Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 239
Filter
1.
Cell Death Dis ; 3: e356, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22833095

ABSTRACT

Diamond-Blackfan anemia (DBA) is caused by aberrant ribosomal biogenesis due to ribosomal protein (RP) gene mutations. To develop mechanistic understanding of DBA pathogenesis, we studied CD34⁺ cells from peripheral blood of DBA patients carrying RPL11 and RPS19 ribosomal gene mutations and determined their ability to undergo erythroid differentiation in vitro. RPS19 mutations induced a decrease in proliferation of progenitor cells, but the terminal erythroid differentiation was normal with little or no apoptosis. This phenotype was related to a G0/G1 cell cycle arrest associated with activation of the p53 pathway. In marked contrast, RPL11 mutations led to a dramatic decrease in progenitor cell proliferation and a delayed erythroid differentiation with a marked increase in apoptosis and G0/G1 cell cycle arrest with activation of p53. Infection of cord blood CD34⁺ cells with specific short hairpin (sh) RNAs against RPS19 or RPL11 recapitulated the two distinct phenotypes in concordance with findings from primary cells. In both cases, the phenotype has been reverted by shRNA p53 knockdown. These results show that p53 pathway activation has an important role in pathogenesis of DBA and can be independent of the RPL11 pathway. These findings shed new insights into the pathogenesis of DBA.


Subject(s)
Anemia, Diamond-Blackfan/metabolism , Erythroid Cells/metabolism , Ribosomal Proteins/genetics , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/pathology , Antigens, CD34/metabolism , Apoptosis , Cell Differentiation , Cell Proliferation , Cells, Cultured , Child, Preschool , Erythroid Cells/cytology , Female , G1 Phase Cell Cycle Checkpoints , Humans , Infant , Infant, Newborn , Male , Phenotype , RNA Interference , RNA, Small Interfering/metabolism , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
Arch Pediatr ; 18(9): 962-5, 2011 Sep.
Article in French | MEDLINE | ID: mdl-21803552

ABSTRACT

Human parvovirus B19 (HP-19) is the only Parvoviridae known to be pathogenic in human. Studies of HP-19 infection and its associated life-threatening complications in sickle cell anemia patients have been reported in Europe and the US. These results justify the development of HP-B19 prevention and strategies to reduce the incidence of severe and life-threatening complications associated with the infection in patients with sickle cell anemia, particularly in sub-Saharan Africa where the sickle cell anemia burden is high. In light of these considerations, we conducted a case-control study including 163 patients with sickle cell anemia and 163 controls. HP-B19 diagnosis was based on the detection of IgG and IgM antibodies specific for HP-B19 using commercially available enzyme immunoassays. Anti-human parvovirus B19 IgG antibodies were found in 105 of 193 (64.8%) patients vs 79 of 193 controls (48.4%). IgM antibodies were found at a higher frequency in sickle cell anemia patients than in controls. This higher frequency was found to be age-dependent. However, the reticulocyte count showed no significant decrease in Malian patients with sickle cell anemia. Further studies are needed to better characterize the implication of HP-B19 infection in sickle cell anemia mortality and morbidity and to develop preventive strategies and efficient management of the resulting complications.


Subject(s)
Anemia, Sickle Cell/complications , Parvoviridae Infections/diagnosis , Parvovirus B19, Human , Adolescent , Antibodies, Viral/blood , Case-Control Studies , Child , Child, Preschool , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Incidence , Infant , Infant, Newborn , Mali/epidemiology , Parvoviridae Infections/blood , Parvoviridae Infections/epidemiology , Parvoviridae Infections/prevention & control , Parvoviridae Infections/virology , Parvovirus B19, Human/immunology , Parvovirus B19, Human/isolation & purification
3.
Transfus Clin Biol ; 17(3): 112-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20655265

