Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Blood Cells Mol Dis ; 108: 102860, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38889660

ABSTRACT

Acquired aplastic anemia (AA) is a rare heterogeneous disorder characterized by pancytopenia and hypoplastic bone marrow. The incidence is 2-3 per million population per year in the Western world, but 3 times higher in East Asia. Survival in severe aplastic anemia (SAA) has improved significantly due to advances in hematopoietic stem cell transplantation (HSCT), immunosuppressive therapy, biologic agents, and supportive care. In SAA, HSCT from a matched sibling donor (MSD) is the first-line treatment. If a MSD is not available, options include immunosuppressive therapy (IST), matched unrelated donor, or haploidentical HSCT. The purpose of this guideline is to provide health care professionals with clear guidance on the diagnosis and management of pediatric patients with AA. A preliminary evidence-based document prepared by a group of pediatric hematologists of the Bone Marrow Failure Study Group of the Italian Association of Pediatric Hemato-Oncology (AIEOP) was discussed, modified and approved during a series of consensus conferences that started online during COVID 19 and continued in the following years, according to procedures previously validated by the AIEOP Board of Directors.

2.
Genes Immun ; 16(2): 151-61, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25569260

ABSTRACT

Lymphocyte apoptosis is mainly induced by either death receptor-dependent activation of caspase-8 or mitochondria-dependent activation of caspase-9. Mutations in caspase-8 lead to autoimmunity/lymphoproliferation and immunodeficiency. This work describes a heterozygous H237P mutation in caspase-9 that can lead to similar disorders. H237P mutation was detected in two patients: Pt1 with autoimmunity/lymphoproliferation, severe hypogammaglobulinemia and Pt2 with mild hypogammaglobulinemia and Burkitt lymphoma. Their lymphocytes displayed defective caspase-9 activity and decreased apoptotic and activation responses. Transfection experiments showed that mutant caspase-9 display defective enzyme and proapoptotic activities and a dominant-negative effect on wild-type caspase-9. Ex vivo analysis of the patients' lymphocytes and in vitro transfection experiments showed that the expression of mutant caspase-9 correlated with a downregulation of BAFFR (B-cell-activating factor belonging to the TNF family (BAFF) receptor) in B cells and ICOS (inducible T-cell costimulator) in T cells. Both patients carried a second inherited heterozygous mutation missing in the relatives carrying H237P: Pt1 in the transmembrane activator and CAML interactor (TACI) gene (S144X) and Pt2 in the perforin (PRF1) gene (N252S). Both mutations have been previously associated with immunodeficiencies in homozygosis or compound heterozygosis. Taken together, these data suggest that caspase-9 mutations may predispose to immunodeficiency by cooperating with other genetic factors, possibly by downregulating the expression of BAFFR and ICOS.


Subject(s)
B-Cell Activation Factor Receptor/biosynthesis , Caspase 9/genetics , Immunologic Deficiency Syndromes/genetics , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Lymphoproliferative Disorders/genetics , Mutation , Adolescent , Adult , Apoptosis/genetics , Apoptosis/immunology , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/immunology , Caspase 9/immunology , Down-Regulation , HEK293 Cells , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/immunology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/metabolism , Male , Pedigree
3.
Rev. esp. med. nucl. imagen mol. (Ed. impr.) ; 33(2): 99-102, mar.-abr. 2014. ilus
Article in English | IBECS | ID: ibc-120942

ABSTRACT

A young patient with undefined autoimmune lymphoproliferative syndrome (ALPS-U) and low back pain underwent a CT and MRI study that showed enhancing vertebral lesions, some pulmonary nodules and diffuse latero-cervical lymphadenopathy. A 18F-FDG-PET/CT scan showed many areas of intense 18F-FDG uptake in multiple vertebrae, in some ribs, in the sacrum, in the liver, in both lungs, in multiple lymph nodes spread in the cervical, thoracic and abdominal chains. A bone marrow biopsy showed a "lymphomatoid granulomatosis", a rare variant of B-cell non-Hodgkin lymphoma (NHL). After the treatment, the 18F-FDG-PET/CT scan showed a complete metabolic response (AU)


