Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Pediatr Infect Dis J ; 35(12): 1350-1351, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27626919

RESUMO

Acanthamoeba infections are rare and mostly occur in immunocompromised patients. Most of the reported cases after stem cell transplantation have been diagnosed postmortem. We present the case of a 3-year-old boy with chronic graft versus host disease post hematopoietic transplantation, who was successfully treated for Acanthamoeba.


Assuntos
Amebíase , Doença Enxerto-Hospedeiro/complicações , Transplante de Células-Tronco Hematopoéticas , Sinusite , Acanthamoeba , Amebíase/complicações , Amebíase/tratamento farmacológico , Amebíase/parasitologia , Amebicidas/uso terapêutico , Anfotericina B/uso terapêutico , Pré-Escolar , Humanos , Masculino , Mucosa Nasal/parasitologia , Mucosa Nasal/patologia , Sinusite/complicações , Sinusite/tratamento farmacológico , Sinusite/parasitologia
2.
Biol Blood Marrow Transplant ; 22(6): 1073-1079, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27038860

RESUMO

Although high absolute lymphocyte count (ALC) early after transplantation is a simple surrogate for immune reconstitution, few studies to date have established the predictive factors for ALC after umbilical cord blood transplantation (UCBT). We retrospectively studied the factors associated with early lymphocyte recovery and the impact of the ALC on day +42 (ALC42) of ≥300 × 10(6)/L on outcomes in 210 consecutive pediatric and adult patients (112 males; median age, 15 years; range, 0.3 to 60 years; interquartile range, 4 to 36 years) who underwent myeloablative in vivo T cell-depleted single UCBT between 2005 and 2014 for malignant and nonmalignant disorders. In a logistic multivariate regression model, factors favoring a higher ALC42 were higher infused CD3(+) cell dose (odds ratio [OR], 2.7; 95% CI, 1.4 to 5.2; P = .004), lower antithymocyte globulin dose (OR, 2.3; 95% CI, 1.2 to 4.5; P = .01), and better HLA match (OR, 2.1; 95% CI, 1.1 to 4.1; P = .03). In multivariate analysis, lower ALC42 was associated with higher nonrelapse mortality (hazard ratio [HR], 1.76; 95% CI, 1.34 to 2.32; P = .001), whereas a higher ALC42 was associated with better disease-free survival (HR, 2.03; 95% CI, 1.15 to 3.6; P < .001) and overall survival (HR, 2.03; 95% CI, 1.17 to 3.6; P < .001). Our study suggests that the selection of better HLA-matched cord blood units containing higher CD3(+) cell counts and the use of conditioning regimens with lower ATG doses could improve immune reconstitution after UCBT.


Assuntos
Complexo CD3/sangue , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Sangue Fetal/imunologia , Depleção Linfocítica , Linfócitos T/citologia , Adolescente , Adulto , Soro Antilinfocitário/administração & dosagem , Criança , Pré-Escolar , Histocompatibilidade , Humanos , Lactente , Recém-Nascido , Contagem de Linfócitos , Pessoa de Meia-Idade , Estudos Retrospectivos , Análise de Sobrevida , Condicionamento Pré-Transplante/métodos , Adulto Jovem
3.
Biol Blood Marrow Transplant ; 21(4): 682-7, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25545727