ABSTRACT

Diamond-Blackfan anemia is a rare inherited bone marrow failure syndrome (five to seven cases per million live births) characterized by an aregenerative, usually macrocytic anemia with an absence or less than 5% of erythroid precursors (erythroblastopenia) in an otherwise normal bone marrow. The platelet and the white cell counts are usually normal but neutropenia, thrombopenia or thrombocytosis have been noted at diagnosis. In 40 to 50% of DBA patients, congenital abnormalities mostly in the cephalic area and in thumbs and upper limbs have been described. Recent analysis did show a phenotype/genotype correlation. Congenital erythroblastopenia of DBA is the first human disease identified to result from defects in ribosomal biogenesis. The first ribosomal gene involved in DBA, ribosomal protein (RP) gene S19 (RPS19 gene), was identified in 1999. Subsequently, mutations in 12 other RP genes out of a total of 78 RP genes have been identified in DBA. All RP gene mutations described to date are heterozygous and dominant inheritance has been documented in 40 to 45% of affected individuals. As RP mutations are yet to be identified in approximately 50% of DBA cases, it is likely that other yet to be identified genes involved in ribosomal biogenesis or other pathways may be responsible for DBA phenotype.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Chromosomes, Human, Pair 10/genetics , Congenital Abnormalities/genetics , Translocation, Genetic , Anemia/etiology , Anemia/therapy , Anemia, Diamond-Blackfan/complications , Anemia, Diamond-Blackfan/epidemiology , Anemia, Diamond-Blackfan/pathology , Anemia, Macrocytic/etiology , Blood Transfusion , Bone Marrow/pathology , Congenital Abnormalities/pathology , Female , Fetal Growth Retardation/genetics , Humans , Incidence , Infant, Newborn , Mutation , Prednisone/therapeutic use , Pregnancy , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 28S/genetics , Ribosomal Proteins/genetics , Ribosomes/pathology
6.
Arch Pediatr ; 15(9): 1464-73, 2008 Sep.
Article in French | MEDLINE | ID: mdl-18556182

ABSTRACT

Hereditary spherocytosis (HS) is the commonest inherited disorder of the erythrocyte membrane in Northern Europe and North America. It is marked by a regenerative anemia which varies widely from asymptomatic patients to severe hemolysis. In 75% of HS patients, inheritance is autosomal dominant. The diagnosis of HS is easily made when there are a family history, hemolytic anemia, reticulocytosis, spherocytes and increased hyperdense cells. Specialized testing to clarify the nature of membrane disorder is required when the film appearance is atypical without a positive family history, in the absence of a family history, in the newborn and before the splenectomy, to rule out the stomatocytosis which is contraindicated. The indication for splenectomy is dependent on the degree of anemia and its clinical manifestation.


Subject(s)
Spherocytosis, Hereditary/diagnosis , Spherocytosis, Hereditary/therapy , Child , Cholecystectomy , Erythrocyte Membrane/physiology , Erythrocyte Transfusion , Erythropoietin , Humans , Recombinant Proteins , Spherocytosis, Hereditary/genetics , Splenectomy
7.
J Pediatr ; 152(1): 136-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18154916

ABSTRACT

DMT1 deficiency causes microcytic hypochromic anemia due to decreased erythroid iron utilization. Anemia is present from birth. Transferrin saturation is high and serum ferritin is mildly elevated, despite liver iron overload. DMT1 deficiency must be considered in the differential diagnosis of microcytic hypochromic anemia observed in the newborn period.


Subject(s)
Anemia, Hypochromic/genetics , Cation Transport Proteins/genetics , Genes, Recessive , Mutation , Anemia, Hypochromic/diagnosis , Anemia, Hypochromic/drug therapy , Cation Transport Proteins/deficiency , Diagnosis, Differential , Erythrocytes/metabolism , Erythropoietin/therapeutic use , Ferritins/blood , Genotype , Humans , Infant, Newborn , Iron Chelating Agents/therapeutic use , Phenotype , Transferrin/metabolism
8.
Transfus Clin Biol ; 14(4): 386-92, 2007 Oct.
Article in French | MEDLINE | ID: mdl-18035571

ABSTRACT

OBJECTIVE: Maternal and fetal risk is often high during pregnancy in sickle cell disease. Our objective was to evaluate the benefits of a transfusion program adapted to each pregnant patient, either by red cell transfusion or by automated red cell exchange, in sickle cell patients with a history of serious obstetrical and/or sickling complications. STUDY DESIGN: We managed 18 pregnancies in 14 patients (12 SS, 1 SC, 1 S/b-thalassemia), seven of whom had a history of one or more pregnancies, with severe maternofetal complications in nine out of 10 cases. The other seven patients were pregnant for the first time and were in care because of a history of severe sickling complications. The aim was to achieve a proportion of abnormal hemoglobin (hemoglobin S or S+C) below 50% and a hemoglobin level between 9 and 11 g/dL. The choice between transfusion and red cell exchange was made in the light of the hemoglobin level. Red cell exchange was done using a Fresenius Com. Tec blood cell separator. Patients had red cell exchange in 10 cases, and transfusions in five cases. In three cases, patients had successive transfusions and red cell exchange. RESULTS: No serious maternal complication was observed. No fetal or perinatal death occurred. In one case, delivery was induced at 36 weeks of gestation because of fetal distress and hypotrophy. CONCLUSIONS: Our study suggests that women with severe sickle cell disease, even if they have a serious obstetrical history, can carry their pregnancy to term, without major obstetric complications, through a combination of early management by a multidisciplinary team and a suitable policy of prophylactic transfusion or automated red cell exchange.