Un paciente joven con síndrome linfoproliferativo autoinmune indefinido (ALPS-U) y dolor lumbar se sometió a una tomografía computarizada y a una resonancia magnética, estudios que mostraron varias lesiones vertebrales, algunos nódulos pulmonares y adenopatías laterocervicales difusas. Una exploración 18F-FDG-PET/TC reveló áreas de captación intensa de 18F-FDG en múltiples vértebras, algunas costillas, sacro, hígado, ambos pulmones y en varios ganglios linfáticos repartidos en las cadenas cervicales, torácica y abdominal. La biopsia de médula ósea diagnosticó una “granulomatosis linfomatoide”, una variante poco frecuente de Linfoma no-Hodgkin de células B (LNH). Tras el tratamiento, la exploración 18F-FDG-PET/TC demostró una respuesta metabólica completa (AU)


Subject(s)
Humans , Male , Autoimmune Lymphoproliferative Syndrome , Lymphoma, Non-Hodgkin , Positron-Emission Tomography , Fluorodeoxyglucose F18 , Low Back Pain/etiology
4.
Rev Esp Med Nucl Imagen Mol ; 33(2): 99-102, 2014.
Article in English | MEDLINE | ID: mdl-23845452

ABSTRACT

A young patient with undefined autoimmune lymphoproliferative syndrome (ALPS-U) and low back pain underwent a CT and MRI study that showed enhancing vertebral lesions, some pulmonary nodules and diffuse latero-cervical lymphadenopathy. A (18)F-FDG-PET/CT scan showed many areas of intense (18)F-FDG uptake in multiple vertebrae, in some ribs, in the sacrum, in the liver, in both lungs, in multiple lymph nodes spread in the cervical, thoracic and abdominal chains. A bone marrow biopsy showed a "lymphomatoid granulomatosis", a rare variant of B-cell non-Hodgkin lymphoma (NHL). After the treatment, the (18)F-FDG-PET/CT scan showed a complete metabolic response.


Subject(s)
Autoimmune Lymphoproliferative Syndrome/diagnosis , Autoimmune Lymphoproliferative Syndrome/therapy , Fluorodeoxyglucose F18 , Lymphoma, Non-Hodgkin/diagnosis , Lymphoma, Non-Hodgkin/therapy , Multimodal Imaging , Positron-Emission Tomography , Radiopharmaceuticals , Tomography, X-Ray Computed , Adult , Humans , Male
5.
Blood Cancer J ; 3: e160, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24241400

ABSTRACT

To diagnose juvenile myelomonocytic leukemia (JMML) is sometimes challenging, because around 10% of patients lack molecular abnormalities affecting Ras-MAPK (mitogen-activated protein kinase) pathway and other diseases such as cytomegalovirus infection can mimic clinical signs of JMML. In order to validate a phospho-specific flow cytometry assay assessing phospho-signal transducer and activator of transcription factor 5 (p-STAT5) as a new diagnostic tool for JMML, we examined 22 samples from children with JMML and 47 controls. CD33+/CD34+ cells from 22 patients with JMML showed hyperphosphorylation of STAT5 induced by sub-saturating doses of granulocyte-macrophage colony-stimulating factor (GM-CSF). Using a training set of samples (11 JMML and 23 controls), we identified a threshold for p-STAT5-positive after stimulation with 0.1 ng/ml GM-CSF (17.17%) that discriminates JMML from controls. This threshold was validated in an independent series (11 JMML, 24 controls and 7 cases with diseases other than JMML) where we demonstrated that patients with JMML could be distinguished from other subjects with a sensitivity of 91% (confidence interval (CI) 59-100%) and a specificity of 87% (CI 70-96%). Positive and negative predictive values were 71% (CI 42-92%) and 96% (CI 82-100%), respectively. In conclusion, flow cytometric p-STAT5 profiling is a reliable diagnostic tool for identifying patients with JMML and can contribute to consistency of current diagnostic criteria.

