Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Br J Haematol ; 136(4): 673-6, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17223910

RESUMO

Although haematopoietic cell transplantation (HCT) is curative for sickle cell anaemia (SCA), concerns about its short- and long-term toxicities limit its application. A potential toxicity is an adverse effect on growth. To identify an HCT growth effect, serial height and weight measurements from 53 children and adolescents with SCA after receiving a transplant were compared to historical controls. Hierarchical Linear Models for longitudinal data were used for analysis. In general growth was not impaired by HCT for SCA in young children; however, diminished growth may occur if HCT is carried out near or during the adolescent growth spurt.


Assuntos
Anemia Falciforme/terapia , Transplante de Medula Óssea , Crescimento , Fatores Etários , Envelhecimento/fisiologia , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/fisiopatologia , Antidrepanocíticos/uso terapêutico , Estatura , Transplante de Medula Óssea/efeitos adversos , Criança , Pré-Escolar , Feminino , Seguimentos , Humanos , Hidroxiureia/uso terapêutico , Masculino , Aumento de Peso
2.
Pediatr Transplant ; 9(2): 254-7, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15787803

RESUMO

Invasive fungal infections often prove difficult to eradicate especially in the stem cell transplant setting. Amphotericin has been the mainstay of treatment for years but has significant toxicity. Newer antifungal agents, such as caspofungin, have shown promising results in adults, particularly when used in combination with amphotericin as both drugs differ in their mode of action. However, there are few data from children and no previous published information about the use of Caspofungin after paediatric stem cell transplantation. We report our experience in children with proven invasive fungal infections after stem cell transplantation. This combination was non-toxic, and two of three patients survived their infections.


Assuntos
Anfotericina B/uso terapêutico , Antifúngicos/uso terapêutico , Transplante de Células-Tronco Hematopoéticas , Micoses/tratamento farmacológico , Peptídeos Cíclicos/uso terapêutico , Adolescente , Aspergilose/tratamento farmacológico , Aspergillus fumigatus , Caspofungina , Quimioterapia Combinada , Equinocandinas , Evolução Fatal , Feminino , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Hospedeiro Imunocomprometido , Lipopeptídeos , Lipossomos , Masculino
3.
Arch Dis Child Fetal Neonatal Ed ; 89(2): F101-7, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14977890

RESUMO

Many previously widely accepted neonatal transfusion practices are changing as neonatologists become more aware of the risks to their patients of multiple blood product transfusions. Recent literature and research on neonatal transfusion practice are here reviewed, and practical guidelines and trigger thresholds for blood products commonly used in neonatal medicine are proposed.


Assuntos
Transfusão de Sangue/métodos , Doenças do Recém-Nascido/terapia , Seleção de Pacientes , Albuminas/administração & dosagem , Transfusão de Sangue/normas , Transfusão de Eritrócitos/métodos , Eritropoetina/administração & dosagem , Transfusão Total/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Humanos , Imunoglobulinas Intravenosas , Recém-Nascido , Substitutos do Plasma/uso terapêutico , Transfusão de Plaquetas/métodos , Proteínas Recombinantes
4.
Bone Marrow Transplant ; 32(7): 653-6, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-13130311