Subject(s)
Anemia, Sickle Cell/therapy , Erythrocyte Transfusion , Exchange Transfusion, Whole Blood , Pregnancy Complications, Hematologic/therapy , Adult , Automation , Female , Hemoglobin, Sickle/analysis , Humans , Pregnancy , Pregnancy Outcome , Retrospective Studies , Treatment Outcome
9.
Clin Lab Haematol ; 28(4): 270-4, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16898969

ABSTRACT

We report a patient in whom hepatosiderosis was diagnosed at the age of 55 years and who has since been treated by regular bleeding. The H63D mutation was found in the heterozygous state in the HFE gene. No mutation was recorded in the SLC11A3 gene (ferroportin). Hepatosiderosis did not seem primary, nevertheless its cause long remained elusive. Only 2 years ago did we find the responsible condition, a very mildly expressed form of dehydrated hereditary stomatocytosis (DHS). This genetic disease is a strongly iron-loading condition. Haemolysis was fully compensated. Kalaemia was slightly elevated, suggesting a pseudohyperkalaemia that may be associated with DHS. Osmotic gradient ektacytometry allowed to assess the diagnosis of DHS. The red cell monovalent Na+ and K+ concentrations were moderately elevated and reduced respectively. The temperature dependence of the ouabain + bumetanide-resistant K+ influx produced a shallow slope, above and parallel to the control curve. These features were consistent with the diagnosis of DHS. The pronounced hepatosiderosis contrasted with the mildly expressed DHS, and with the ferritinaemia that was slightly elevated, if at all, prior to bleeding. Bleeding caused ferritinaemia to decrease and hepatosiderosis to recede. The whole picture accounts for a misleading presentation of DHS, in which the primary condition long remained hidden behind one of its remotest complications, hepatosiderosis.


Subject(s)
Anemia, Hemolytic, Congenital/complications , Hemosiderosis/etiology , Histocompatibility Antigens Class I/genetics , Liver Diseases/etiology , Membrane Proteins/genetics , Aged , Anemia, Hemolytic, Congenital/genetics , Erythrocyte Membrane/genetics , Hemochromatosis Protein , Hemosiderosis/therapy , Humans , Liver Diseases/therapy , Male , Osmotic Fragility/genetics , Phlebotomy
10.
Arch Pediatr ; 13(4): 367-70, 2006 Apr.
Article in French | MEDLINE | ID: mdl-16524705

ABSTRACT

Defective apoptosis caused by mutations of the Fas gene can lead to an autoimmune lymphoproliferative syndrome (ALPS). The main autoimmune manifestations are haematological: hemolytic anemia, thrombocytopenia and neutropenia. We described 3 patients with ALPS presenting as a lymphoproliferative syndrome associated with a Coomb's negative autoimmune hemolytic anemia and dyserythropoiesis predominating on the more mature erythroblasts. Fas apoptosis deficiency was evidenced in the 3 patients by the demonstration of an increased number of CD4(-)CD8(-)TCRalphabeta(+) T cells, a decreased apoptotic response of activated T lymphocytes to anti-Apo 1-3 monoclonal antibody and the presence of a heterozygous mutation of the Fas receptor gene.


Subject(s)
Anemia, Dyserythropoietic, Congenital/genetics , Anemia, Hemolytic, Autoimmune/genetics , Apoptosis/genetics , fas Receptor/genetics , Child, Preschool , Hepatomegaly/genetics , Humans , Infant , Male , Mutation , Splenomegaly/genetics
11.
J Pediatr ; 147(5): 669-73, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16291361