6.
J Clin Pathol ; 59(4): 440-2, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16567474

ABSTRACT

BACKGROUND: Familial tumoral calcinosis (FTC) is a rare autosomal recessive disease characterised by the development of multiple calcified masses in periarticular soft tissues; GALNT3 gene mutations have recently been described in an African American and in a Druse Arab family with FTC. OBJECTIVE: To report the clinical and histological features caused by a new GALNT3 mutation in a white family. RESULTS: Homozygosity for the nonsense mutation Lys463X was found in both affected siblings, who displayed a classic phenotype, the male also having testicular microlithiasis. He is the first subject described with testicular microlithiasis in FTC. CONCLUSIONS: The high testicular expression of GALNT3 suggests that the gene alteration could act locally by causing deposition of calcium, and the testis may be an underestimated site of calcification in FTC. Autoimmune diseases are present in several members of the family. Although immune disorders have been described in FTC, autoimmunity does not segregate with the GALNT3 mutation in this family.


Subject(s)
Calcinosis/genetics , Codon, Nonsense , Lithiasis/genetics , N-Acetylgalactosaminyltransferases/genetics , Neoplasm Proteins/genetics , Testicular Diseases/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Calcinosis/metabolism , Calcinosis/pathology , Child , Humans , Italy , Lithiasis/metabolism , Lithiasis/pathology , Male , Pedigree , Testicular Diseases/ethnology , Testicular Diseases/metabolism , White People , Polypeptide N-acetylgalactosaminyltransferase
7.
Bone Marrow Transplant ; 35 Suppl 1: S89-92, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15812539

ABSTRACT

Aplastic anemia (AA) is a rare disease with a major autoimmune pathogenetic component. CTLA4 is a T-lymphocyte surface molecule involved in the maintenance of immune tolerance. Some polymorphisms associated with a reduced expression of CTLA4, and thus presumably with increased tendency to autoimmunity, have been associated with various autoimmune diseases. In this study, we evaluated the distribution of the low expression polymorphisms -318C > T and 49A > G of CTLA4 in a population of 67 patients with acquired AA and in 100 normal controls. There was no difference in the distribution of the tested polymorphism between patients and controls and, within the patient group, between those who responded to immunosuppression vs those who did not respond. This study indicates that the polymorphisms -318C > T and 49A > G of CTLA4 do not affect the risk of developing AA and do not influence the response to immunosuppression.


Subject(s)
Anemia, Aplastic/genetics , Antigens, Differentiation/genetics , Exons/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Adolescent , Adult , Antigens, CD , CTLA-4 Antigen , Case-Control Studies , Child , Child, Preschool , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Infant , Male , Middle Aged , Risk Factors , White People
8.
Clin Exp Immunol ; 133(3): 430-7, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12930371

ABSTRACT

Triggering of the Fas receptor induces T cell apoptosis and is involved in shutting-off the immune response. Inherited defects impairing Fas function cause the autoimmune lymphoproliferative syndrome, and may play a role in other autoimmune diseases. The aim of this work was to analyse the Fas function in paediatric patients with thyroid autoimmunities. We found that T cells from 24/28 patients with Graves' disease (GD) and 12/35 patients with Hashimoto's thyroiditis (HT) displayed defective Fas function. In HT, the defect was more frequent in patients requiring replacement therapy (11/20) than in those not requiring (1/15); moreover, in untreated HT the highest defect was displayed by patients with the highest levels of autoantibodies. Fas was always expressed at normal levels and no Fas mutations were detected. Analysis of the healthy parents of seven Fas-resistant patients showed that several of them were Fas-resistant, which suggests a genetic component. Fusion of Fas-resistant T cells with the Fas-sensitive HUT78 T cell line generated Fas-resistant hybrid cells, which suggests the presence of molecules exerting a dominant negative effect on Fas function. Analysis of Fas-induced activation of caspase-8 and -9 showed decreased activity of both caspases in HT, whereas activity of caspase-9 was increased and that of caspase-8 was decreased in GD. These data suggest that heterogeneous inherited defects impairing Fas function favour the development of thyroid autoimmunities.


Subject(s)
Graves Disease/immunology , T-Lymphocytes/immunology , Thyroiditis, Autoimmune/immunology , fas Receptor/immunology , Adolescent , Adult , Age of Onset , Apoptosis , Case-Control Studies , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Line , Child , Enzyme Activation , Female , Graves Disease/genetics , Humans , Hybridomas , Lymphocyte Activation , Male , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Thyroiditis, Autoimmune/genetics
9.
Clin Exp Rheumatol ; 21(3): 377-85, 2003.
Article in English | MEDLINE | ID: mdl-12846062