RESUMO

Allogeneic haemopoietic stem cell transplantation (SCT) is the only curative option for severe bone marrow (BM) failure in patients with Fanconi anaemia (FA). We have developed a non total body irradiation (TBI) conditioning protocol consisting of fludarabine (120-150 mg/m(2)), low dose of cyclophosphamide (40 mg/kg) and antilymphocyte globulin (45 mg/kg). Graft-versus-host disease (GVHD) prophylaxis was with cyclosporin alone for sibling allografts but also included Campath-1 H (days 1-5 post SCT) for the unrelated allografts. We have performed two sibling and two unrelated BM transplants with a follow-up of 11-51 months. All patients experienced minimal toxicity and were discharged from hospital 28-32 days post SCT. Neutrophil and platelet engraftment occurred from days 11 to 19 and 15 to 34, respectively. All patients achieved stable full donor haemopoiesis with normalisation of the peripheral blood count despite one of them having myelodysplasia (MDS) with 8% blasts prior to the SCT. The only site of acute GVHD was in the skin (grade I-II) and only one patient progressed to limited chronic GVHD. This protocol is associated with reduced toxicity and prompt engraftment in FA patients with AA and/or MDS undergoing SCT using sibling or unrelated donors.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Anemia de Fanconi/terapia , Transplante de Células-Tronco Hematopoéticas/métodos , Condicionamento Pré-Transplante/métodos , Vidarabina/análogos & derivados , Adolescente , Soro Antilinfocitário/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Criança , Pré-Escolar , Ciclofosfamida/administração & dosagem , Anemia de Fanconi/complicações , Feminino , Sobrevivência de Enxerto , Doença Enxerto-Hospedeiro/prevenção & controle , Hematopoese , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Teste de Histocompatibilidade , Humanos , Cinética , Masculino , Irmãos , Transplante Homólogo/imunologia , Vidarabina/administração & dosagem
5.
Mol Hum Reprod ; 9(8): 497-502, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12837927

RESUMO

Strategies for genetic prenatal diagnosis on fetal cells in the maternal circulation have been limited by lack of a cell type present only in fetal blood. However, the recent identification of mesenchymal stem cells (MSC) in first trimester fetal blood offers the prospect of targeting MSC for non-invasive prenatal diagnosis. We developed protocols for fetal MSC enrichment from maternal blood and determined sensitivity and specificity in mixing experiments of male fetal MSC added to female blood, in dilutions from 1 in 10(5) to 10(8). We then used the optimal protocol to isolate fetal MSC from maternal blood in the first trimester, using blood taken after surgical termination of pregnancy as a model of increased feto-maternal haemorrhage. In model mixtures, we could amplify one male fetal MSC in 2.5 x 10(7) adult female nucleated cells, yielding a 100% pure population of fetal cells, but not one fetal MSC in 10(8) nucleated cells. Fetal MSC were identified in one of 20 post-termination maternal blood samples and confirmed as fetal MSC by XY fluorescence in-situ hybridization (FISH), immunophenotyping and osteogenic and adipogenic differentiation. We report the isolation of fetal MSC from maternal blood; however, their rarity in post-termination blood suggests they are unlikely to have a role in non-invasive prenatal diagnosis. Failure to locate these cells routinely may be attributed to their low frequency in maternal blood, to sensitivity limitations of enrichment technology, and/or to their engraftment in maternal tissues soon after transplacental passage. We speculate that gender microchimerism in post-reproductive maternal tissues might result from feto-maternal trafficking of MSC in early pregnancy.


Assuntos
Transfusão Feto-Materna , Feto/anatomia & histologia , Mesoderma/citologia , Diagnóstico Pré-Natal/métodos , Células-Tronco/metabolismo , Adulto , Separação Celular/métodos , Feminino , Humanos , Hibridização in Situ Fluorescente , Masculino , Gravidez , Primeiro Trimestre da Gravidez , Células-Tronco/citologia
6.
Transfus Med ; 12(1): 35-41, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11967135

RESUMO

Platelet transfusions are frequently given to neonatal intensive care unit (NICU) patients with severe thrombocytopenia (platelets less than 50 x 10(9) L(-1)) but no study has assessed whether this is clinically appropriate. To address this we conducted a retrospective review of platelet transfusion practice in patients developing severe thrombocytopenia over 3 years in a single NICU. Out of 901 admissions, 53 (6%) developed severe thrombocytopenia. Twenty-seven neonates received a total of 63 platelet transfusions, the main triggers being: platelet count less than 30 x 10(9) L(-1) (all patients), or less than 50 x 10(9) L(-1) in those with previous haemorrhage or clinical instability. No major haemorrhage occurred during severe thrombocytopenia either in neonates in whom platelet transfusions were withheld (26/53) or in neonates given platelets who survived to discharge (22/27). Five preterm neonates given platelets died but all had overwhelming sepsis or necrotizing enterocolitis and none died directly as a result of haemorrhage. Although the widely used liberal triggers for neonatal platelet transfusion highlighted in this review reflect available guidelines, and represent cautious ('safe') haemostatic practice, they are likely to result in unnecessary transfusion for a significant number of NICU patients. Improved practice requires definition of a safe lower limit for platelet count in stable neonates; effective platelet transfusion strategies for sick neonates; and improved therapies for conditions precipitating severe thrombocytopenia.


Assuntos
Unidades de Terapia Intensiva Neonatal/normas , Transfusão de Plaquetas/normas , Trombocitopenia/terapia , Gerenciamento Clínico , Feminino , Hemorragia/etiologia , Humanos , Recém-Nascido , Masculino , Trabalho de Parto Prematuro , Contagem de Plaquetas , Transfusão de Plaquetas/estatística & dados numéricos , Guias de Prática Clínica como Assunto , Gravidez , Estudos Retrospectivos , Trombocitopenia/complicações , Trombocitopenia/mortalidade , Resultado do Tratamento , Reino Unido
7.
Blood ; 98(8): 2396-402, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11588036

RESUMO

Human mesenchymal stem/progenitor cells (MSCs) have been identified in adult bone marrow, but little is known about their presence during fetal life. MSCs were isolated and characterized in first-trimester fetal blood, liver, and bone marrow. When 10(6) fetal blood nucleated cells (median gestational age, 10(+2) weeks [10 weeks, 2 days]) were cultured in 10% fetal bovine serum, the mean number (+/- SEM) of adherent fibroblastlike colonies was 8.2 +/- 0.6/10(6) nucleated cells (69.6 +/- 10/microL fetal blood). Frequency declined with advancing gestation. Fetal blood MSCs could be expanded for at least 20 passages with a mean cumulative population doubling of 50.3 +/- 4.5. In their undifferentiated state, fetal blood MSCs were CD29(+), CD44(+), SH2(+), SH3(+), and SH4(+); produced prolyl-4-hydroxylase, alpha-smooth muscle actin, fibronectin, laminin, and vimentin; and were CD45(-), CD34(-), CD14(-), CD68(-), vWF(-), and HLA-DR(-). Fetal blood MSCs cultured in adipogenic, osteogenic, or chondrogenic media differentiated, respectively, into adipocytes, osteocytes, and chondrocytes. Fetal blood MSCs supported the proliferation and differentiation of cord blood CD34(+) cells in long-term culture. MSCs were also detected in first-trimester fetal liver (11.3 +/- 2.0/10(6) nucleated cells) and bone marrow (12.6 +/- 3.6/10(6) nucleated cells). Their morphology, growth kinetics, and immunophenotype were comparable to those of fetal blood-derived MSCs and similarly differentiated along adipogenic, osteogenic, and chondrogenic lineages, even after sorting and expansion of a single mesenchymal cell. MSCs similar to those derived from adult bone marrow, fetal liver, and fetal bone marrow circulate in first-trimester human blood and may provide novel targets for in utero cellular and gene therapy.


Assuntos
Diferenciação Celular/fisiologia , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Mesoderma/citologia , Primeiro Trimestre da Gravidez/sangue , Tecido Adiposo/citologia , Tecido Adiposo/embriologia , Antígenos CD/análise , Diferenciação Celular/imunologia , Divisão Celular , Separação Celular , Células Cultivadas , Condrogênese/imunologia , Feminino , Fibroblastos/citologia , Citometria de Fluxo , Antígenos HLA-DR/análise , Humanos , Imunofenotipagem , Fígado/citologia , Fígado/embriologia , Masculino , Osteogênese/imunologia , Gravidez , Pele/citologia , Pele/imunologia
8.
Br J Haematol ; 114(1): 219-25, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11472371

RESUMO

We have prospectively assessed the relative contribution of host and donor to haemopoiesis following stem cell transplantation (SCT) in children with beta-thalassaemia major (n = 35), using karyotype analysis or Southern blot/polymerase chain reaction analysis of variable number tandem repeats on genomic DNA from peripheral blood. Early haemopoiesis was fully donor in origin in 24 out of 35 cases and remained so throughout the post-transplant course in all but one patient, who evolved to stable mixed chimaerism. The remaining 11 cases (31%) initially showed mixed chimaerism: four of these rejected, one eventually eradicated host haemopoiesis to become fully donor haemopoietic, and the remaining six had persistent mixed chimaerism, with 5--38% host haemopoiesis. The risk of graft rejection was high when > 15% host haemopoiesis was present at 3 months post transplant: four out of six such patients rejected their grafts; conversely, zero out of 29 patients with < 15% host haemopoiesis at 3 months rejected (P < 0.0001). There was a higher incidence of significant acute and chronic graft-versus-host disease in patients with full donor chimaerism. These studies confirm that the mixed chimaeric state is common following SCT for thalassaemia, often persists (with up to 4 years follow-up) and is compatible with long-term cure. Analysis of chimaerism in patients undergoing SCT for beta-thalassaemia enables monitoring of engraftment in the early post-transplant period, provides insight into the biology of engraftment and may be useful in identifying patients at high risk of rejection.


Assuntos
Rejeição de Enxerto/genética , Transplante de Células-Tronco Hematopoéticas , Repetições Minissatélites , Talassemia beta/terapia , Sistema ABO de Grupos Sanguíneos , Adolescente , Criança , Pré-Escolar , Quimera , Doença Enxerto-Hospedeiro/genética , Humanos , Estudos Prospectivos , Condicionamento Pré-Transplante , Talassemia beta/genética
9.
Proc Natl Acad Sci U S A ; 98(6): 3294-8, 2001 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-11248072

RESUMO

CD1 molecules are specialized in presenting lipids to T lymphocytes, but identification and isolation of CD1-restricted lipid specific T cells has been hampered by the lack of reliable and sensitive techniques. We here report the construction of CD1d-glycolipid tetramers from fully denatured human CD1d molecules by using the technique of oxidative refolding chromatography. We demonstrate that chaperone- and foldase-assisted refolding of denatured CD1d molecules and beta(2)-microglobulin in the presence of synthetic lipids is a rapid method for the generation of functional and specific CD1d tetramers, which unlike previously published protocols ensures isolation of CD1d tetramers loaded with a single lipid species. The use of human CD1d-alpha-galactosylceramide tetramers for ex vivo staining of peripheral blood lymphocytes and intrahepatic T cells from patients with viral liver cirrhosis allowed for the first time simultaneous analysis of frequency and specificity of natural killer T cells in human clinical samples. Application of this protocol to other members of the CD1 family will provide powerful tools to investigate lipid-specific T cell immune responses in health and in disease.


Assuntos
Antígenos CD1/metabolismo , Ceramidas/metabolismo , Glicolipídeos/metabolismo , Dobramento de Proteína , Animais , Antígenos CD1/genética , Antígenos CD1d , Células Cultivadas , Hepatite C/sangue , Hepatite C/metabolismo , Humanos , Ligantes , Cirrose Hepática/sangue , Cirrose Hepática/metabolismo , Camundongos , Camundongos Knockout , Oxirredução , Coloração e Rotulagem
10.
Br J Haematol ; 112(2): 427-9, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11167843

RESUMO

Allogeneic stem cell transplantation (SCT) represents the treatment of choice for severe bone marrow (BM) failure in patients with Fanconi's anaemia (FA). However, for FA patients developing leukaemic or myelodysplastic transformation, the results of SCT are much less encouraging. We present a 17-year-old girl with myelodysplastic transformation of FA (refractory anaemia with excess blasts) and oculocutaneous albinism, who was treated by sibling SCT using conditioning with fludarabine, cyclophosphamide (CY) and anti-lymphocyte globulin (ALG). She had rapid engraftment with no toxicity and no graft-versus-host disease (GVHD). Twenty-two months after SCT, she had 100% donor chimaerism on Southern blot analysis.


Assuntos
Anemia Refratária com Excesso de Blastos/cirurgia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Anemia de Fanconi/cirurgia , Transplante de Células-Tronco Hematopoéticas/métodos , Condicionamento Pré-Transplante/métodos , Vidarabina/análogos & derivados , Adolescente , Adulto , Ciclofosfamida/administração & dosagem , Ciclosporina/uso terapêutico , Doença Enxerto-Hospedeiro/prevenção & controle , Humanos , Imunoglobulinas/uso terapêutico , Imunossupressores/uso terapêutico , Masculino , Transplante Homólogo , Vidarabina/administração & dosagem
11.
Curr Opin Pediatr ; 13(1): 16-21, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11176238

RESUMO

The healthy fetus has a platelet count of greater than 150 x 10(9)/L by the second trimester of pregnancy and only 2% of term infants are thrombocytopenic at birth. Severe thrombocytopenia (platelets < 50 x 10(9)/L) occurs in fewer than three per 1000 term infants, the most important cause being alloimmune thrombocytopenia. In contrast, in infants admitted to neonatal intensive care units, thrombocytopenia develops in 25% and in up to half of sick preterm infants. Recent evidence shows that these infants mostly have evidence of underlying impaired fetal megakaryocytopoiesis and platelet production following pregnancy complications characterized by placental insufficiency or fetal hypoxia. The mechanism of this is unknown. However, many neonatal complications exacerbate this thrombocytopenic potential and 20% of thrombocytopenias in neonatal intensive care unit patients are severe. Evidence-based guidelines for platelet transfusion therapy in these patients are yet to be defined, but as platelet underproduction underlies most neonatal thrombocytopenias, recombinant hemopoietic growth factors, including thrombopoietin and interkeukin-11, may be useful future therapies.


Assuntos
Trombocitopenia/etiologia , Trombocitopenia/terapia , Feminino , Doenças Fetais/imunologia , Humanos , Recém-Nascido , Unidades de Terapia Intensiva Neonatal , Interleucina-11/uso terapêutico , Troca Materno-Fetal , Transfusão de Plaquetas , Gravidez , Trombocitopenia/classificação , Trombopoetina/uso terapêutico
12.
Best Pract Res Clin Haematol ; 14(4): 807-22, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11924923

RESUMO

Beta-thalassaemia major and sickle-cell disease (SCD) reduce lifespan and quality of life for >300000 children and young adults worldwide. The only cure for both disorders is allogeneic stem cell transplantation (SCT). The decision-making processes in recommending SCT for patients with thalassaemia and SCD are different. For thalassaemia, where transfusion-related iron overload is universal, SCT should be offered to all patients <17 years because long-term survival and thalassaemia-free survival are about 80 and 70% respectively. For thalassaemics unable to comply with medical treatment, SCT offers a significant survival advantage; however, for patients with optimal medical care, short-term survival after SCT is inferior to medical treatment, and SCT instead offers a life free from transfusions and iron chelation. The clinical heterogeneity of SCD means that SCT is recommended only for selected patients with severe disease, particularly sickle-related neurological problems, for whom long-term survival and SCD-free survival after SCT approach 92 and 86% respectively. We here review the evidence available to help physicians evaluate the role of SCT for individual patients with thalassaemia major or SCD.


Assuntos
Hemoglobinopatias/terapia , Transplante de Células-Tronco/mortalidade , Anemia Falciforme/mortalidade , Anemia Falciforme/terapia , Hemoglobinopatias/mortalidade , Humanos , Prognóstico , Transplante de Células-Tronco/métodos , Transplante Homólogo/métodos , Transplante Homólogo/mortalidade , Talassemia beta/mortalidade , Talassemia beta/terapia
13.
Br J Haematol ; 115(4): 1010-4, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11843843

RESUMO

There is mounting evidence to suggest that T-cell-mediated suppression of haemopoiesis is a pathogenetic mechanism in three bone marrow failure syndromes: aplastic anaemia (AA), paroxysmal nocturnal haemoglobinuria (PNH) and myelodysplasia (MDS). T-cell microclones can be detected by sensitive polymerase chain reaction (PCR)-based methods in all three disorders. Recently, larger clonal populations of T-cell large granular lymphocytes (T-LGLs) have been observed in some patients with AA and MDS. Here, we report the development of a large clonal T-LGL population in a patient with bona fide PNH. In this patient, we defined part of the sequence of the T-cell receptor (TCR) beta-chain gene, and we have shown that the large T-LGL population emerged from a background of multiple smaller T-cell clones. Thus, T-LGL clones in AA, MDS and PNH probably expand as a result of antigenic stimulation. It is postulated that the antigen driving clonal T-cell proliferations in these disorders exists on haemopoietic stem cells.


Assuntos
Hemoglobinúria Paroxística/imunologia , Leucemia de Células T/complicações , Linfócitos T/patologia , Adulto , Anemia Aplástica/imunologia , Divisão Celular , Células Clonais , Técnicas de Cocultura , Feminino , Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/patologia , Hemoglobinúria Paroxística/genética , Humanos , Leucemia de Células T/genética , Leucemia de Células T/imunologia , Reação em Cadeia da Polimerase/métodos
14.
Biol Blood Marrow Transplant ; 7(12): 665-73, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11787529

RESUMO

A multicenter investigation of allogeneic bone marrow transplantation for children with sickle cell disease was conducted that included 27 European and North American transplant centers. Fifty-nine patients who ranged in age from 3.3 to 15.9 years (median, 10.1 years) received HLA-identical sibling marrow allografts between September 1991 and April 2000. Fifty-five patients survive, and 50 survive free from sickle cell disease, with a median follow-up of 42.2 months (range, 11.8 to 115 months) after transplantation. Of the 50 patients with successful allografts, 13 developed stable mixed donor-host hematopoietic chimerism. The level of donor chimerism, measured > or =6 months after transplantation in peripheral blood, varied between 90% and 99% in 8 patients. Five additional patients had a lower proportion of donor cells (range, 11% to 74%). Among these 5 patients, hemoglobin levels varied between 11.2 and 14.2 g/dL (median, 11.3 g/dL; mean, 12.0 g/dL). In patients who had donors with a normal hemoglobin genotype (Hb), the Hb S fractions were 0%, 0%, and 7%, corresponding to donor chimerism levels of 67%, 74%, and 11%, respectively. Among patients who had donors with sickle trait, the Hb S fractions were 36% and 37%, corresponding to donor chimerism levels of 25% and 60%, respectively. Thus, allograft recipients with stable mixed chimerism had Rb S levels similar to donor levels, and only 1 patient required a red blood cell transfusion beyond 90 days posttransplantation. None of the patients have experienced painful events or other clinical complications related to sickle cell disease after transplantation. These observations strongly suggest that patients with sickle cell disease who develop persistent mixed hematopoietic chimerism after transplantation experience a significant ameliorative effect.


Assuntos
Anemia Falciforme/terapia , Transplante de Medula Óssea , Transplante de Medula Óssea/métodos , Hematopoese , Quimeras de Transplante , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/mortalidade , Criança , Pré-Escolar , Feminino , Seguimentos , Sobrevivência de Enxerto , Hemoglobina Falciforme/análise , Hemoglobinas/análise , Humanos , Masculino , Estudos Prospectivos , Doadores de Tecidos , Transplante Isogênico , Resultado do Tratamento
15.
Blood ; 96(7): 2613-20, 2000 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11001919

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired clonal disorder of the hematopoietic stem cell (HSC). Somatic mutations in the PIG-A gene result in the deficiency of several glycosylphosphatidylinositol-linked proteins from the surface of blood cells. This explains intravascular hemolysis but does not explain the mechanism of bone marrow failure that is almost invariably seen in PNH. In view of the close relationship between PNH and idiopathic aplastic anemia (IAA), it has been suggested that the 2 disorders might have a similar cellular pathogenesis, namely, that autoreactive T-cell clones are targeting HSCs. In this paper, we searched for abnormally expanded T-cell clones by size analysis of the complementarity-determining region 3 (CDR3) in the beta variable chain (BV) messenger RNA (mRNA) of the T-cell receptor (TCR) in 19 patients with PNH, in 7 multitransfused patients with hemoglobinopathy. and in 11 age-matched healthy individuals. We found a significantly higher degree of skewness in the TCR BV repertoire of patients with PNH, compared with controls (R(2) values 0.82 vs 0.91, P <.001). The mean frequency of skewed families per individual was increased by more than 2-fold in patients with PNH, compared with controls (28% +/- 19.6% vs 11.4% +/- 6%, P =.002). In addition, several TCR BV families were significantly more frequently skewed in patients with PNH than in controls. These findings provide experimental support for the concept that PNH, like IAA, has an immune pathogenesis. In addition, the identification of expanded T-cell clones by CDR3 size analysis will help to investigate the effect of HSC-specific T cells on normal and PNH HSCs.