RESUMO

Graft dilution and DMSO washing before cord blood (CB) administration using an automated system may offer low incidence of adverse infusion events (AIE), ensuring reproducible cell yields. Hence, we analyzed the incidences and significance of immediate AIE, cellular yield, and engraftment after single CB infusion. One hundred and fifty-seven patients (median age, 20 years; range, 1 to 60) received a single CB unit for treatment of hematologic and nonhematologic malignancies with myeloablative conditioning after graft dilution and washing. The median total nucleated cell (TNC) doses was 3.4 × 10(7)/kg (range, 2 to 26) and the median post-thaw recovery was 84% (range, 45 to 178). The cumulative incidence of neutrophil engraftment at 50 days was 84% (95% confidence interval [CI], 83 to 93). A total of 118 immediate AIE were observed in fifty-two (33%) patients. All reported AIE were transient, graded from 1 to 2 by Common Terminology Adverse Events version 4. The most frequent toxicity was cardiovascular but without any life-threatening reaction. Infused TNC, recipient's weight, and rate of infusion per kilogram were risk factors associated with cardiovascular AIE in multivariate analysis (odds ratio [OR], 1.2 (95% CI, 1.1 to 1.4); P < .001; OR, .94 (95% CI, .9 to .97); P < .001; and OR, 1.5 (95% CI, 1.2 to 1.8); P < .001; respectively). In summary, use of an automated method for graft washing before CB administration showed low incidence of AIE without compromising cell yields and engraftment. Infused TNC dose, recipient's weight, and rate of infusion per kilogram were risk factors associated with infusion reactions.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Sobrevivência de Enxerto , Células-Tronco Hematopoéticas , Neoplasias/terapia , Condicionamento Pré-Transplante , Adolescente , Adulto , Aloenxertos , Criança , Pré-Escolar , Transplante de Células-Tronco de Sangue do Cordão Umbilical/efeitos adversos , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Neoplasias/mortalidade , Fatores de Risco
4.
Pediatr Dermatol ; 30(4): 484-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23557135

RESUMO

Congenital erythropoietic porphyria (CEP), or Günther's disease, is an inborn error of metabolism produced by a deficiency of uroporphyrinogen III synthase (UROS), the fourth enzyme of the heme biosynthesis pathway. This enzymatic defect induces the accumulation of isomer I porphyrins in erythrocytes, skin, and tissues, producing various clinical manifestations. Severe cases are characterized by extreme photosensitivity, causing scarring and mutilations, and by hemolytic anemia, reducing life expectancy. CEP is caused by mutations in the UROS gene, and one of the most severe forms of the disease is associated with a cysteine to arginine substitution at residue 73 of the protein (C73R). CEP has been successfully treated only by the transplantation of hematopoietic precursors. We report the case of a male infant with severe postdelivery symptoms diagnosed with CEP and found to be homozygous for the C73R mutation. He underwent successful allogeneic bone marrow transplantation from a matched unrelated donor at 7 months of age. The hemolytic anemia was corrected and the porphyrin overproduction was significantly reduced. The patient remained asymptomatic after 1 year. This new case confirms that patients with severe CEP can benefit from early postnatal hematopoietic stem cell transplantation.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Teste de Histocompatibilidade , Porfiria Eritropoética/terapia , Humanos , Lactente , Masculino , Porfiria Eritropoética/genética , Resultado do Tratamento , Uroporfirinogênio III Sintetase/genética
5.
Blood ; 117(14): 3759-69, 2011 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-21273304

RESUMO

Fanconi anemia is characterized by congenital abnormalities, bone marrow failure, and cancer predisposition. To investigate the origin, functional role, and clinical impact of FANCA mutations, we determined a FANCA mutational spectrum with 130 pathogenic alleles. Some of these mutations were further characterized for their distribution in populations, mode of emergence, or functional consequences at cellular and clinical level. The world most frequent FANCA mutation is not the result of a mutational "hot-spot" but results from worldwide dissemination of an ancestral Indo-European mutation. We provide molecular evidence that total absence of FANCA in humans does not reduce embryonic viability, as the observed frequency of mutation carriers in the Gypsy population equals the expected by Hardy-Weinberg equilibrium. We also prove that long distance Alu-Alu recombination can cause Fanconi anemia by originating large interstitial deletions involving FANCA and 2 adjacent genes. Finally, we show that all missense mutations studied lead to an altered FANCA protein that is unable to relocate to the nucleus and activate the FA/BRCA pathway. This may explain the observed lack of correlation between type of FANCA mutation and cellular phenotype or clinical severity in terms of age of onset of hematologic disease or number of malformations.