ABSTRACT

OBJECTIVES: To study the frequency and risk factors of growth retardation (GR) in patients with Diamond-Blackfan anemia. STUDY DESIGN: A cross-sectional survey including the 95 patients followed by hematologists affiliated with the French Society of Pediatric Hematology and Immunology for whom growth data were available; 43 patients were transfusion dependent, 32 were steroid dependent, and 20 patients were off treatment. GR was defined as height below 2 SD. RESULTS: Growth retardation was observed in 29.5% (28) patients. The proportion of GR increased significantly with age (16% <10, 32% among 10 to 16, 47.6% among 17 to 25, 41.7% among >16 years) and was higher in on-treatment than in off-treatment patients (35% among transfusion-dependent, 37% among steroid-dependent vs 5% among off-treatment). GR was significantly linked to associated malformations (OR, 2.3 [1.1 to 8.0]; P = .02) and intrauterine growth retardation (OR, 6.0 [1.1 to 11.6]; P = .021). GR remained independently associated with age, malformations, and treatment in a logistic regression. CONCLUSIONS: Our study showed that the risk of GR increases with age and is associated with treatment dependence. This result addresses the question of the respective part, in the pathogenesis of GR, of the disease severity, illustrated by treatment dependence on the one hand and of the deleterious effects of long-term treatments on the other hand.


Subject(s)
Anemia, Diamond-Blackfan/epidemiology , Growth Disorders/epidemiology , Adolescent , Adult , Anemia, Diamond-Blackfan/therapy , Child , Cross-Sectional Studies , Female , France/epidemiology , Humans , Logistic Models , Male , Multivariate Analysis , Risk Factors
12.
Br J Haematol ; 127(1): 118-22, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15384986

ABSTRACT

The family of two siblings with severe hereditary spherocytosis was investigated. The decrease was evident on both the alpha- and the beta-chains. The parents were haematologically normal. The mother was heterozygous for the low-expression polymorphic allele alphaLEPRA. The father was heterozygous for a novel combination in which one allele showed the alpha-spectrin low expression polymorphic allele alphaLELY, while his other allele showed the alphaLELY polymorphism in cis with a G-->A substitution, named Bicêtre, found at the extreme 3' end of exon 51. This combination was designated alpha(LELY-Bicêtre). The children were compound heterozygotes for alleles alphaLEPRA and alpha(LELY-Bicêtre). Reverse transcription polymerase chain reaction detected only trace amounts of the mRNA coding for alpha(LELY-Bicêtre). Mutation is therefore an essentially null mutation with no functional protein product. The lack of disease in the alphaLELY/(LELY-Bicêtre) father compared with the marked haemolysis in the alphaLEPRA/alpha(LELY-Bicêtre) children showed that expression of allele alphaLELY is not low enough to expose null alpha-spectrin alleles on the other chromosome. Quantitative estimations from these findings suggest that, to evoke spherocytosis, it is necessary that alpha-spectrin expression must be reduced to less than 25% of normal, while a reduction to 8% is sufficient.


Subject(s)
Spectrin/genetics , Spherocytosis, Hereditary/genetics , Alleles , Female , Humans , Infant, Newborn , Male , Mutation , Pedigree , Spherocytosis, Hereditary/blood
13.
Blood ; 98(10): 2894-9, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698268

ABSTRACT

Spherocytic red cells with reduced membrane surface area are a feature of hereditary spherocytosis (HS) and some forms of autoimmune hemolytic anemia (AIHA). It is generally assumed that membrane loss in spherocytic red cells occurs during their sojourn in circulation. The structural basis for membrane loss in HS is improper assembly of membrane proteins, whereas in AIHA it is due to partial phagocytosis of circulating red cells by macrophages. A hypothesis was formed that these different mechanisms should lead to temporal differences in surface area loss during red cell genesis and during sojourn in circulation in these 2 spherocytic syndromes. It was proposed that cell surface loss could begin at the reticulocyte stage in HS, whereas surface area loss in AIHA involves only circulating mature red cells. The validity of this hypothesis was established by documenting differences in cellular features of reticulocytes in HS and AIHA. Using a novel technique to quantitate cell surface area, the decreased membrane surface area of both reticulocytes and mature red cells in HS compared with normal cells was documented. In contrast, in AIHA only mature red cells but not reticulocytes exhibited decreased membrane surface area. These data imply that surface area loss in HS, but not in AIHA, is already present at the circulating reticulocyte stage. These findings imply that loss of cell surface area is an early event during genesis of HS red cells and challenge the existing concepts that surface area loss in HS occurs predominantly during the sojourn of mature red cells in circulation.


Subject(s)
Anemia, Hemolytic, Autoimmune/blood , Autoimmune Diseases/blood , Cell Membrane/ultrastructure , Reticulocytes/ultrastructure , Spherocytes/ultrastructure , Spherocytosis, Hereditary/blood , Adolescent , Adult , Aged , Anemia, Hemolytic, Autoimmune/diagnosis , Autoimmune Diseases/diagnosis , Child , Child, Preschool , Coombs Test , Desiccation , Diagnosis, Differential , Erythrocyte Aging , Erythrocyte Deformability , Erythrocyte Indices , Erythrocyte Membrane/ultrastructure , Female , Hemoglobins/analysis , Humans , Macrophages/physiology , Male , Middle Aged , Phagocytosis , Reticulocytes/chemistry , Spherocytes/chemistry , Spherocytosis, Hereditary/diagnosis , Spherocytosis, Hereditary/surgery , Splenectomy , Surface Properties , Time Factors
14.
Eur J Obstet Gynecol Reprod Biol ; 99(1): 124-5, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11604201

ABSTRACT

Thrombocytopenia of the May-Hegglin anomaly type was diagnosed in a woman with no past history of bleeding diathesis, who had been followed during her three pregnancies. No abnormal bleeding occurred although no platelet transfusion was administered during the second and third cesarean sections. Routine platelets transfusion is unnecessary but platelets should be available for use if abnormal bleeding occurs.


Subject(s)
Blood Platelets/pathology , Delivery, Obstetric/methods , Pregnancy Complications, Hematologic/surgery , Thrombocytopenia/pathology , Adult , Cesarean Section , Female , Humans , Infant, Newborn , Platelet Transfusion , Pregnancy , Thrombocytopenia/complications
15.
Br J Haematol ; 114(4): 837-44, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11564072

ABSTRACT

Acquired haemophilia is a life-threatening disorder caused by circulating auto-antibodies that inhibit factor VIII coagulant activity (FBIII:C). Immunoadsorption on protein A sepharose (IA-PA) was performed in two bleeding patients with acquired haemophilia: we observed a dramatic and quick decrease in the anti-FVIII:C inhibitor titre leading to a normal, albeit transient, haemostatic status. In one case, IA-PA was the only procedure which succeeded in stopping massive haemorrhage. In the second case, IA-PA reinforced the haemostatic effect of recombinant activated factor VII by increasing the endogenous plasma factor VIII level. The efficacy of IA-PA was sustained with immunosuppressive treatment introduced, respectively, 10 and 15 d before the IA-PA procedures. Our experience with IA-PA suggests that this extracorporeal anti-FVIII:C removal procedure is a valuable therapeutic tool for acquired haemophilia and can alleviate life-threatening haemorrhages.


Subject(s)
Hemophilia A/therapy , Immunosorbent Techniques , Plasmapheresis/methods , Staphylococcal Protein A , Adult , Blood Loss, Surgical/prevention & control , Factor VIIa/therapeutic use , Hemophilia A/immunology , Humans , Immunoglobulin G/analysis , Male , Middle Aged , Peptic Ulcer Hemorrhage/therapy , Recombinant Proteins/therapeutic use
17.
Blood ; 97(7): 2145-50, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11264183

ABSTRACT

Diamond-Blackfan anemia (DBA) is a rare congenital hypoplastic anemia that usually presents early in infancy and is inherited in 10% to 20% of cases. Linkage analysis has shown that DBA in many of both dominant and recessive DBA families mapped to chromosome 19q13.2 leading to the cloning of a gene on chromosome 19q13.2 that encodes a ribosomal protein, RPS19. However, subsequently, mutations of the RPS19 gene have only been identified in 25% of all patients with DBA. This study analyzed 14 multiplex DBA families, 9 of which had 19q13.2 haplotypes inconsistent with 19q linkage. A genome-wide search for linked loci suggested the presence of a second DBA locus in a 26.4-centimorgan (cM) interval on human chromosome 8p. Subsequently, 24 additional DBA families were ascertained and all 38 families were analyzed with additional polymorphic markers on chromosome 8p. In total, 18 of 38 families were consistent with linkage to chromosome 8p with a maximal LOD score with heterogeneity of 3.55 at D8S277 assuming 90% penetrance. The results indicate the existence of a second DBA gene in the 26.4-cM telomeric region of human chromosome 8p23.3-p22, most likely within an 8.1-cM interval flanked by D8S518 and D8S1825. Seven families were inconsistent with linkage to 8p or 19q and did not reveal mutations in the RPS19 gene, suggesting further genetic heterogeneity. (Blood. 2001;97:2145-2150)