Assuntos
Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Hemoglobinúria Paroxística/imunologia , Linfócitos T/imunologia , Adolescente , Adulto , Feminino , Células-Tronco Hematopoéticas/imunologia , Hemoglobinúria Paroxística/genética , Humanos , Região Variável de Imunoglobulina/genética , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , RNA Mensageiro/química , Análise de Sequência de RNA
16.
Early Hum Dev ; 59(1): 1-12, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10962163

RESUMO

C-mpl ligand or thrombopoietin (Tpo) is increasingly recognised as the major regulator of platelet homeostasis in humans. Relatively little is known about Tpo in the fetus and neonate but no evidence has yet been found to suggest any fundamental difference in Tpo structure, function and regulation in the fetus and neonate compared to older age groups. Tpo mRNA transcripts have been detected in the fetus as early as 6 weeks post conception and the liver appears to be the main site of Tpo production in both the fetus and neonate. The vast majority of healthy newborns have detectable levels of circulating Tpo and raised Tpo levels are commonly, but not consistently, found in thrombocytopenic neonates. In adults receptor binding and subsequent metabolism of Tpo is proposed as the main method of regulation of the circulating Tpo level. Preliminary studies in neonates showing increased Tpo levels most often during thrombocytopenia accompanied by reduced megakaryocytopoiesis supports this concept. In addition to this demonstrable fetal and neonatal endogenous Tpo production megakaryocyte progenitor and precursor cells from the fetus and from preterm and term newborns proliferate and differentiate extensively in-vitro in response to exogenous Tpo. Furthermore a recent study has shown a marked rise in platelet count in newborn rhesus monkeys administered one form of recombinant Tpo. Although these studies remain at an early stage together these findings strongly suggest that, as in adults, Tpo is the major regulator of platelet homeostasis in the fetus and neonate. Thrombocytopenia is common in sick neonates and progress in understanding this important clinical problem is likely to be greatly enhanced by the current and future research into Tpo production, function and regulation in the healthy and thrombocytopenic fetus and neonate.


Assuntos
Feto/fisiologia , Recém-Nascido/fisiologia , Trombopoetina/fisiologia , Adulto , Animais , Animais Recém-Nascidos , Feminino , Humanos , Gravidez
17.
Blood ; 95(6): 1918-24, 2000 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10706855

RESUMO

Fifty children who had symptomatic sickle cell disease received matched sibling marrow allografts between September 1991 and March 1999, with Kaplan-Meier probabilities of survival and event-free survival of 94% and 84%, respectively. Twenty-six patients (16 male, 10 female) had at least 2 years of follow-up after transplantation and were evaluated for late effects of transplantation and for its impact on sickle cell-related central nervous system (CNS) and pulmonary disease. Patients ranged between 3.3 and 14.0 (median, 9. 4) years of age and had a median follow-up of 57.9 (range 38-95) months after transplantation. Among 22 of 26 patients who had stable donor engraftment, complications related to sickle cell disease resolved, and none experienced further episodes of pain, stroke, or acute chest syndrome. All 10 engrafted patients with a prior history of stroke had stable or improved cerebral magnetic resonance imaging results. Pulmonary function tests were stable in 22 of the 26 patients, worse in two, and not studied in two. Seven of eight patients transplanted for recurrent acute chest syndrome had stable pulmonary function. Linear growth measured by median height standard deviation score improved from -0.7 before transplantation to -0.2 after transplantation. An adverse effect of busulfan conditioning on ovarian function was demonstrated in five of seven evaluable females who are currently at least 13 years of age. None of the four males tested had elevated serum gonadotropin levels. These data confirm that allogenic bone marrow transplantation establishes normal erythropoiesis and is associated with improved growth and stable CNS imaging and pulmonary function in most patients. (Blood. 2000;95:1918-1924)