Assuntos
Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteína do Grupo de Complementação A da Anemia de Fanconi/fisiologia , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Mutação , Adolescente , Idade de Início , Sequência de Bases , Técnicas de Cultura de Células , Células Cultivadas , Criança , Pré-Escolar , Aberrações Cromossômicas , Hibridização Genômica Comparativa , Análise Mutacional de DNA , Anemia de Fanconi/diagnóstico , Anemia de Fanconi/epidemiologia , Proteína do Grupo de Complementação A da Anemia de Fanconi/metabolismo , Frequência do Gene , Humanos , Lactente , Modelos Biológicos , Dados de Sequência Molecular , Mutação/fisiologia , Fenótipo , Espanha/epidemiologia
6.
J Med Genet ; 48(4): 242-50, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21217111

RESUMO

BACKGROUND: Fanconi anaemia (FA) is a rare syndrome characterized by bone marrow failure, malformations and cancer predisposition. Chromosome fragility induced by DNA interstrand crosslink (ICL)-inducing agents such as diepoxybutane (DEB) or mitomycin C (MMC) is the 'gold standard' test for the diagnosis of FA. OBJECTIVE: To study the variability, the diagnostic implications and the clinical impact of chromosome fragility in FA. METHODS: Data are presented from 198 DEB-induced chromosome fragility tests in patients with and without FA where information on genetic subtype, cell sensitivity to MMC and clinical data were available. RESULTS: This large series allowed quantification of the variability and the level of overlap in ICL sensitivity among patients with FA and the normal population. A new chromosome fragility index is proposed that provides a cut-off diagnostic level to unambiguously distinguish patients with FA, including mosaics, from non-FA individuals. Spontaneous chromosome fragility and its correlation with DEB-induced fragility was also analysed, indicating that although both variables are correlated, 54% of patients with FA do not have spontaneous fragility. The data reveal a correlation between malformations and sensitivity to ICL-inducing agents. This correlation was also statistically significant when the analysis was restricted to patients from the FA-A complementation group. Finally, chromosome fragility does not correlate with the age of onset of haematological disease. CONCLUSIONS: This study proposes a new chromosome fragility index and suggests that genome instability during embryo development may be related to malformations in FA, while DEB-induced chromosome breaks in T cells have no prognostic value for the haematological disease.


Assuntos
Fragilidade Cromossômica , Anemia de Fanconi/genética , Reagentes de Ligações Cruzadas/farmacologia , Compostos de Epóxi/farmacologia , Anemia de Fanconi/diagnóstico , Humanos , Mitomicina/farmacologia , Mosaicismo , Fenótipo
7.
Mol Ther ; 17(6): 1083-92, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19277017

RESUMO

Previous clinical trials based on the genetic correction of purified CD34(+) cells with gamma-retroviral vectors have demonstrated clinical efficacy in different monogenic diseases, including X-linked severe combined immunodeficiency, adenosine deaminase deficient severe combined immunodeficiency and chronic granulomatous disease. Similar protocols, however, failed to engraft Fanconi anemia (FA) patients with genetically corrected cells. In this study, we first aimed to correlate the hematological status of 27 FA patients with CD34(+) cell values determined in their bone marrow (BM). Strikingly, no correlation between these parameters was observed, although good correlations were obtained when numbers of colony-forming cells (CFCs) were considered. Based on these results, and because purified FA CD34(+) cells might have suboptimal repopulating properties, we investigated the possibility of genetically correcting unselected BM samples from FA patients. Our data show that the lentiviral transduction of unselected FA BM cells mediates an efficient phenotypic correction of hematopoietic progenitor cells and also of CD34(-) mesenchymal stromal cells (MSCs), with a reported role in hematopoietic engraftment. Our results suggest that gene therapy protocols appropriate for the treatment of different monogenic diseases may not be adequate for stem cell diseases like FA. We propose a new approach for the gene therapy of FA based on the rapid transduction of unselected hematopoietic grafts with lentiviral vectors (LVs).


Assuntos
Anemia de Fanconi/metabolismo , Anemia de Fanconi/terapia , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Lentivirus/genética , Células-Tronco Mesenquimais/metabolismo , Antígenos CD34/metabolismo , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Linhagem Celular , Células Cultivadas , Anemia de Fanconi/patologia , Humanos
9.
J Med Genet ; 44(4): 241-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17105750

RESUMO

BACKGROUND: Fanconi anaemia is a heterogeneous genetic disease, where 12 complementation groups have been already described. Identifying the complementation group in patients with Fanconi anaemia constitutes a direct procedure to confirm the diagnosis of the disease and is required for the recruitment of these patients in gene therapy trials. OBJECTIVE: To determine the subtype of Fanconi anaemia patients in Spain, a Mediterranean country with a relatively high population (23%) of Fanconi anaemia patients belonging to the gypsy race. METHODS: Most patients could be subtyped by retroviral complementation approaches in peripheral blood T cells, although some mosaic patients were subtyped in cultured skin fibroblasts. Other approaches, mainly based on western blot analysis and generation of nuclear RAD51 and FANCJ foci, were required for the subtyping of a minor number of patients. RESULTS AND CONCLUSIONS: From a total of 125 patients included in the Registry of Fanconi Anaemia, samples from 102 patients were available for subtyping analyses. In 89 cases the subtype could be determined and in 8 cases exclusions of common complementation groups were made. Compared with other international studies, a skewed distribution of complementation groups was observed in Spain, where 80% of the families belonged to the Fanconi anaemia group A (FA-A) complementation group. The high proportion of gypsy patients, all of them FA-A, and the absence of patients with FA-C account for this characteristic distribution of complementation groups.


Assuntos
Algoritmos , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Anemia de Fanconi/classificação , Heterogeneidade Genética , Roma (Grupo Étnico)/genética , Células Cultivadas/química , Células Cultivadas/efeitos dos fármacos , Consanguinidade , Resistência a Medicamentos/genética , Compostos de Epóxi/farmacologia , Anemia de Fanconi/epidemiologia , Anemia de Fanconi/genética , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/análise , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Fibroblastos/química , Fibroblastos/patologia , Teste de Complementação Genética , Genótipo , Humanos , Incidência , Mitomicina/farmacologia , Mosaicismo , Sistema de Registros , Retroviridae/genética , Espanha/epidemiologia , Linfócitos T/química , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Transdução Genética
11.
Haematologica ; 88(3): 290-9, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12651268

RESUMO

BACKGROUND AND OBJECTIVES: Although chemotherapy in childhood acute myeloid leukemia (AML) has improved in the last decade, except for a group of better-risk patients (approximately one third), more than half the other patients relapse. The main objective of this study was to evaluate the results obtained with bone marrow transplants, either allogeneic (allo-BMT) or autologous (auto-BMT), following two intensive consolidation courses in a series of children with high-risk (HR) AML according to morphologic and early-response BFM criteria. A second objective was to compare the results of auto-BMT with those of allo-BMT. DESIGN AND METHODS: From April 1988 to May 2001, 79 children (< 15 years old) with de novo AML entered the prospective AML-88 trial in a single institution: 50 (63%) were qualified as having high-risk disease and are the subject of this study. After 1 or 2 induction courses, depending on early response, and two consolidations, patients with an HLA-identical sibling received an allo-BMT and all the others an auto-BMT. The conditioning regimen was cyclophosphamide and total body irradiation (TBI) in children over 3 years old and busulfan and etoposide in younger children. Bone marrow was purged with mafosfamide in auto-BMT and cyclosporine alone was given as graft-versus-host disease (GVHD) prophylaxis in allo-BMT. RESULTS: At the end of the chemotherapy phase (induction and consolidation ), 46 of the 50 HR patients (92%) had attained complete remission (CR) after one (n=29), two (n=11) or three (n=6) courses; 2 more were in partial remission (PR) and 2 had died. The 48 patients in CR or PR received either an allo-BMT (17) or an auto-BMT (31). Hematologic reconstitution was significantly slower in auto-BMT recipients. Forty-one percent of patients who received allo-BMT suffered acute GVHD grades II-IV. Toxic deaths and relapse rates were 5.9% and 17.6%, respectively, in allo-BMT and 3.2% and 25.8%, respectively, in auto-BMT. Post-transplant 8-year event-free survival (EFS) was 74.5% (54-96) in allo-BMT and 74.2% (59-89) in auto-BMT. EFS and OS in all the series (50 patients) were 71% (59-83) and 73% (61-85), respectively, with a median follow-up of 7.2 years. INTERPRETATION AND CONCLUSIONS: This study indicates that improved results in children with HR-AML can be obtained by either allo- or auto-BMT performed after two courses of intensive consolidation therapy provided good supportive therapy is given and reduced transplant -related mortality (TRM) is minimized.