ABSTRACT

OBJECTIVE: A human lymphoproliferative syndrome characterized by a defect of the Fas-mediated apoptosis pathway in the absence of a fas gene mutation (Autoimmune Lymphoproliferative Disease) has recently been described and characterized by autoimmune phenomena. The aim of this study was to investigate the presence of antinuclear and antiphospholipid antibodies and to define their specificity in 5 pediatric patients with this syndrome. METHODS: Antinuclear antibodies were investigated by Western Blot and IIF performed under standard as well as apoptotic conditions. The fine specificity of antiphospholipid antibodies was dissected by an ELISA for anti-beta 2-glycoprotein I, anti-prothrombin, anti-annexin V and anti-protein S antibodies, and by immunostaining on thin layer chromatography plates for antiphospholipid molecule antibodies. RESULTS: This study showed that the autoantibodies found in these patients targeted a broad spectrum of nuclear antigens which undergo redistribution from the nucleus to the cytoplasm and plasma membrane during the course of the apoptotic process. This reactivity does not comprise known specificities such as anti-extractable nuclear antigens or anti-dsDNA. Antiphospholipid antibodies were also found in these sera. A further characterization of the antiphospholipid antibodies showed the presence of a heterogeneous response with antibodies directed to negatively-charged phospholipids and antibodies targeting coagulation-related proteins (beta 2-GPI, prothrombin, annexin V) which are considered relevant antigens in the antiphospholipid syndrome. CONCLUSIONS: These results suggest that lack of tolerance due to a defect of Fas-mediated apoptosis allows the survival of B and T clones involved in the antinuclear and antiphospholipid immune responses.


Subject(s)
Antibodies, Antinuclear/blood , Antibodies, Antiphospholipid/blood , Antiphospholipid Syndrome/immunology , Lymphoproliferative Disorders/immunology , Adolescent , Adult , Antibodies, Antinuclear/analysis , Antibodies, Antiphospholipid/analysis , Antibody Specificity , Antiphospholipid Syndrome/physiopathology , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Blotting, Western , Child , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique, Indirect , Humans , Lymphoproliferative Disorders/physiopathology , Male , Prognosis , Sampling Studies , Sensitivity and Specificity , Severity of Illness Index
10.
Br J Haematol ; 115(2): 334-40, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11703332

ABSTRACT

Hereditary hyperferritinaemia-cataract syndrome (HHCS) is an autosomal dominant disease caused by mutations in the iron responsive element (IRE) of the l-ferritin gene. Despite the elucidation of the genetic basis, the overall clinical spectrum of HHCS has been less well studied as, to date, only individual case reports have been described. Therefore, we studied a total of 62 patients in 14 unrelated families, with nine different mutations. No relevant symptoms other than visual impairment were found to be associated with the syndrome. A marked phenotypic variability was observed, particularly with regard to ocular involvement (i.e. age range at which cataract was diagnosed in 16 subjects with the C39T: 6-40 years). Similarly, serum ferritin levels varied substantially also within subjects sharing the same mutation (i.e. range for the A40G: 700-2412 microg/l). We followed an HHCS newborn in whom well-defined lens opacities were not detectable either at birth or at 1 year. The lens ferritin content was analysed in two subjects who underwent cataract surgery at different ages, with different cataract morphology. Values were similar and about 1500-fold higher than in controls. These observations suggest that: (i) in HHCS the cataract is not necessarily congenital; (ii) in addition to the IRE genotype, other genetic or environmental factors may modulate the phenotype, especially the severity of the cataract.


Subject(s)
Cataract/blood , Ferritins/blood , Adolescent , Adult , Age of Onset , Aged , Cataract/genetics , Child , Child, Preschool , Disease Progression , Female , Ferritins/genetics , Follow-Up Studies , Humans , Infant, Newborn , Iron-Regulatory Proteins , Iron-Sulfur Proteins/genetics , Male , Middle Aged , Mutation , Pedigree , RNA-Binding Proteins/genetics , Syndrome
11.
Lab Invest ; 81(7): 1025-37, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11454991

