Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Hum Genet ; 110(11): 1938-1949, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37865086

ABSTRACT

Fanconi anemia (FA) is a clinically variable and genetically heterogeneous cancer-predisposing disorder representing the most common bone marrow failure syndrome. It is caused by inactivating predominantly biallelic mutations involving >20 genes encoding proteins with roles in the FA/BRCA DNA repair pathway. Molecular diagnosis of FA is challenging due to the wide spectrum of the contributing gene mutations and structural rearrangements. The assessment of chromosomal fragility after exposure to DNA cross-linking agents is generally required to definitively confirm diagnosis. We assessed peripheral blood genome-wide DNA methylation (DNAm) profiles in 25 subjects with molecularly confirmed clinical diagnosis of FA (FANCA complementation group) using Illumina's Infinium EPIC array. We identified 82 differentially methylated CpG sites that allow to distinguish subjects with FA from healthy individuals and subjects with other genetic disorders, defining an FA-specific DNAm signature. The episignature was validated using a second cohort of subjects with FA involving different complementation groups, documenting broader genetic sensitivity and demonstrating its specificity using the EpiSign Knowledge Database. The episignature properly classified DNA samples obtained from bone marrow aspirates, demonstrating robustness. Using the selected probes, we trained a machine-learning model able to classify EPIC DNAm profiles in molecularly unsolved cases. Finally, we show that the generated episignature includes CpG sites that do not undergo functional selective pressure, allowing diagnosis of FA in individuals with reverted phenotype due to gene conversion. These findings provide a tool to accelerate diagnostic testing in FA and broaden the clinical utility of DNAm profiling in the diagnostic setting.


Subject(s)
Fanconi Anemia , Humans , Fanconi Anemia/diagnosis , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , DNA Methylation/genetics , Proteins/genetics , DNA/metabolism
2.
Br J Haematol ; 204(6): 2453-2457, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38594875

ABSTRACT

ACTN1-related thrombocytopenia is a rare disorder caused by heterozygous variants in the ACTN1 gene characterized by macrothrombocytopenia and mild bleeding tendency. We describe for the first time two patients affected with ACTN1-RT caused by a homozygous variant in ACTN1 (c.982G>A) with mild heart valve defects unexplained by any other genetic variants investigated by WES. Within the reported family, the homozygous sisters have moderate thrombocytopenia and marked platelet macrocytosis with giant platelets, revealing a more severe haematological phenotype compared to their heterozygous relatives and highlighting a significant effect of allelic burden on platelet size. Moreover, we hypothesize that some ACTN1 variants, especially when present in the homozygous state, may also contribute to the cardiac abnormalities.


Subject(s)
Actinin , Homozygote , Phenotype , Thrombocytopenia , Humans , Thrombocytopenia/genetics , Actinin/genetics , Female , Male , Pedigree , Mutation , Adult
3.
Br J Haematol ; 205(1): 306-315, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38815995

ABSTRACT

Thrombocytopenia 4 (THC4) is an autosomal-dominant thrombocytopenia caused by mutations in CYCS, the gene encoding cytochrome c (CYCS), a small haeme protein essential for electron transport in mitochondria and cell apoptosis. THC4 is considered an extremely rare condition since only a few patients have been reported so far. These subjects presented mild thrombocytopenia and no or mild bleeding tendency. In this study, we describe six Italian families with five different heterozygous missense CYCS variants: p.Gly42Ser and p.Tyr49His previously associated with THC4, and three novel variants (p.Ala52Thr, p.Arg92Gly, and p.Leu99Val), which have been classified as pathogenic by bioinformatics and segregation analyses. Moreover, we supported functional effects of p.Ala52Thr and p.Arg92Gly on oxidative growth and respiratory activity in a yeast model. The clinical characterization of the 22 affected individuals, the largest series of THC4 patients ever reported, showed that this disorder is characterized by mild-to-moderate thrombocytopenia, normal platelet size, and function, low risk of bleeding, and no additional clinical phenotypes associated with reduced platelet count. Finally, we describe a significant correlation between the region of CYCS affected by mutations and the extent of thrombocytopenia, which could reflect different degrees of impairment of CYCS functions caused by different pathogenetic variants.


