herpesvirus Saimiriíneo 2/fisiologia , Ativação Linfocitária/fisiologia , Proteínas Tirosina Quinases/metabolismo , Linfócitos T/fisiologia , Comunicação Autócrina , Antígenos CD2/fisiologia , Complexo CD3/fisiologia , Divisão Celular , Células Cultivadas , Precursores Enzimáticos/metabolismo , Citometria de Fluxo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Tirosina Quinases/deficiência , Receptores de Antígenos de Linfócitos T/metabolismo , Quinase Syk , Linfócitos T/citologia , Linfócitos T/virologia , Proteína-Tirosina Quinase ZAP-70
14.
J Gene Med ; 3(3): 201-6, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11437325

RESUMO

Recent advances in gene transfer in human hematopoietic cells, combined with a better understanding of the genetic aspects of several immunodeficiencies, has offered new opportunities in the domain of gene therapy. Severe combined immunodeficiency (SCID) appear to represent a good model for the application of gene therapy, combining an expected selective advantage for transduced cells, an absence of immunological response to the vector and/or the therapeutic transgene, together with accessibility to hematopoietic stem cells (HSC). Ex vivo retroviral transduction of a therapeutic transgene in HSC prior to transplantation appears to be a particularly effective and long-lasting means of restoring the expression of a mutated gene in the lymphoid lineage. Furthermore, encouraging therapeutic benefits as a result of a gene therapy protocol for the treatment of X-linked severe combined immunodeficiencies (SCID-X1) invites many questions as to the reasons for this therapeutic benefit. This review outlines the results that have been achieved in gene therapy for SCID-X1, ADA-SCID as well as other types of SCID, and discusses the possible relationship between the physiopathology of each disease and the success of relevant trials.


Assuntos
Terapia Genética , Imunodeficiência Combinada Severa/terapia , Adenosina Desaminase/deficiência , Ligação Genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Cromossomo X/genética
16.
Br J Haematol ; 113(2): 432-4, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11380411
17.
Nat Genet ; 27(3): 277-85, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11242109

RESUMO

The molecular basis of X-linked recessive anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID) has remained elusive. Here we report hypomorphic mutations in the gene IKBKG in 12 males with EDA-ID from 8 kindreds, and 2 patients with a related and hitherto unrecognized syndrome of EDA-ID with osteopetrosis and lymphoedema (OL-EDA-ID). Mutations in the coding region of IKBKG are associated with EDA-ID, and stop codon mutations, with OL-EDA-ID. IKBKG encodes NEMO, the regulatory subunit of the IKK (IkappaB kinase) complex, which is essential for NF-kappaB signaling. Germline loss-of-function mutations in IKBKG are lethal in male fetuses. We show that IKBKG mutations causing OL-EDA-ID and EDA-ID impair but do not abolish NF-kappaB signaling. We also show that the ectodysplasin receptor, DL, triggers NF-kappaB through the NEMO protein, indicating that EDA results from impaired NF-kappaB signaling. Finally, we show that abnormal immunity in OL-EDA-ID patients results from impaired cell responses to lipopolysaccharide, interleukin (IL)-1beta, IL-18, TNFalpha and CD154. We thus report for the first time that impaired but not abolished NF-kappaB signaling in humans results in two related syndromes that associate specific developmental and immunological defects.


Assuntos
Displasia Ectodérmica/genética , Displasia Ectodérmica/imunologia , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/genética , Adolescente , Criança , Pré-Escolar , Códon de Terminação/genética , Displasia Ectodérmica/metabolismo , Ectodisplasinas , Ligação Genética , Humanos , Quinase I-kappa B , Imunidade Celular , Síndromes de Imunodeficiência/metabolismo , Lactente , Masculino , Proteínas de Membrana/metabolismo , Mutação , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Síndrome , Cromossomo X/genética
18.
Proc Natl Acad Sci U S A ; 98(3): 1166-70, 2001 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-11158612

RESUMO

Somatically mutated IgM(+)-only and IgM(+)IgD(+)CD27(+) B lymphocytes comprise approximately 25% of the human peripheral B cell pool. These cells phenotypically resemble class-switched B cells and have therefore been classified as postgerminal center memory B cells. X-linked hyper IgM patients have a genetic defect characterized by a mutation of the CD40L gene. These patients, who do not express a functional CD40 ligand, cannot switch Ig isotypes and do not form germinal centers and memory B cells. We report here that an IgM(+)IgD(+)CD27(+) B cell subset with somatically mutated Ig receptors is generated in these patients, implying that these cells expand and diversify their Ig receptors in the absence of classical cognate T-B collaboration. The presence of this sole subset in the absence of IgM(+)-only and switched CD27(+) memory B cells suggests that it belongs to a separate diversification pathway.


Assuntos
Linfócitos B/imunologia , Antígenos CD40/genética , Ligante de CD40/genética , Genes de Imunoglobulinas , Imunoglobulina M/genética , Síndromes de Imunodeficiência/genética , Mutação , Adolescente , Adulto , Processamento Alternativo , Subpopulações de Linfócitos B/imunologia , Antígenos CD40/imunologia , Ligante de CD40/imunologia , Criança , Pré-Escolar , Códon de Terminação , Sangue Fetal/imunologia , Rearranjo Gênico , Humanos , Imunoglobulina A/sangue , Imunoglobulina D/genética , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Síndromes de Imunodeficiência/sangue , Síndromes de Imunodeficiência/imunologia , Recém-Nascido , Valores de Referência , Deleção de Sequência
20.
J Immunol ; 165(8): 4725-30, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11035117

RESUMO

Common variable immunodeficiency (CVID) is an heterogeneous syndrome characterized by decreased levels of serum Ig and recurrent bacterial infection. Here, we were interested to study whether a qualitative defect of the affinity Ab maturation process could be combined to the low level of serum Ig in a cohort of 38 CVID patients. For this, we designed a novel and rapid screening test for the detection of hypomutated V gene expressed by memory B cells. This test delineated a subset of 9/38 (23%) CVID patients with an abnormal pattern of Ig V gene mutation. The mean frequency of V gene mutation of this subset was significantly lower (1.74%) compared with other CVID patients (5.46%) and normal donors (6.5%) (p<0.0001). The mean age of this subgroup was significantly higher than other hypogammaglobulinemic patients with normal levels of V gene mutation (p<0.02), whereas no difference in the duration of symptoms was noted between the two groups. This suggests that hypomutation characterizes patients who began CVID late in life. Recently, it was shown that non-Ig sequences, such as the intronic BCL-6 gene, could be the target of the somatic hypermutation process in normal memory B cells. Our finding of a normal mutation frequency of the BCL-6 gene in two hypomutated CVID point to a defect of the Ig targeting of hypermutation machinery in these cases.


Assuntos
Afinidade de Anticorpos/genética , Imunodeficiência de Variável Comum/genética , Imunodeficiência de Variável Comum/imunologia , Adulto , Idoso , Alanina/genética , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Células Clonais , Códon , Imunodeficiência de Variável Comum/sangue , Sequência Consenso , Análise Mutacional de DNA , Proteínas de Ligação a DNA/sangue , Proteínas de Ligação a DNA/genética , Genes de Imunoglobulinas , Humanos , Regiões Constantes de Imunoglobulina/genética , Imunoglobulina G/genética , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Memória Imunológica/genética , Pessoa de Meia-Idade , Mutação , Proteínas Proto-Oncogênicas/sangue , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-6 , Serina/genética , Fatores de Transcrição/sangue , Fatores de Transcrição/genética , Transcrição Gênica/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA