Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cytometry B Clin Cytom ; 98(2): 179-192, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31705743

RESUMEN

BACKGROUND: The diagnosis of paroxysmal nocturnal hemoglobinuria (PNH) relies on flow cytometric demonstration of loss of glycosyl-phosphatidyl inositol (GPI)-anchored proteins from red blood cells (RBC) and white blood cells (WBC). High-sensitivity multiparameter assays have been developed to detect loss of GPI-linked structures on PNH neutrophils and monocytes. High-sensitivity assays to detect PNH phenotypes in RBCs have also been developed that rely on the loss of GPI-linked CD59 on CD235a-gated mature RBCs. The latter is used to delineate PNH Type III (total loss of CD59) and PNH Type II RBCs (partial loss of CD59) from normal (Type I) RBCs. However, it is often very difficult to delineate these subsets, especially in patients with large PNH clones who continue to receive RBC transfusions, even while on eculizumab therapy. METHODS: We have added allophycocyanin (APC)-conjugated CD71 to the existing CD235aFITC/CD59PE RBC assay allowing simultaneous delineation and quantification of PNH Type III and Type II immature RBCs (iRBCs). RESULTS: We analyzed 24 medium to large-clone PNH samples (>10% PNH WBC clone size) for PNH Neutrophil, PNH Monocyte, Type III and Type II PNH iRBCs, and where possible, Type III and Type II PNH RBCs. The ability to delineate PNH Type III, Type II, and Type I iRBCs was more objective compared to that in mature RBCs. Additionally, total PNH iRBC clone sizes were very similar to PNH WBC clone sizes. CONCLUSIONS: Addition of CD71 significantly improves the ability to analyze PNH clone sizes in the RBC lineage, regardless of patient hemolytic and/or transfusion status.


Asunto(s)
Antígenos CD/fisiología , Eritrocitos/metabolismo , Citometría de Flujo/métodos , Hemoglobinuria Paroxística/diagnóstico , Receptores de Transferrina/fisiología , Antígenos CD/sangre , Antígenos CD59/metabolismo , Diferenciación Celular , Estudios de Cohortes , Diagnóstico Diferencial , Eritrocitos/patología , Citometría de Flujo/instrumentación , Citometría de Flujo/normas , Glicoforinas/metabolismo , Hemoglobinuria Paroxística/sangre , Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/patología , Humanos , Inmunofenotipificación/instrumentación , Inmunofenotipificación/métodos , Inmunofenotipificación/normas , Recuento de Leucocitos/instrumentación , Recuento de Leucocitos/métodos , Leucocitos/patología , Monocitos/metabolismo , Monocitos/patología , Neutrófilos/metabolismo , Neutrófilos/patología , Receptores de Transferrina/sangre
2.
Rev. pediatr. electrón ; 16(3): 21-27, oct. 2019. tab
Artículo en Español | LILACS | ID: biblio-1046282

RESUMEN

La hemoglobinuria paroxística nocturna (HPN) es una enfermedad clonal y adquirida causada por una mutación somática en el gen PIG-A que se encuentra en el cromosoma X y codifica una proteína involucrada en la síntesis del glicosilfosfatidilinositol (GPI), el cual le sirve como anclaje a muchas proteínas de la membrana celular produciendo mayor sensibilidad al complemento. Los distintos signos y síntomas que se presentan tienen gran impacto en la calidad de vida de los pacientes, por lo que un diagnóstico correcto es de vital importancia. Actualmente, la citometría de flujo multiparamétrica es la metodología de elección para detectar y seguir al paciente con HPN.


Paroxysmal nocturnal hemoglobinuria (PNH) is a clonal and acquired disease caused by a somatic mutation in the PIG-A gene found on the X chromosome and encoding a protein involved in the synthesis of glycosylphosphatidylinositol (GPI), which serves as anchoring to many proteins of the cell membrane producing greater sensitivity to complement. The different signs and symptoms that appear have a great impact on the quality of life of patients, so a correct diagnosis is of vital importance. Currently, multiparameter flow cytometry is the methodology of choice to detect and follow the patient with PNH.


Asunto(s)
Humanos , Niño , Hemoglobinuria Paroxística/diagnóstico , Hemoglobinuria Paroxística/terapia , Diagnóstico Diferencial , Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/etiología
3.
Intern Med J ; 46(9): 1044-53, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27305361

RESUMEN

BACKGROUND: Paroxysmal nocturnal haemoglobinuria (PNH) is a rare disease. Although much progress has been made in the understanding of the pathophysiology of the disease, far less is known with respect to the clinical outcomes of patients with PNH. Few retrospective studies provide survival estimates, and even fewer have explored the clinical heterogeneity of the disease. Haemolytic and aplastic anaemia (AA) forms of the disease have been recognised as main disease categories, with the haemolytic form being associated with the worst prognosis by the largest studied cohort some years ago. AIMS: To describe mortality and causes of death in PNH overall and by PNH classification and to evaluate risk factors associated with mortality. METHODS: We analysed data of 2356 patients enrolled in the International PNH Registry with multivariate analyses, using time-dependent covariates. Patients were classified into haemolytic, AA/PNH syndrome or intermediate PNH. RESULTS: Overall, 122 (5.2%) patients died after enrolment, the incidence according to subcategories being 5.1, 11.7, 2.0 and 4.8% for patients with haemolytic PNH, AA-PNH, intermediate and insufficient data respectively. Older age and decreased performance status also affected survival in multivariate analysis. Improved outcome of patients with haemolytic PNH suggests that eculizumab treatment in PNH may be associated with improved survival. CONCLUSION: A detailed analysis of clinical presentations and causes of death in patients with PNH, overall and by disease subcategories, provide evidence that in the current era, patients with haemolytic PNH are no longer those who harbour the worst prognosis. This finding differs sharply from what has been previously reported.


Asunto(s)
Anemia Aplásica/epidemiología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Hemoglobinuria Paroxística/mortalidad , Hemoglobinuria Paroxística/terapia , Trombosis/epidemiología , Adulto , Causas de Muerte , Transfusión de Eritrocitos , Femenino , Francia , Hemoglobinuria Paroxística/clasificación , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Análisis Multivariante , Pronóstico , Sistema de Registros , Estudios Retrospectivos , Factores de Riesgo , Análisis de Supervivencia
4.
Ann Hematol ; 95(1): 125-133, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26416513

RESUMEN

We retrospectively assessed the clinical characteristics of patients with paroxysmal nocturnal hemoglobinuria (PNH) according to severity of cytopenia. A total of 282 patients with hematological parameters assessed at the time of diagnosis of PNH were included. There were 24 patients with PNH/severe aplastic anemia (SAA) (at least two of the three criteria; hemoglobin ≤8 g/dL; absolute neutrophil count (ANC) <0.5 × 10(9)/L; platelet count <20 × 10(9)/L), 96 patients with PNH/aplastic anemia (AA) (at least two of the three criteria; hemoglobin ≤10 g/dL; ANC 0.5-1.5 × 10(9)/L; platelet count 20-100 × 10(9)/L), and 162 classic PNH patients. Compared with the classic PNH subgroup, the PNH/SAA subgroup had a significantly lower median granulocyte PNH clone size (26.7 vs. 51.0 %, P = 0.021) and lower incidence of lactate dehydrogenase ≥1.5 times the upper limit of normal (52.9 vs. 80.0 %, P = 0.049). The incidence of thromboembolism was similar in both subgroups. Overall survival was significantly lower in the PNH/SAA subgroup than in the classic PNH subgroup (P = 0.033). Our findings suggest that identification of patients with PNH/SAA at the time of diagnosis is important because of different clinical manifestations and poorer outcome compared with patients with classic PNH (clinicaltrials.gov identifier: #NCT01224483).


Asunto(s)
Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/diagnóstico , Pancitopenia/clasificación , Pancitopenia/diagnóstico , Sistema de Registros , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Femenino , Hemoglobinuria Paroxística/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Pancitopenia/mortalidad , República de Corea/epidemiología , Estudios Retrospectivos , Tasa de Supervivencia/tendencias , Adulto Joven
5.
Rev. bras. hematol. hemoter ; 37(2): 90-97, Mar-Apr/2015. tab, graf
Artículo en Inglés | LILACS | ID: lil-746093

RESUMEN

Background: Paroxysmal nocturnal hemoglobinuria is an acquired chronic hemolytic ane- mia, which often manifests as peripheral blood cytopenias and thrombosis. Objective: The aim of this study is to describe a Brazilian population of paroxysmal nocturnal hemoglobinuria patients. Methods: One hundred and three paroxysmal nocturnal hemoglobinuria cases were retrospectively reviewed and the clinical presentation, thrombosis, survival, and clone size were assessed. Diagnosis was established by flow cytometry. Results: Fifty-two male and 51 female patients with a median age of 24.1 years (5.5-62 years) were studied. Clinical symptoms included hemoglobinuria (18.4%), infection (46.6%) and thrombosis (16.5%), and 80.6% had pancytopenia. Patients were classified as classic parox- ysmal nocturnal hemoglobinuria (10), paroxysmal nocturnal hemoglobinuria with aplastic anemia (39), and paroxysmal nocturnal hemoglobinuria with subclinical features and aplas- tic anemia (54). There were significant differences in terms of median age, size of clone, clinical symptoms, and peripheral blood cell counts between the three subcategories. The clone size in erythrocytes and granulocytes were respectively 0.04% (range: 0-18%) and 7.3% (range: 0.3-68.7%) in patients with subclinical features and aplastic anemia, 15.8% (range: 0-99.7%) and 63.0% (range: 1.7-99.8%) in patients with aplastic anemia alone, and 82.2% (range: 0-99.85%) and 98.0% (81.3-100.0%) in Classic disease. Statistical differences were identified for platelets (p-value = 0.001), lactate dehydrogenase (p-value = 0.002) and the clone size (p-value < 0.001) in patients who suffered thrombotic events compared to those who did not. Overall survival was 81.7%, with patients with subclinical features and aplastic anemia having lower overall survival (76.5%). Conclusion: This retrospective review of 103 patients over an 11-year period represents the largest collection of paroxysmal...


Asunto(s)
Humanos , Citometría de Flujo , Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/diagnóstico , Médula Ósea/patología
6.
Int J Hematol ; 96(1): 34-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22562435

RESUMEN

Distinguishing hypoplastic myelodysplastic syndrome from aplastic anemia (AA) is challenging. In the present study, Japanese and Chinese pediatric hematologists and pathologists conducted a joint review of bone marrow (BM) smears and trephine biopsies in 100 children with acquired BM failure syndrome, using the criteria proposed in the 2008 edition of the World Health Organization classification of hematopoietic and lymphoid tissues. The final consensus for the diagnoses of 100 children was AA in 29 patients, refractory cytopenia of childhood (RCC) in 58 patients, and refractory cytopenia with multilineage dysplasia (RCMD) in 13 patients. No significant differences between Japanese and Chinese children were found with regards to clinical and laboratory findings, or the distribution of diagnoses. Patients with RCC/RCMD showed milder disease severity and BM hypocellularity than those with AA. To establish the provisional entities for RCC, it is essential to prospectively compare the clinical outcomes between AA and RCC groups in a large number of patients.


Asunto(s)
Anemia Aplásica/diagnóstico , Anemia Refractaria/diagnóstico , Pueblo Asiatico , Hemoglobinuria Paroxística/diagnóstico , Adolescente , Anemia Aplásica/clasificación , Anemia Refractaria/clasificación , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Niño , Preescolar , China , Diagnóstico Diferencial , Femenino , Hemoglobinuria Paroxística/clasificación , Humanos , Lactante , Japón , Masculino , Organización Mundial de la Salud
7.
Eur J Haematol ; 83(6): 503-11, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19686268

RESUMEN

Paroxysmal nocturnal haemoglobinuria (PNH) is a rare acquired clonal disorder of haematopoietic stem cells. The molecular defect in PNH is mutation in the phosphotidylinositol glycan complementation class A (PIGA gene) causing defect in glycosylphosphatidylinositol anchored proteins (Cell, 73, 1993, 703). The deficiency of these GPI-anchored proteins on the membranes of haematopoietic cells lead to the various clinical manifestations of PNH. Clinically PNH is classified into classic PNH, PNH in the setting of another specified bone marrow disorder and sub clinical PNH. Size of the PNH clone differs in these different subtypes. The management of PNH has been revolutionized by the advent of monoclonal antibody, eculizumab. Thus, today it is important to have sensitive tests to diagnose and monitor the clone size in patients of PNH. Before 1990, diagnosis of PNH was made using complement based tests. However in the last decade, flowcytometry has become the gold standard diagnostic test as it has increased sensitivity to detect small clones, ability to measure clone size and is not affected by blood transfusions. This review is aimed to focus mainly on the different methods available for the detection of PNH clone and the recent advances and recommendations for the flowcytometric diagnosis of PNH.


Asunto(s)
Separación Celular/métodos , Citometría de Flujo/métodos , Hemoglobinuria Paroxística/diagnóstico , Examen de la Médula Ósea , Antígenos CD55/análisis , Antígenos CD59/análisis , Tamaño de la Célula , Células Clonales/patología , Ensayo de Actividad Hemolítica de Complemento , Eritrocitos/química , Eritrocitos/ultraestructura , Colorantes Fluorescentes/análisis , Glicosilfosfatidilinositoles/metabolismo , Pruebas de Hemaglutinación , Hemoglobinuria Paroxística/sangre , Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/complicaciones , Hemoglobinuria Paroxística/genética , Humanos , Inmunofenotipificación/normas , Leucocitos/química , Leucocitos/ultraestructura , Proteínas de la Membrana/análisis , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Nefelometría y Turbidimetría/normas , Sensibilidad y Especificidad , Trombofilia/sangre , Trombofilia/etiología
8.
Lancet ; 373(9665): 759-67, 2009 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-19144399

RESUMEN

The complement system plays a central part in both innate and acquired immunity, but the contribution of complement activation to pathobiology is largely ancillary. An exception to the non-dominant role of complement in disease is the haemolytic anaemia of paroxysmal nocturnal haemoglobinuria (PNH). The intravascular haemolysis that is the clinical hallmark of PNH is a consequence of deficiency of the complement inhibitory proteins decay accelerating factor (DAF, CD55) and membrane inhibitor of reactive lysis (MIRL, CD59). Eculizumab is a humanised monoclonal antibody that binds and prevents activation of complement C5 and the subsequent formation of the cytolytic membrane attack complex of complement. Eculizumab inhibits the intravascular haemolysis of PNH, reduces transfusion requirements, stabilises haemoglobin concentration, and improves quality of life. Although chronic treatment with eculizumab increases the risk of infections with Neisseria meningitides, the drug is generally safe and well tolerated. But as is the case with other drugs developed for treatment of ultra-orphan diseases, eculizumab is expensive, and treatment must continue indefinitely because C5 inhibition does not affect the process (ie, clonal proliferation of haemopoietic stem cells with a mutant phosphatidylinositol glycan complementation class A [PIGA] gene) that underlies PNH. Moreover, due to the heterogeneous nature of the disease, treatment with eculizumab is not appropriate for all patients with PNH.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Hemoglobinuria Paroxística/tratamiento farmacológico , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Ensayos Clínicos Fase II como Asunto , Método Doble Ciego , Femenino , Semivida , Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/fisiopatología , Hemólisis/efectos de los fármacos , Humanos , Masculino , Estudios Multicéntricos como Asunto , Ensayos Clínicos Controlados Aleatorios como Asunto , Distribución Tisular
9.
Haematologica ; 94(1): 29-37, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19001281

RESUMEN

BACKGROUND: The presence of paroxysmal nocturnal hemoglobinuria clones in the setting of aplastic anemia or myelodysplastic syndrome has been shown to have prognostic and therapeutic implications. However, the status of paroxysmal nocturnal hemoglobinuria clones in various categories of myelodysplastic syndrome and in other bone marrow disorders is not well-studied. DESIGN AND METHODS: By using multiparameter flow cytometry immunophenotypic analysis with antibodies specific for four glycosylphosphatidylinositol-anchored proteins (CD55, CD59, CD16, CD66b) and performing an aerolysin lysis confirmatory test in representative cases, we assessed the paroxysmal nocturnal hemoglobinuria-phenotype granulocytes in 110 patients with myelodysplastic syndrome, 15 with myelodysplastic/myeloproliferative disease, 5 with idiopathic myelofibrosis and 6 with acute myeloid leukemia. RESULTS: Paroxysmal nocturnal hemoglobinuria-phenotype granulocytes were detected in nine patients with low grade myelodysplastic syndrome who showed clinicopathological features of bone marrow failure, similar to aplastic anemia. All paroxysmal nocturnal hemoglobinuria-positive cases demonstrated loss of the four glycosylphosphatidylinositol-anchored proteins, with CD16(-)CD66b(-) clones being larger than those of CD55(-)CD59(-) (p<0.05). Altered glycosylphosphatidylinositol-anchored protein expression secondary to granulocytic hypogranulation, immaturity, and/or immunophenotypic abnormalities was present in a substantial number of cases and diagnostically challenging. CONCLUSIONS: These results show that routine screening for paroxysmal nocturnal hemoglobinuria clones in patients with an intrinsic bone marrow disease who show no clinical evidence of hemolysis has an appreciable yield in patients with low grade myelodysplastic syndromes. The recognition of diagnostic caveats and pitfalls associated with the underlying intrinsic bone marrow disease is essential in interpreting paroxysmal nocturnal hemoglobinuria testing correctly. In our experience, the CD16/CD66b antibody combination is superior to CD55/CD59 in screening for subclinical paroxysmal nocturnal hemoglobinuria because it detects a large clone size and is less subject to analytical interference.


Asunto(s)
Enfermedades de la Médula Ósea/complicaciones , Enfermedades de la Médula Ósea/diagnóstico , Hemoglobinuria Paroxística/complicaciones , Hemoglobinuria Paroxística/diagnóstico , Síndromes Mielodisplásicos/complicaciones , Síndromes Mielodisplásicos/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Enfermedades de la Médula Ósea/sangre , Enfermedades de la Médula Ósea/clasificación , Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Diferenciación Celular , Femenino , Hemoglobinuria Paroxística/sangre , Hemoglobinuria Paroxística/clasificación , Humanos , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/clasificación , Fenotipo
10.
Blood ; 112(8): 3099-106, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18535202

RESUMEN

The natural history of paroxysmal nocturnal hemoglobinuria (PNH) clinical subcategories (classic PNH and aplastic anemia [AA]/PNH syndrome) is still unknown. We retrospectively studied 460 PNH patients diagnosed in 58 French hematologic centers from 1950 to 2005. The median (SE) follow-up time was 6.8 (0.5) years. The median survival time (SE) was 22 (2.5) years. We identified 113 patients with classic PNH, 224 patients with AA-PNH syndrome, and 93 (22%) intermediate patients who did not fit within these 2 categories. At presentation, classic PNH patients were older, with more frequent abdominal pain and displayed higher levels of GPI-AP-deficient granulocytes. A time-dependent improved survival was observed. In classic PNH, diagnoses before 1986 (hazard ratio [HR]: 3.6; P = .01) and increasing age (P < .001) were associated with worse survival prognoses, whereas use of androgens within the first year after diagnosis was protective (HR, 0.17; P = .01). Transfusions before 1996 (HR, 2.7; P = .007) led to lower survival rates in patients with AA-PNH syndrome, whereas immunosuppressive treatment was associated with better outcomes (HR, 0.33; P = .03). Evolution to thrombosis affected survival in both subcategories (classic PNH: HR, 7.8 [P < .001]; AA-PNH syndrome: HR, 33.0 [P < .001]). Evolution to bicytopenia or pancytopenia for classic PNH (HR, 7.3, P < .001) and malignancies for AA-PNH syndrome (HR, 48.8; P < .001) were associated with worse outcomes. Although clinical presentation and prognosis factors are different, classic PNH and AA-PNH syndrome present roughly similar outcomes, affected mainly by complications.


Asunto(s)
Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/diagnóstico , Adulto , Femenino , Hematología/métodos , Hemoglobinuria Paroxística/complicaciones , Humanos , Inmunosupresores/uso terapéutico , Masculino , Persona de Mediana Edad , Análisis Multivariante , Pronóstico , Modelos de Riesgos Proporcionales , Riesgo , Trombosis/genética , Factores de Tiempo , Resultado del Tratamiento
11.
J Neurol ; 251(9): 1050-9, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15372245

RESUMEN

The clinical diagnosis of Marchiafava-Bignami disease (MBD) has considerably changed during recent decades with brain MRI providing the opportunity of a reliable in-vivo diagnosis. However, semiologic and neuroimaging characteristics of the currently known spectrum of MBD have not been investigated systematically, and knowledge of clinicoradiologic associations is sketchy. We report an illustrative case with limited callosal involvement on MRI and a favorable outcome and have reviewed literature on clinical and radiologic features in 50 cases of MBD diagnosed in vivo since 1985. Our reviewed data suggest the differentiation of two clinicoradiologic subtypes: Type A is characterized by major impairment of consciousness, T2-hyperintense swelling of the entire corpus callosum on early MRI and poor outcome. Type B shows at most slight impairment of consciousness, partial callosal lesions on MRI and a favorable outcome. Differentiation of these clinicoradiologic subtypes may help resolve inconsistencies of the established clinical classification resulting from new insights into the clinical course and prognosis of MBD by structural neuroimaging.


Asunto(s)
Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/diagnóstico por imagen , Adulto , Anciano , Femenino , Hemoglobinuria Paroxística/diagnóstico , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Radiografía
12.
Int J Hematol ; 77(2): 113-20, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12627845

RESUMEN

The clinical pathology of paroxysmal nocturnal hemoglobinuria (PNH) involves 3 complications: hemolytic anemia, thrombosis, and hematopoietic deficiency. The first 2 are clearly the result of the cellular defect in PNH, the lack of proteins anchored to the membrane by the glycosylphosphatidylinositol anchor. The hemolytic anemia results in syndromes primarily related to the fact that the hemolysis is extracellular. Thrombosis is most significant in veins within the abdomen, although a number of other thrombotic syndromes have been described. The hematopoietic deficiency may be the same as that in aplastic anemia, a closely related disorder, and may not be due to the primary biochemical defect. The relationship to aplastic anemia suggests a nomenclature that emphasizes the predominant clinical manifestations in a patient. This relationship does not explain cases that appear to be related to myelodysplastic syndromes or the transition of some cases of PNH to leukemia. Treatment, except for bone marrow transplantation, remains noncurative and in need of improvement.


Asunto(s)
Hemoglobinuria Paroxística , Hemoglobinuria Paroxística/complicaciones , Anemia Aplásica/etiología , Anemia Hemolítica/complicaciones , Anemia Hemolítica/etiología , Anemia Hemolítica/terapia , Enfermedades de la Médula Ósea/etiología , Hematopoyesis/fisiología , Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/terapia , Humanos , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/etiología , Trombosis de la Vena/patología
13.
Exp Hematol ; 31(1): 65-72, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12543108

RESUMEN

Cellular prion protein (PrP(c)) is a glycosylphosphatidylinositol (GPI)-anchored protein (GPI-AP) constitutively expressed by neurons but also in hematopoietic cells. In trasmissible spongiform encephalopathies, the protease-resistant form of prion (PrP (s c)) converts the host PrP(c) into the pathologic form. We have investigated PrP(c) expression in hematopoietic cells from paroxysmal nocturnal hemoglobinuria (PNH). In this disease, due to somatic mutations in PIG-A gene, biosynthesis of the (GPI)-anchor is impaired and affected cells lack membrane expression of all GPI-AP. Normal and PNH hematopoietic progenitors and paired wild-type (WT) and PIG-A mutant cell lines were used for analysis of intracellular and surface PrP(c) expression using flow cytometry and Western blot.By flow cytometry, PrP(c) was constitutively present on normal CD34(+) cells, including more immature CD38(dim) cells, as well as hematopoietic cell lines. Similar results were obtained in purified CD34(+). Phospholipase C treatment confirmed that PrP(c) was expressed on the membrane via the GPI-anchor. In PNH patients, GPI-AP-deficient CD34(+) cells lacked PrP(c) membrane expression. PIG-A-mutated cell lines (Jurkat, K562, C(EBV), A(EBV)), in contrast to their normal counterparts, did not express surface PrP(c). However, we detected intracellular PrP(c) at approximately equivalent levels in both normal and PIG-A-mutated cells using intracellular flow cytometry and Western blotting. Cells and cell lines with PNH phenotype together with their normal counterparts may be a suitable system to explore the function of membrane PrP(c) in the hematopoietic system. Conversely, PrP(c) is a good model to elucidate the fate of GPI-AP in PIG-A-deficient cells.


Asunto(s)
Células Madre Hematopoyéticas/química , Hemoglobinuria Paroxística/sangre , Proteínas PrPC/deficiencia , Antígenos CD34/análisis , Membrana Celular/química , Células Cultivadas , Citometría de Flujo , Expresión Génica , Glicosilfosfatidilinositoles/deficiencia , Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/genética , Humanos , Células K562 , Proteínas de la Membrana/análisis , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Fosfatidilinositol Diacilglicerol-Liasa , Proteínas PrPC/análisis , Proteínas PrPC/fisiología , Fosfolipasas de Tipo C/farmacología
14.
Eur J Haematol ; 65(1): 8-16, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10914934

RESUMEN

In paroxysmal nocturnal haemoglobinuria (PNH), somatic mutation of the PIG-A gene is thought to result in altered expression of glycosylphosphatidylinositol (GPI)-anchored proteins. This study was performed to determine if there were any heterogeneities of cellular phenotypes between two major peripheral blood cells, erythrocytes and granulocytes. Using CD59-based immunocytometry, the patterns of CD59 expression were shown to be conserved in the circulating erythroid cells (reticulocytes and mature erythrocytes) in all 29 patients with PNH. Twenty-one patients had distinct combinations of PNH type I, II, and III cells in different lineages. Only eight patients exhibited similar patterns of CD59 expression between the two lineages. Approximately one third of the patients had PNH type II cells in either or both of the two lineages indicating variable lineage involvement. The proportion of abnormal granulocytes was higher than those of abnormal reticulocytes and erythrocytes. In patients with appropriate erythropoietic responses to haemolysis (RPI > 2.0), shift reticulocytes display predominantly PNH phenotypes. These immature erythroid cells with altered expression of GPI-anchored proteins may dominate the peripheral blood during periods of increased marrow activity resulting in greater phenotypic mosaicism in such patients. Discrepancies in expression of GPI-anchored proteins in PNH which are highly variable between the two lineages may be the result of their different life spans and the influence of complement-mediated cytolysis. The phenomena also indicated the possible occurrence of more than one PNH clones with variable clonal dominance.


Asunto(s)
Médula Ósea/patología , Antígenos CD55/análisis , Antígenos CD59/análisis , Eritrocitos/patología , Granulocitos/patología , Hemoglobinuria Paroxística/patología , Reticulocitos/patología , Adolescente , Adulto , Anciano , Anemia Aplásica/etiología , Biomarcadores , Recuento de Células Sanguíneas , Linaje de la Célula , Células Clonales/patología , Eritrocitos/química , Femenino , Citometría de Flujo , Glicosilfosfatidilinositoles/deficiencia , Granulocitos/química , Enfermedades Hematológicas/patología , Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/complicaciones , Hemoglobinuria Paroxística/diagnóstico , Hemoglobinuria Paroxística/genética , Humanos , Inmunofenotipificación , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Fenotipo , Reticulocitos/química
15.
Rev. bioméd. (México) ; 10(2): 129-36, abr.-jun. 1999.
Artículo en Español | LILACS | ID: lil-254631

RESUMEN

La hemoglobinuria Paroxística Nocturna (HPN) fue descrita inicialmente por William Gull en 1866, en Londres Inglaterra y representa desde su descripción inicial un buen ejemplo de la evolución de los conceptos y conocimientos, en función del tiempo, en torno a una enfermedad. A William Gull le corresponde el mérito de haber descrito que el pigmento excretado en orina no correspondía a glóbulos rojos (GR). En 1882 Paul Strübing comunicó la siguiente descripción de la HPN. Strübing describió la asociación entre la hemoglobinuria y el ejercicio físico; propuso que la anormalidad residía en los GR, que al circular por los riñones sufrían hemólisis y describió la asociación entre la administración de hierro y las crisis de hemólisis. El nombre de HPN fue establecido en 1928 por Enneking. En 1911, Hijmans-van den Berg demostró que la acidificación de sueros normales o de pacientes con HPN, inducía lisis en los GR de pacientes con HPN. Sin embargo las observaciones de Thomas Hale Ham en 1937, que le permitieron proponer que el defecto de los GR en la HPN consistía en una mayor susceptibilidad a la lisis por el complemento (C'). Pangburn y col. y el grupo de Nicholson-Weller en 1983, describieron que en la HPN existe disminución cuantitativa del factor que acelera la degradación de las convertasas del C' fijadas a la membrana (DAF = "decay accelerating factor", o CD55). En 1987 y 1988, Zalman y col. y Blaas y cols., respectivamente, describen la deficiencia en esta células de la proteina reguladora de la fracción C-59, el inhibidor de membrana de la lisis reactiva o CD59. En 1992, Mahoney y col. y Hirose y su grupo demostraron que en la HPN la síntesis del glucosilfosfatidil inositol (PIG) era defectuosa, lo que en su turno impedía se anclaran las proteinas antes descritas. Estudios realizados por Takeda y col., en la Universidad de Osaka Japón, y publicados en 1993 permitieron clonar el gen PIG (gen PIG-A) e identificaron en la HPN una mutación somática que ocasionaba la pérdida de la función del gen PIG-A. En la actualidad se postula que la clona de HPN emerge como defensa a algún factor externo o interno que inhiba la hematopoyesis normal, pero incapaz de inhibir las células hematopoyéticas deficientes en las protéinas ancladas en el PIG


Asunto(s)
Humanos , Anemia Aplásica , Eritrocitos/química , Eritrocitos/citología , Glicosilfosfatidilinositoles , Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/fisiopatología , Hemoglobinuria Paroxística/genética , Hemoglobinuria Paroxística/historia , Hemólisis , Terminología
16.
Transfusion ; 38(4): 337-42, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9595015

RESUMEN

BACKGROUND: Complement-mediated lysis of red cells (RBCs) is a classic feature of paroxysmal nocturnal hemoglobinuria (PNH) that is traditionally studied with a combination of radiolabeling of RBCs and in vitro hemolysis tests. Phenotyping of reticulocytes was used as an alternative method for the evaluation of the relative life span of normal RBCs (PNH I) and RBCs that were partially (PNH II) or completely (PNH III) deficient in CD59. STUDY DESIGN AND METHODS: Murine monoclonal antibodies CD55, CD58, and CD59 and thiazole orange were used to phenotype reticulocytes by two-color flow cytometry in nine PNH patients. RBC survival could be calculated from the ratio of CD59- or CD59low mature RBCs to CD59- or CD59low reticulocytes obtained from these patients who had not received a transfusion. RESULTS: The life span of PNH III RBCs varied from about 17 to 60 days. PNH II reticulocytes were found in the four patients with PNH II RBCs. The life span of PNH II RBCs varied with their residual expression of CD59, and cells with 15 to 20 percent of the normal amount of CD59 were protected against in vivo hemolysis. CONCLUSION: Phenotyping of reticulocytes is a convenient and reliable tool for evaluating the relative survival of normal and PNH RBCs. PNH II and PNH III reticulocytes are phenotypically distinct, and some PNH II RBCs may be sensitive to complement-mediated lysis in vitro, but normally they are complement-resistant in vivo.