Subject(s)
Cytochromes c , Thrombocytopenia , Humans , Thrombocytopenia/genetics , Female , Male , Cytochromes c/genetics , Adult , Middle Aged , Pedigree , Mutation, Missense , Aged , Adolescent , Mutation , Young Adult , Child
4.
Br J Haematol ; 203(5): 852-859, 2023 12.
Article in English | MEDLINE | ID: mdl-37610030

ABSTRACT

MECOM-associated syndrome (MECOM-AS) is a rare disease characterized by amegakaryocytic thrombocytopenia, progressive bone marrow failure, pancytopenia and radioulnar synostosis with high penetrance. The clinical phenotype may also include finger malformations, cardiac and renal alterations, hearing loss, B-cell deficiency and predisposition to infections. The syndrome, usually diagnosed in the neonatal period because of severe thrombocytopenia, is caused by mutations in the MECOM gene, encoding for the transcription factor EVI1. The mechanism linking the alteration of EVI1 function and thrombocytopenia is poorly understood. In a paediatric patient affected by severe thrombocytopenia, we identified a novel variant of the MECOM gene (p.P634L), whose effect was tested on pAP-1 enhancer element and promoters of targeted genes showing that the mutation impairs the repressive activity of the transcription factor. Moreover, we demonstrated that EVI1 controls the transcriptional regulation of MPL, a gene whose mutations are responsible for congenital amegakaryocytic thrombocytopenia (CAMT), potentially explaining the partial overlap between MECOM-AS and CAMT.


Subject(s)
Pancytopenia , Thrombocytopenia , Infant, Newborn , Humans , Child , Pancytopenia/etiology , Transcription Factors/genetics , Thrombocytopenia/diagnosis , Bone Marrow Failure Disorders , Mutation , Receptors, Thrombopoietin/genetics , MDS1 and EVI1 Complex Locus Protein/genetics
5.
Haematologica ; 108(5): 1385-1393, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36226497

ABSTRACT

Congenital amegakaryocytic thrombocytopenia (CAMT) is a recessive disorder characterized by severe reduction of megakaryocytes and platelets at birth, which evolves toward bone marrow aplasia in childhood. CAMT is mostly caused by mutations in MPL (CAMT-MPL), the gene encoding the receptor of thrombopoietin (THPO), a crucial cytokine regulating hematopoiesis. CAMT can be also due to mutations affecting the THPO coding region (CAMT-THPO). In a child with the clinical picture of CAMT, we identified the homozygous c.-323C>T substitution, affecting a potential regulatory region of THPO. Although mechanisms controlling THPO transcription are not characterized, bioinformatics and in vitro analysis showed that c.-323C>T prevents the binding of transcription factors ETS1 and STAT4 to the putative THPO promoter, impairing THPO expression. Accordingly, in the proband the serum THPO concentration indicates defective THPO production. Based on these findings, the patient was treated with the THPO-mimetic agent eltrombopag, which induced a significant increase in platelet count and stable remission of bleeding symptoms. Herein, we report a novel pathogenic variant responsible for CAMT and provide new insights into the mechanisms regulating transcription of the THPO gene.


Subject(s)
Receptors, Thrombopoietin , Thrombopoietin , Child , Infant, Newborn , Humans , Thrombopoietin/pharmacology , Receptors, Thrombopoietin/genetics , Mutation , Megakaryocytes/pathology , Promoter Regions, Genetic , Proto-Oncogene Protein c-ets-1/genetics , STAT4 Transcription Factor/genetics
6.
Haematologica ; 108(7): 1909-1919, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-36519321

ABSTRACT

Inherited thrombocytopenias (IT) are genetic diseases characterized by low platelet count, sometimes associated with congenital defects or a predisposition to develop additional conditions. Next-generation sequencing has substantially improved our knowledge of IT, with more than 40 genes identified so far, but obtaining a molecular diagnosis remains a challenge especially for patients with non-syndromic forms, having no clinical or functional phenotypes that raise suspicion about specific genes. We performed exome sequencing (ES) in a cohort of 116 IT patients (89 families), still undiagnosed after a previously validated phenotype-driven diagnostic algorithm including a targeted analysis of suspected genes. ES achieved a diagnostic yield of 36%, with a gain of 16% over the diagnostic algorithm. This can be explained by genetic heterogeneity and unspecific genotype-phenotype relationships that make the simultaneous analysis of all the genes, enabled by ES, the most reasonable strategy. Furthermore, ES disentangled situations that had been puzzling because of atypical inheritance, sex-related effects or false negative laboratory results. Finally, ES-based copy number variant analysis disclosed an unexpectedly high prevalence of RUNX1 deletions, predisposing to hematologic malignancies. Our findings demonstrate that ES, including copy number variant analysis, can substantially contribute to the diagnosis of IT and can solve diagnostic problems that would otherwise remain a challenge.