Assuntos
Anemia Falciforme/terapia , Transplante de Medula Óssea , Adolescente , Anemia Falciforme/complicações , Anemia Falciforme/mortalidade , Estatura , Doenças Cardiovasculares/etiologia , Criança , Pré-Escolar , Estudos de Coortes , Intervalo Livre de Doença , Glândulas Endócrinas/metabolismo , Feminino , Seguimentos , Humanos , Pulmão/fisiologia , Masculino , Fatores de Tempo , Doadores de Tecidos
18.
Br J Haematol ; 111(4): 1158-63, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11167756

RESUMO

The mechanism responsible for the bone marrow failure that is almost invariable in paroxysmal nocturnal haemoglobinuria (PNH) is unknown. Based on the close association between PNH and idiopathic aplastic anaemia, a plausible pathogenetic model predicts that, in PNH, autoreactive T cells specific for haemopoietic stem cells (HSCs) cause depletion of normal HSCs, whereas PNH HSCs escape this T-cell-mediated attack. In this study, we addressed the hypothesis that PNH HSCs are resistant to the cytotoxic effect of T cells because they lack surface expression of one or more glycosylphosphatidylinositol (GPI)-linked molecules. We tested the sensitivity of normal and PNH Epstein-Barr virus (EBV)-transformed B-cell lymphoblastoid cell lines (BLCLs) to the cytotoxic effect of autologous EBV-specific T-cell lines and clones from a patient with PNH in an in vitro experimental system. We found that the PNH BLCLs were no less sensitive to T-cell-mediated cytotoxicity than non-PNH isogenic BLCLs, indicating that GPI-linked molecules on the surface of target cells are not required for killing by T cells. This suggests that the mechanism whereby PNH HSCs survive T-cell attack is not because of the lack of surface expression of one or more GPI-linked molecules. By implication, other mechanisms become more probable.


Assuntos
Hemoglobinúria Paroxística/imunologia , Células-Tronco/imunologia , Linfócitos T Citotóxicos/imunologia , Antígenos CD59/metabolismo , Morte Celular , Linhagem Celular Transformada , Técnicas de Cocultura , Citotoxicidade Imunológica , Herpesvirus Humano 4 , Humanos , Linfoma de Células B/imunologia , Linfoma de Células B/metabolismo
20.
Br J Haematol ; 106(1): 162-6, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10444180

RESUMO

There are considerable differences in haemopoietic activity between young children and adults on the one hand, and between adults and the elderly on the other. A fundamental unanswered question is whether these differences relate to discrete stages or are part of a continuous process. We have sought to define aspects of the haematological ageing process, and have found that results from children with bone marrow failure syndromes differ from age-matched reference values. Haemopoietic cells were obtained from umbilical cord blood, from blood and bone marrow of healthy individuals and from the blood of young patients with bone marrow failure syndromes. Clonogenic myeloid progenitors (CFU-GM) were grown in semi-solid medium to measure their frequency; the proliferative capacity of myeloid progenitors was measured by replating colonies and observing secondary colony formation. We found that the frequency of CFU-GM in normal marrow increased and their proliferative capacity decreased exponentially with age. The proliferative capacity of CFU-GM in normal blood also decreased exponentially with age. This relationship extrapolated back to the levels of proliferation measured for cord blood CFU-GM (age = 0). The proliferative capacities of CFU-GM from children with bone marrow failure syndromes were severely reduced compared with age-matched reference values. These results indicate that a decline in haemopoietic progenitor cell function begins at birth and continues throughout life. This decline may occur prematurely in childhood marrow failure syndromes with a predisposition to leukaemia.


Assuntos
Envelhecimento/patologia , Doenças da Medula Óssea/patologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/patologia , Adolescente , Adulto , Divisão Celular , Criança , Pré-Escolar , Humanos , Lactente , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...