Asunto(s)
Envejecimiento Eritrocítico/fisiología , Hemoglobinuria Paroxística/sangre , Inmunofenotipificación , Reticulocitos/inmunología , Antígenos CD55/sangre , Antígenos CD58/sangre , Antígenos CD59/sangre , Eritrocitos/inmunología , Femenino , Citometría de Flujo , Hemoglobinuria Paroxística/clasificación , Humanos , Masculino
17.
Sangre (Barc) ; 42(3): 171-7, 1997 Jun.
Artículo en Español | MEDLINE | ID: mdl-9381257

RESUMEN

PURPOSE: To compare the clinical patterns and survival of young and adult (AP) versus paediatric (PP) patients with paroxysmal nocturnal haemoglobinuria (PNH). PATIENTS AND METHODS: The clinical records of 117 patients (82% AP, 18% PP) seen in four cities of the Mexican Republic were analysed, the clinical course and survival of both groups being compared. RESULTS: No sex difference was found in the two patient-groups: 51% and 52% males, 49% and 48% females in AP and PP, respectively. The onset of PNH had similar distribution for the two groups of patients: aplastic form, 45% in AP and 62% in PP; cytopenias, 24% in AP versus 27% in PP; haemolysis, 28% in AP and 9% in PP, and thrombosis, 3% in AP versus 0% in PP. The clinical features with significant difference were: anaemic+haemorrhagic syndrome (39 AP (40%) vs 14 PP (67%), p = 0.02), initial diagnosis of immunologic thrombocytopenic purpura (7 AP (7%) vs 7 PP (33%), p = 0.003), and death rate (17 AP (18%) vs 8 PP (38%), p = 0.04). The actuarial survival curves showed significant differences between both groups (p = 0.045, Cox-Mantel), with estimated 10-year survival of 81% for AP and 55% for PP, and 15-year survivals of 64% for AP and 55% for PP. CONCLUSIONS: Seemingly, PNH in paediatric age has poorer prognosis than in adults, which is associated to higher incidence of fatal haemorrhages due to thrombocytopenia.


Asunto(s)
Hemoglobinuria Paroxística/epidemiología , Enfermedad Aguda , Adolescente , Adulto , Anemia/etiología , Anemia Refractaria con Exceso de Blastos/etiología , Médula Ósea/patología , Niño , Preescolar , Diagnóstico Diferencial , Femenino , Hemoglobinuria Paroxística/clasificación , Hemoglobinuria Paroxística/complicaciones , Hemoglobinuria Paroxística/patología , Hemorragia/etiología , Humanos , Incidencia , Lactante , Leucemia Mieloide/etiología , Tablas de Vida , Masculino , México/epidemiología , Pronóstico , Púrpura Trombocitopénica Idiopática/diagnóstico , Estudios Retrospectivos , Análisis de Supervivencia , Trombosis/etiología
20.
Chin Med J (Engl) ; 106(5): 323-30, 1993 May.
Artículo en Inglés | MEDLINE | ID: mdl-7691484

RESUMEN

The surface phosphatidylinositol (PI)-linked proteins on membrane of paroxysmal nocturnal hemoglobinuria (PNH) erythrocytes (PNHE) was analysed by a flowcytometer (FACS 420). It was found that the loss of acetylcholinesterase (AchE) and decay accelerating factor (DAF), two PI-linked proteins, from cell membrane of PNHE was not synchronous. The hemolysis rates of DAF (-) and AchE (-) PNHE were much higher than that of mixed population in cobra-venom factor (CoF) lysis test. Intact PNHE remaining after CoF lysis had relatively lower immunofluorescent labeling rate of AchE on membrane in comparison with normal erythrocytes. It implied that this subpopulation, in spite of being insensitive to complement lysis, was still abnormal in terms of the amount of PI-linked protein on cell membrane. When these intact PNHE remaining after CoF lysis were incubated with activated polymorphonuclear leukocytes (PMN) for three hours, immunofluorescent labeling of AchE on PNHE was prominently decreased. This indicated that reactive oxidants released from activated PMN might injure PI-linked proteins.


Asunto(s)
Acetilcolinesterasa/metabolismo , Antígenos CD/metabolismo , Proteínas Inactivadoras de Complemento/deficiencia , Hemoglobinuria Paroxística/sangre , Glicoproteínas de Membrana/metabolismo , Acetilcolinesterasa/deficiencia , Proteínas Sanguíneas/deficiencia , Proteínas Sanguíneas/metabolismo , Antígenos CD55 , Proteínas Inactivadoras de Complemento/metabolismo , Membrana Eritrocítica/metabolismo , Citometría de Flujo , Hemoglobinuria Paroxística/clasificación , Humanos , Neutrófilos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...