Subject(s)
Genetic Testing , Thrombocytopenia , Humans , Exome Sequencing , Phenotype , Genetic Testing/methods , Genotype , Thrombocytopenia/diagnosis , Thrombocytopenia/genetics
7.
Haematologica ; 107(1): 260-267, 2022 01 01.
Article in English | MEDLINE | ID: mdl-33472357

ABSTRACT

GFI1B is a transcription factor essential for the regulation of erythropoiesis and megakaryopoiesis, and pathogenic variants have been associated with thrombocytopenia and bleeding. Analysing thrombocytopenic families by whole exome sequencing, we identified a novel GFI1B variant (c.648+5G>A), which causes exon 9 skipping and overexpression of a shorter p32 isoform. We report the clinical data of our patients and critically review the phenotype observed in individuals with different GFI1B variants leading to the same effect on the p32 expression. Since p32 is increased in acute and chronic leukemia cells, we tested the expression level of genes playing a role in various type of cancers, including hematological tumors and found that they are significantly dysregulated, suggesting a potential role for GFI1B in carcinogenesis regulation. Increasing the detection of individuals with GFI1B variants will allow us to better characterize this rare disease and determine whether it is associated with an increased risk of developing malignancies.


Subject(s)
Germ-Line Mutation , Thrombocytopenia , Carcinogenesis , Humans , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Repressor Proteins/genetics , Thrombocytopenia/genetics
8.
J Cell Physiol ; 236(8): 5664-5675, 2021 08.
Article in English | MEDLINE | ID: mdl-33432587

ABSTRACT

Warsaw breakage syndrome (WABS), is caused by biallelic mutations of DDX11, a gene coding a DNA helicase. We have recently reported two affected sisters, compound heterozygous for a missense (p.Leu836Pro) and a frameshift (p.Lys303Glufs*22) variant. By investigating the pathogenic mechanism, we demonstrate the inability of the DDX11 p.Leu836Pro mutant to unwind forked DNA substrates, while retaining DNA binding activity. We observed the accumulation of patient-derived cells at the G2/M phase and increased chromosomal fragmentation after mitomycin C treatment. The phenotype partially overlaps with features of the Fanconi anemia cells, which shows not only genomic instability but also defective mitochondria. This prompted us to examine mitochondrial functionality in WABS cells and revealed an altered aerobic metabolism. This opens the door to the further elucidation of the molecular and cellular basis of an impaired mitochondrial phenotype and sheds light on this fundamental process in cell physiology and the pathogenesis of these diseases.


Subject(s)
DNA Helicases/genetics , Fanconi Anemia/genetics , Genomic Instability/genetics , Kearns-Sayre Syndrome/metabolism , Mitochondrial Myopathies/metabolism , Abnormalities, Multiple/genetics , DEAD-box RNA Helicases/genetics , DNA Helicases/metabolism , Fanconi Anemia/metabolism , Genomics , Humans , Kearns-Sayre Syndrome/genetics , Mitochondrial Myopathies/genetics , Mutation/genetics
9.
Blood ; 133(12): 1346-1357, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30591527

ABSTRACT

Inherited thrombocytopenias (ITs) are a heterogeneous group of disorders characterized by low platelet count that may result in bleeding tendency. Despite progress being made in defining the genetic causes of ITs, nearly 50% of patients with familial thrombocytopenia are affected with forms of unknown origin. Here, through exome sequencing of 2 siblings with autosomal-recessive thrombocytopenia, we identified biallelic loss-of-function variants in PTPRJ . This gene encodes for a receptor-like PTP, PTPRJ (or CD148), which is expressed abundantly in platelets and megakaryocytes. Consistent with the predicted effects of the variants, both probands have an almost complete loss of PTPRJ at the messenger RNA and protein levels. To investigate the pathogenic role of PTPRJ deficiency in hematopoiesis in vivo, we carried out CRISPR/Cas9-mediated ablation of ptprja (the ortholog of human PTPRJ) in zebrafish, which induced a significantly decreased number of CD41+ thrombocytes in vivo. Moreover, megakaryocytes of our patients showed impaired maturation and profound defects in SDF1-driven migration and formation of proplatelets in vitro. Silencing of PTPRJ in a human megakaryocytic cell line reproduced the functional defects observed in patients' megakaryocytes. The disorder caused by PTPRJ mutations presented as a nonsyndromic thrombocytopenia characterized by spontaneous bleeding, small-sized platelets, and impaired platelet responses to the GPVI agonists collagen and convulxin. These platelet functional defects could be attributed to reduced activation of Src family kinases. Taken together, our data identify a new form of IT and highlight a hitherto unknown fundamental role for PTPRJ in platelet biogenesis.