Assuntos
Antineoplásicos/uso terapêutico , Transplante de Medula Óssea/mortalidade , Leucemia Mieloide/terapia , Doença Aguda , Adolescente , Antineoplásicos/toxicidade , Transplante de Medula Óssea/efeitos adversos , Transplante de Medula Óssea/métodos , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Leucemia Mieloide/complicações , Leucemia Mieloide/mortalidade , Masculino , Qualidade de Vida , Risco , Transplante Autólogo/mortalidade , Transplante Homólogo/mortalidade , Resultado do Tratamento
12.
J Pediatr Hematol Oncol ; 24(9): 772-6, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12468924

RESUMO

We report a boy with Wiskott-Aldrich syndrome (WAS) who developed autoimmune thyroiditis 19 months after allogenic bone marrow transplantation (BMT). Possible causes of his autoimmune illness were 1) transference of autoimmune cells from the donor, which was ruled out because of the absence of autoimmune illness in his healthy HLA-identical brother (donor); 2) persistent mixed chimerism after BMT ruled out by post-BMT molecular analysis of the proband's peripheral lymphocytes; and 3) patient's predisposition to autoimmune disease secondary to an dysregulated immune system because of WAS and his HLA haplotype. This case brings previously unreported findings to the spectrum of WAS.


Assuntos
Transplante de Medula Óssea/efeitos adversos , Proteínas/genética , Tireoidite Autoimune/etiologia , Síndrome de Wiskott-Aldrich/terapia , Sequência de Aminoácidos , Substituição de Aminoácidos , Sequência de Bases , Pré-Escolar , Cromossomos Humanos X , Éxons , Humanos , Masculino , Mutação , Tireoidite Autoimune/genética , Transplante Homólogo , Síndrome de Wiskott-Aldrich/genética , Proteína da Síndrome de Wiskott-Aldrich
13.
Radiographics ; 22(6): 1411-28, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12432112

RESUMO

Recent advances in therapy for pediatric hematologic neoplasms have greatly improved the prognosis but have resulted in an increased incidence of associated complications and toxic effects. The main neuroimaging features in pediatric patients with leukemia or lymphoma treated with chemotherapy or radiation therapy were retrospectively reviewed. To simplify the approach and facilitate differential diagnosis, the neuroimaging features have been classified into three main categories: central nervous system manifestations of primary disease, side effects of therapeutic procedures (radiation therapy, chemotherapy, bone marrow transplantation), and complications due to immunosuppression, particularly infections. Manifestations of primary disease include cerebrovascular complications (hemorrhage, cerebral infarction) and central nervous system involvement (infiltration of the meninges, parenchyma, bone marrow, orbit, and spine). Effects of radiation therapy include white matter disease, mineralizing microangiopathy, parenchymal brain volume loss, radiation-induced cryptic vascular malformations, and second neoplasms. Effects of chemotherapy and bone marrow transplantation include hemorrhage, dural venous thrombosis, white matter disease, reversible posterior leukoencephalopathy syndrome, and anterior lumbosacral radiculopathy. Both the underlying malignancy and antineoplastic therapy can cause immunosuppression. Fungi are the most frequent causal microorganisms in immunosuppressed patients with infection. Familiarity with the imaging findings is essential for proper diagnosis of neurologic symptoms in pediatric patients with oncohematologic disease.


Assuntos
Doenças do Sistema Nervoso Central/diagnóstico , Leucemia/diagnóstico , Linfoma/diagnóstico , Adolescente , Doenças do Sistema Nervoso Central/complicações , Doenças do Sistema Nervoso Central/terapia , Criança , Pré-Escolar , Diagnóstico Diferencial , Humanos , Lactente , Leucemia/complicações , Leucemia/terapia , Linfoma/complicações , Linfoma/terapia , Imageamento por Ressonância Magnética , Tomografia Computadorizada por Raios X
14.
Mutat Res ; 504(1-2): 75-83, 2002 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-12106648