ABSTRACT

Flt-3 ligand (FL) is a cytokine that promotes the survival, proliferation, and differentiation of hematopoietic progenitors in synergy with other growth factors, such as stem cell factor. Previously we have demonstrated that stem cell factor and its receptor c-kit are expressed in neural crest-derived tumor cells and that a c-kit block induces their apoptosis. Here we have evaluated the expression of flt-3 and its ligand in 12 neuroectodermal tumor cell lines from neuroblastoma (NB), neuroepithelioma (NE), Ewing sarcoma (ES), and peripheral neuroectodermal tumor (PNET) and in 38 biopsies: 19 from NB and 19 from ES and PNET. RT-PCR demonstrated the expression of flt-3 and FL in all lines. Coexpression was observed in 42% of NB and in 74% of ES and PNET biopsies. Flow cytometry confirmed the presence of membrane and cytoplasmic flt-3 and membrane FL in all lines, whereas soluble FL protein was not measurable in their supernatants. Microphysiometric demonstration of acidification of the medium provided evidence of the specific response of cell lines to FL stimulation. Specific flt-3 phosphorylation after FL treatment was also demonstrated by Western blotting analysis. In cells growing in RPMI plus 1% fetal calf serum, FL revealed a significant proliferating activity, more evident in NB and NE lines (mean increase of viable cells, 73 +/- 26% after 1 day). Treatment with flt-3 antisense oligonucleotides significantly inhibited cell growth. FL also displayed an antiapoptotic activity: after a 12-hour culture in the presence of 0.1% fetal calf serum, FL caused a 50% reduction of apoptotic cells. These results provide further evidence that neuroectodermal and hematopoietic cells share common regulatory pathways, and could be of interest in the clinical management of neuroectodermal tumors.


Subject(s)
Cell Division , Cell Survival , Membrane Proteins/genetics , Nervous System Neoplasms/metabolism , Neural Crest/pathology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Base Sequence , Cell Line , DNA Primers , Flow Cytometry , Humans , Ligands , Membrane Proteins/metabolism , Nervous System Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , fms-Like Tyrosine Kinase 3
12.
Diabetes ; 50(3): 483-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11246866

ABSTRACT

Fas (CD95) triggers programmed cell death and is involved in cell-mediated cytotoxicity and in shutting off the immune response. Inherited loss-of-function mutations hitting the Fas system cause the autoimmune/lymphoproliferative syndrome (ALPS). We have recently shown that ALPS patients' families display increased frequency of common autoimmune diseases, including type 1 diabetes. This work evaluates Fas function in type 1 diabetic patients without typical ALPS. Cell death induced by anti-Fas monoclonal antibody was investigated in T-cells from 13 patients with type 1 diabetes alone and 19 patients with type 1 diabetes plus other autoimmune diseases (IDDM-P). Moreover, we analyzed 19 patients with thyroiditis alone (TYR), because most IDDM-P patients displayed thyroiditis. Frequency of resistance to Fas-induced cell death was significantly higher in patients with IDDM-P (73%) than in type 1 diabetic (23%) or TYR (16%) patients or in normal control subjects (3%). The defect was specific because resistance to methyl-prednisolone-induced cell death was not significantly increased in any group. Fas was always expressed at normal levels, and no Fas mutations were detected in four Fas-resistant IDDM-P patients. Analysis of the families of two Fas-resistant patients showing that several members were Fas-resistant suggests that the defect has a genetic component. Moreover, somatic fusion of T-cells from Fas-resistant subjects and the Fas-sensitive HUT78 cell line generates Fas-resistant hybrid cells, which suggests that the Fas resistance is due to molecules exerting a dominant-negative effect on a normal Fas system. These data suggest that Fas defects may be a genetic factor involved in the development of polyreactive type 1 diabetes.


Subject(s)
Autoimmune Diseases/complications , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/physiopathology , fas Receptor/physiology , Adolescent , Adult , Antibodies, Monoclonal/pharmacology , Cell Death/drug effects , Cell Death/physiology , Cell Line , Child , Diabetes Mellitus, Type 1/genetics , Drug Resistance , Female , Humans , Male , Mutation , Reference Values , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Thyroiditis/physiopathology , fas Receptor/analysis , fas Receptor/genetics , fas Receptor/immunology
13.
Blood Cells Mol Dis ; 26(5): 417-22, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11112378

ABSTRACT

Diamond-Blackfan anemia (DBA) is a congenital disease characterized by defective erythroid progenitor maturation and physical malformations. Most cases are sporadic, but dominant or, more rarely, recessive inheritance is observed in 10% of patients. Mutations in the gene encoding ribosomal protein (RP) S19 have recently been found in 25% of patients with either the dominant or the sporadic form. DBA is the first human disease due to mutations in a ribosomal structural protein. Families unlinked to this locus have also been reported. In an investigation of 23 individuals, we identified eight different mutations in 9 patients. These include five missense, one frameshift, one splice site defect, and one 4-bp insertion in the regulatory sequence. Seven mutations are new; one has so far been found in 8 patients and is a relatively common de novo event. Two mutations are predicted to generate a truncated protein. We also report the prevalence of RPS 19 mutations in the Italian DBA population, as shown by an analysis of 56 patients. No genotype-phenotype correlation was found between patients with the same mutation. The main clinical applications for molecular analysis are clinical diagnosis of patients with an incomplete form of DBA and testing of siblings of a patient with a severe form so as to avoid using those who carry a mutation and a silent phenotype as allogeneic stem cell donors.


Subject(s)
Fanconi Anemia/genetics , Ribosomal Proteins/genetics , Adult , Amino Acid Substitution , Base Sequence , Child , Cohort Studies , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Fanconi Anemia/pathology , Female , Genetic Heterogeneity , Genotype , Humans , Italy , Male , Mutagenesis, Insertional , Mutation , Phenotype , Point Mutation , Sequence Deletion
14.
Paediatr Drugs ; 2(5): 345-55, 2000.
Article in English | MEDLINE | ID: mdl-11022796

ABSTRACT

Diamond Blackfan Anaemia (DBA) is a congenital disease characterised by defective erythroid progenitor maturation. It is usually diagnosed during the first year of life. The main clinical sign is profound isolated normochromic or macrocytic anaemia, with normal numbers and function of the other haemopoietic cells. Reticulocyte counts in patients with DBA are very low. Bone marrow reflects the defective erythropoiesis, showing a very low number of erythropoietic precursors and a reduction of erythroid burst-forming unit progenitor cells. The proliferation and differentiation of the other lineages are normal. More than one-third of patients have malformations, most often involving the upper limbs and head, and the urogenital or cardiovascular systems. However, the link between these malformations and defective erythropoiesis is unclear and a defect in a molecule acting on both early embryonic development and haematopoiesis has been proposed. Whereas most cases are sporadic, inheritance is observed in 10% of patients, with a dominant or, more rarely, recessive pattern. One locus on chromosome 19q13.2 encoding ribosomal protein S19 accounts for a quarter of patients with either the dominant or the sporadic form. Families not linked with this locus have also been described. The diagnosis of DBA may be difficult and differential diagnoses include Fanconi's anaemia and acquired erythroid aplasias. Erythrocyte adenosine deaminase levels are generally high in DBA patients, which may help in the diagnosis, but they are not pathognomic. Corticosteroids are the main treatment option in DBA and these agents induce erythropoiesis in over 60% of patients. Some patients achieve complete remission, which may be either corticosteroid-induced or spontaneous. The increased in vitro erythropoiesis occasionally induced by the addition of specific cytokines, namely interleukin (IL)-3 and stem cell factor (SCF), has suggested their use in vivo. However, few patients have responded to IL-3, whereas SCF administration, though interesting in theory, has not yet been attempted. Patients who do not respond to corticosteroids and those who have to discontinue treatment because of adverse events must rely on long term transfusions, and are thus exposed to all of the associated complications. Bone marrow or cord blood transplantation has been performed in some patients. The former approach is burdened with severe complications and high mortality.


Subject(s)
Fanconi Anemia/diagnosis , Fanconi Anemia/therapy , Adrenal Cortex Hormones/therapeutic use , Blood Transfusion , Bone Marrow Transplantation , Fanconi Anemia/etiology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Infant , Interleukin-3/therapeutic use
15.
Clin Exp Immunol ; 121(1): 53-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10886239

ABSTRACT

To ascertain whether alterations of lymphocyte switching off may be associated with clustering of autoimmune diseases in children, Fas- and C2-ceramide-induced cell death was evaluated on T cell lines derived from three patients affected by clustering of autoimmune disorders. Three patterns were found: patient 3 was resistant to Fas- and C2-ceramide, patient 1 was resistant to Fas, but sensitive to C2-ceramide, patient 2 was resistant to C2-ceramide, but sensitive to Fas. By contrast, Fas- and C2-ceramide-induced cell death was normal in five children with systemic juvenile rheumatoid arthritis, five children with insulin-dependent diabetes and 10 age-matched healthy controls. Surface expression of Fas was low in patient 1, but normal in patients 2 and 3. Together with normal Fas transcripts, patients 2 and 3 displayed a transcript 152 bp longer than the normal one retaining intron 5. Our data indicate that polyreactive autoimmune syndromes may be associated with heterogeneous alteration of the immune response switching-off system.