Subject(s)
Chromosomes, Human, Pair 8/genetics , Fanconi Anemia/genetics , Genetic Heterogeneity , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 8/ultrastructure , DNA Mutational Analysis , Female , Genetic Markers , Genetic Testing , Haplotypes/genetics , Humans , Lod Score , Male , Pedigree , Phenotype
18.
Blood ; 97(3): 826-8, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11157507

ABSTRACT

A new megathrombocytopenic syndrome with giant platelets in peripheral blood and severe thrombocytopenia was diagnosed in a 4-month-old boy. His clinical course included repeated hemorrhagic incidents leading to death at age 37 months. Bone marrow ultrastructural analysis revealed numerous dystrophic megakaryocytes with giant membrane complexes. Although these features were similar to those described for megakaryocytes in mice lacking the gene for transcription factor p45-NF-E2, no abnormalities in the p45-NF-E2 gene could be documented. Platelet membrane analysis showed a reduction in glycoprotein (GP) Ib, but normal content of GPIIb and GPIIIa. Analysis of genes encoding for GPIb alpha and beta, GPV, and GPIX ruled out the possibility that the observed platelet abnormality is a variant of Bernard-Soulier syndrome. A moderate neutropenia was associated with a complete lack of expression of sialyl-Lewis-X on the surface of polymorphonuclear neutrophils. A common defect in posttranslational modification of glycoproteins could account for the diverse cellular abnormalities.


Subject(s)
Megakaryocytes/ultrastructure , Neutropenia/diagnosis , Oligosaccharides/metabolism , Thrombocytopenia/diagnosis , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Humans , Infant , Leukocyte Count , Male , Neutropenia/pathology , Neutrophils/metabolism , Platelet Count , Platelet Membrane Glycoproteins/metabolism , Sialyl Lewis X Antigen , Syndrome , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
19.
Curr Opin Pediatr ; 13(1): 10-5, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11176237

ABSTRACT

Diamond-Blackfan Anemia (DBA) is a rare, congenital hypoplastic anemia often diagnosed early in infancy. A moderate to severe aregenerative anemia is found in association with erythroblastopenia in an otherwise normocellular bone marrow. In 40% of these infants with DBA, diverse developmental abnormalities are also noted. A majority of patients with DBA respond to steroid therapy. Recent molecular studies have identified mutations in the gene encoding the ribosomal protein RPS19 on chromosome 19 in 25% of patients with DBA. In another subset of patients, linkage analysis has identified another locus on chromosome 8p in association with DBA. There are, however, other cases of DBA that are linked neither to the RPS19 gene nor to the locus on 8p, implying the involvement of yet-to-be-defined genetic defects in the cause of DBA. The pathogenesis of DBA is still to be fully defined and it is anticipated that further molecular studies will lead to a better understanding of this complex disease.


Subject(s)
Fanconi Anemia , Abnormalities, Multiple , Child, Preschool , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 8/genetics , Failure to Thrive , Fanconi Anemia/diagnosis , Fanconi Anemia/epidemiology , Fanconi Anemia/genetics , Fanconi Anemia/physiopathology , Genetic Heterogeneity , Humans , Infant , Mutation
20.
Curr Opin Hematol ; 8(2): 68-73, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11224679

ABSTRACT

Immune-mediated disorders of erythropoiesis can result in acquired severe anemia, low reticulocyte counts, and bone marrow exhibiting pure red cell aplasia or ineffective erythropoiesis. Erythropoiesis can be suppressed or impaired by humoral or cellular mechanisms. In vitro inhibition of erythroid colony growth by immunoglobulins or lymphocytes can be a strong argument for the immune origin of the disease. Classical etiologies are thymoma and hematologic malignancies such as chronic lymphocytic leukemia (CLL). Clonal proliferation of T cells has been incriminated. Recently, acquired circulating autoantibodies directed against erythropoietin have been detected in a case of pure red cell aplasia. Autoimmune mechanisms have also been detected or suggested in synartesis and in Fas-associated dyserythropoiesis, two distinct syndromes recently described where morphologic abnormalities specific to the erythroid lineage illustrate ineffective erythropoiesis.


Subject(s)
Autoimmune Diseases/immunology , Erythropoiesis/immunology , Animals , Autoantibodies/immunology , Autoimmune Diseases/etiology , Humans , Red-Cell Aplasia, Pure/complications , Red-Cell Aplasia, Pure/immunology , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...