RESUMO

Fanconi anemia (FA) is a chromosome instability syndrome, characterized by progressive pancytopenia and cancer susceptibility. Other cellular features of FA cells are hypersensitivity to DNA cross-linking agents and accelerated telomere shortening. We have quantified overall genome chromosome fragility and euploidy as well as chromosomes 7 and 8 aneuploidy in peripheral blood lymphocytes from a group of FA patients and age-matched controls that were previously measured for telomere length. The haematology of FA samples were also characterized in terms of whole blood cell, neuthrophil and platelet counts, transfusion dependency, requirement of androgens, cortico-steroids or bone marrow transplantation, and the development of bone marrow clonal cytogenetic abnormalities, myelodysplastic syndrome or acute myeloid leukemia. As expected, a high frequency of spontaneous chromosome breaks was observed in FA patients, especially of chromatid-type. No differences in chromosomes 7 and 8 monosomy, polysomy and non-disjunction were detected between FA patients and controls. The same was true for overall genome haploidy or polyploidy. Interestingly, the spontaneous levels of chromosome fragility but not of numerical abnormalities were correlated to the severity of the haematological disease in FA. None of the variables included in the present investigation (chromosome fragility, chromosome numerical abnormalities and haematological status) were correlated to telomere length.


Assuntos
Aneuploidia , Fragilidade Cromossômica/genética , Anemia de Fanconi/genética , Criança , Cromossomos Humanos Par 7/genética , Cromossomos Humanos Par 8/genética , Anemia de Fanconi/patologia , Feminino , Humanos , Hibridização in Situ Fluorescente , Masculino , Índice de Gravidade de Doença
15.
Haematologica ; 87(3): 331-2, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11869952

RESUMO

Twenty-four children with acute leukemia (21) or chronic myeloid leukemia (3) who relapsed after a first hematopoietic stem cell transplantation (HSCT) underwent a second allogeneic HSCT. Sixteen patients died from relapse or transplant related causes and 8 are alive and disease-free with a probability of event-free survival at 5 years of 32%. These results show that this procedure offers a chance to a subset of these patients.


Assuntos
Transplante de Células-Tronco Hematopoéticas/mortalidade , Leucemia/terapia , Adolescente , Criança , Proteção da Criança , Pré-Escolar , Feminino , Seguimentos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Leucemia/mortalidade , Masculino , Recidiva , Espanha/epidemiologia , Taxa de Sobrevida , Transplante Homólogo/efeitos adversos , Transplante Homólogo/imunologia , Transplante Homólogo/mortalidade
16.
Hum Mol Genet ; 11(4): 439-44, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11854176

RESUMO

Fanconi anemia (FA) is a rare genetic disease characterized by chromosome instability, progressive pancytopenia and cancer susceptibility. Telomeres are intimately related to chromosome stability and play an important role in organismal viability at the hematological level. Since previous works suggested an accelerated shortening of telomeres in FA, we have studied several markers of telomere integrity and function in FA patients and age-matched controls to get insights into the mechanisms and consequences of telomere erosion in FA. A higher frequency of extra-chromosomic TTAGGG signals and of chromosome ends with undetectable TTAGGG repeats was observed in FA cells by fluorescence in situ hybridization (FISH), suggesting intensive breakage at telomeric sequences. This was proven by measuring the frequency of excess of telomeric signals per cell, which was 2.8-fold higher in FA. Consistent with previous reports, quantitative FISH analysis showed an accelerated telomere shortening of 0.68 kb in FA, which occurred concurrently in both chromosome arms in a similar magnitude. Our data therefore suggest that the telomere erosion in FA is caused by a higher rate of breakage at TTAGGG sequences in vivo in differentiated cells, in addition to mere replicative shortening during lymphocyte proliferation. Consistent with impaired telomeres in FA patients, we observed a >10-fold increase in chromosome end fusions in FA compared to normal controls. This observation was independent of TRF2, a telomere binding factor that protects human telomeres from end fusions, since immunohistochemistry studies in FA cell lines and corrected counterparts by retrovirus-mediated transfer of FANCA and FANCD2 cDNA showed that a functional FA pathway is not required for telomere binding of TRF2.


Assuntos
Aberrações Cromossômicas , Anemia de Fanconi/genética , Telômero , Adolescente , Adulto , Criança , Pré-Escolar , Proteínas de Ligação a DNA/metabolismo , Feminino , Humanos , Masculino , Ligação Proteica , Telômero/patologia , Proteína 2 de Ligação a Repetições Teloméricas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...