Subject(s)
Apoptosis/immunology , Autoimmune Diseases/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Adolescent , Apoptosis/drug effects , Autoimmune Diseases/blood , Autoimmune Diseases/genetics , Cell Survival , Cells, Cultured , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Female , Humans , Immunophenotyping , Leukocytes, Mononuclear/classification , Leukocytes, Mononuclear/immunology , Male , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , T-Lymphocytes/drug effects , fas Receptor/genetics , fas Receptor/metabolism
16.
Blood ; 95(10): 3176-82, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10807785

ABSTRACT

Fas/Apo-1 (CD95) triggers programmed cell death (PCD) and is involved in immune response control and cell-mediated cytotoxicity. In the autoimmune/lymphoproliferative syndrome (ALPS), inherited loss-of-function mutations of the Fas gene cause nonmalignant lymphoproliferation and autoimmunity. We have recently identified an ALPS-like clinical pattern (named autoimmune lymphoproliferative disease [ALD]) in patients with decreased Fas function, but no Fas gene mutation. They also displayed decreased PCD response to ceramide, triggering a death pathway partially overlapping that used by Fas, which suggests that ALD is caused by downstream alterations of the Fas signaling pathway. Decreased Fas function is also involved in tumor development, because somatic mutations hitting the Fas system may protect neoplastic cells from immune surveillance. This work assessed the inherited component of the ALD defect by evaluating Fas- and ceramide-induced T-cell death in both parents and 4 close relatives of 10 unrelated patients with ALD. Most of them (22 of 24) displayed defective Fas- or ceramide-induced (or both) cell death. Moreover, analysis of the family histories showed that frequencies of autoimmunity and cancer were significantly increased in the paternal and maternal line, respectively. Defective Fas- or ceramide-induced T-cell death was also detected in 9 of 17 autoimmune patients from 7 families displaying more than a single case of autoimmunity within first- or second-degree relatives (multiple autoimmune syndrome [MAS] patients). Autoimmune diseases displayed by ALD and MAS families included several organ-specific and systemic forms. These data suggest that ALD is due to accumulation of several defects in the same subject and that these defects predispose to development of cancer or autoimmune diseases other than ALPS/ALD.


Subject(s)
Apoptosis/genetics , Autoimmune Diseases/genetics , Genetic Predisposition to Disease , Lymphoproliferative Disorders/genetics , fas Receptor/genetics , Adolescent , Adult , Aged , Autoimmune Diseases/etiology , Child , Child, Preschool , Female , Gene Expression Regulation , Humans , Lymphoproliferative Disorders/etiology , Male , Mutation , Neoplasms/etiology , Neoplasms/genetics , Signal Transduction/genetics
18.
Blood ; 95(4): 1195-8, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10666190

ABSTRACT

Severe congenital neutropenia (CN) (Kostmann syndrome) is a hematologic disorder characterized by a maturation arrest of myelopoiesis at the promyelocyte/myelocyte stage of development. This arrest results in severe neutropenia leading to absolute neutrophil counts (ANC) below 0.2 x 10(9)/L associated with severe bacterial infections from early infancy. Data on over 300 patients with CN collected by the Severe Chronic Neutropenia International Registry (SCNIR) beginning in 1994 indicate that more than 90% of these patients respond to recombinant human granulocyte-colony stimulating factor (r-HuG-CSF) treatment with an ANC greater than 1. 0 x 10(9)/L. For patients who are refractory to r-HuG-CSF treatment and continue to have severe and often life-threatening bacterial infections, hematopoietic stem cell transplantation is the only currently available treatment. We report on a total of 11 patients with CN reported to the SCNIR who underwent transplantation for reasons other than malignant transformation between 1976 and 1998. Of these patients, 8 were nonresponders or showed only partial response to r-HuG-CSF treatment with ongoing infections. Results from these patients suggest that transplantation of stem cells from an HLA-identical sibling is beneficial for patients refractory to r-HuG-CSF. (Blood. 2000;95:1195-1198)


Subject(s)
Hematopoietic Stem Cell Transplantation , Neutropenia/therapy , Adolescent , Child , Child, Preschool , Female , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Infant , Male , Neutropenia/congenital , Recombinant Proteins , Syndrome
19.
Blood ; 94(12): 4294-306, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10590074

ABSTRACT

Mutations of the ribosomal protein S19 (RPS19) gene were recently identified in 10 patients with Diamond Blackfan anemia (DBA). To determine the prevalence of mutations in this gene in DBA and to begin to define the molecular basis for the observed variable clinical phenotype of this disorder, the genomic sequence of the 6 exons and the 5' untranslated region of the RPS19 gene was directly assessed in DBA index cases from 172 new families. Mutations affecting the coding sequence of RPS19 or splice sites were found in 34 cases (19.7%), whereas mutations in noncoding regions were found in 8 patients (4.6%). Mutations included nonsense, missense, splice sites, and frameshift mutations. A hot spot for missense mutations was identified between codons 52 and 62 of the RPS19 gene in a new sequence consensus motif W-[YFW]-[YF]-x-R-[AT]-A-[SA]-x-[AL]-R-[HRK]-[ILV]-Y. No correlation between the nature of mutations and the different patterns of clinical expression, including age at presentation, presence of malformations, and therapeutic outcome, could be documented. Moreover, RPS19 mutations were also found in some first-degree relatives presenting only with isolated high erythrocyte adenosine deaminase activity and/or macrocytosis. The lack of a consistent relationship between the nature of the mutations and the clinical phenotype implies that yet unidentified factors modulate the phenotypic expression of the primary genetic defect in families with RPS19 mutations.


Subject(s)
Fanconi Anemia/genetics , Mutation , Ribosomal Proteins/genetics , Adolescent , Adult , Amino Acid Sequence , Base Sequence , Child, Preschool , Fanconi Anemia/physiopathology , Female , Humans , Infant , Male , Molecular Sequence Data , Pedigree
20.
AIDS Res Hum Retroviruses ; 15(14): 1255-63, 1999 Sep 20.
Article in English | MEDLINE | ID: mdl-10505674

ABSTRACT

CD4 cross-linking by HIV gp120 triggers CD4+ T cell death. Several authors have suggested that this effect is mediated by CD95, but this possibility is debated by other authors. In a previous work, we found by co-capping that gp120(451) and gp120MN, but not gp120(IIIB), induce lateral association of CD4 with CD95 on the T cell surface. In this work, we used fluorescence resonance energy transfer to confirm that CD4/CD95 lateral association is induced by gp120(451), but not gp120(IIIB). Moreover, we found that gp120 ability to induce the CD4/CD95 association correlates with ability to induce cell death, since gp120(451) and gp120MN induced higher levels of cell death than did gp120(IIIB) in PHA-derived CD4+ T cell lines. CD95 involvement in gp120-induced cell death was confirmed by showing that gp120(451) and gp120MN did not induce death in CD4+ T cells derived from patients with autoimmune/lymphoproliferative disease (ALD) and decreased CD95 function. Cell death induced by gp120MN was inhibited by a recombinant CD95/IgG.Fc molecule blocking the CD95/CD95L interaction. However, inhibition was late and only partial. These data suggest that the gp120-induced CD4/CD95 association exerts a dual effect: an early effect that is independent of CD95L and may be due to direct triggering of CD95 by gp120, and a late effect that may be due to sensitization of CD95 to triggering by CD95L. In line with the former effect, cell treatment with gp120MN activated caspase 3 in the presence of Fas/IgG.Fc, which shows that cell death induced by gp120MN independently of CD95L uses the same pathway as CD95.


Subject(s)
Apoptosis , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/physiology , HIV Envelope Protein gp120/metabolism , HIV-1/metabolism , Membrane Glycoproteins/metabolism , fas Receptor/metabolism , Apoptosis/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Caspase 3 , Caspases/metabolism , Cells, Cultured , Fas Ligand Protein , Humans , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...