Subject(s)
Blood Platelets/pathology , Genetic Predisposition to Disease , Megakaryocytes/pathology , Mutation , Thrombocytopenia/pathology , Adolescent , Adult , Animals , Blood Platelets/metabolism , CRISPR-Cas Systems , Child , Female , Follow-Up Studies , Hematopoiesis , Humans , Male , Megakaryocytes/metabolism , Middle Aged , Pedigree , Prognosis , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Thrombocytopenia/etiology , Thrombocytopenia/genetics , Zebrafish
10.
Int J Mol Sci ; 22(19)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34638529

ABSTRACT

Bernard-Soulier syndrome (BSS) is an autosomal-recessive bleeding disorder caused by biallelic variants in the GP1BA, GP1BB, and GP9 genes encoding the subunits GPIbα, GPIbß, and GPIX of the GPIb-IX complex. Pathogenic variants usually affect the extracellular or transmembrane domains of the receptor subunits. We investigated a family with BSS caused by the homozygous c.528_550del (p.Arg177Serfs*124) variant in GP1BB, which is the first mutation ever identified that affects the cytoplasmic domain of GPIbß. The loss of the intracytoplasmic tail of GPIbß results in a mild form of BSS, characterized by only a moderate reduction of the GPIb-IX complex expression and mild or absent bleeding tendency. The variant induces a decrease of the total platelet expression of GPIbß; however, all of the mutant subunit expressed in platelets is correctly assembled into the GPIb-IX complex in the plasma membrane, indicating that the cytoplasmic domain of GPIbß is not involved in assembly and trafficking of the GPIb-IX receptor. Finally, the c.528_550del mutation exerts a dominant effect and causes mild macrothrombocytopenia in heterozygous individuals, as also demonstrated by the investigation of a second unrelated pedigree. The study of this novel GP1BB variant provides new information on pathophysiology of BSS and the assembly mechanisms of the GPIb-IX receptor.


Subject(s)
Bernard-Soulier Syndrome/genetics , Platelet Glycoprotein GPIb-IX Complex/genetics , Thrombocytopenia/genetics , Adult , Bernard-Soulier Syndrome/blood , Bernard-Soulier Syndrome/pathology , Blood Platelets/physiology , Female , Humans , Male , Middle Aged , Pedigree , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Domains/genetics , Thrombocytopenia/pathology , von Willebrand Factor/metabolism
11.
Br J Haematol ; 183(2): 276-288, 2018 10.
Article in English | MEDLINE | ID: mdl-30351444

ABSTRACT

The inherited thrombocytopenias (IT) are a heterogeneous group of diseases resulting from mutations in more than 30 different genes. Among them, ACTN1-related thrombocytopenia (ACTN1-RT; Online Mendelian Inheritance in Man: 615193) is one of the most recently identified forms. It has been described as a mild autosomal dominant macrothrombocytopenia caused by mutations in ACTN1, a gene encoding for one of the two non-muscle isoforms of α-actinin. We recently identified seven new unrelated families with ACTN1-RT caused by different mutations. Two of them are novel missense variants (p.Trp128Cys and p.Pro233Leu), whose pathogenic role has been confirmed by in vitro studies. Together with the 10 families we have previously described, our cohort of ACTN1-RT now consists of 49 individuals carrying ACTN1 mutations. This is the largest case series ever collected and enabled a critical evaluation of the clinical aspects of the disease. We concluded that ACTN1-RT is the fourth most frequent form of IT worldwide and it is characterized by platelet macrocytosis in all affected subjects and mild thrombocytopenia in less than 80% of cases. The risk of bleeding, either spontaneous or upon haemostatic challenge, is negligible and there are no other associated defects, either congenital or acquired. Therefore, ACTN1-RT is a benign form of IT, whose diagnosis provides affected individuals and their families with a good prognosis.


Subject(s)
Actinin/genetics , Hematologic Diseases/genetics , Mutation , Thrombocytopenia/genetics , Adult , Aged , Blood Cell Count , Blood Platelets/pathology , Child , DNA Mutational Analysis/methods , Erythrocytes, Abnormal/pathology , Female , Hematologic Diseases/blood , Hematologic Diseases/pathology , Humans , Male , Middle Aged , Mutation, Missense , Pedigree , Platelet Aggregation , Thrombocytopenia/blood
12.
Haematologica ; 103(3): 417-426, 2018 03.
Article in English | MEDLINE | ID: mdl-29269525

ABSTRACT

Fanconi anemia is a rare disease characterized by congenital malformations, aplastic anemia, and predisposition to cancer. Despite the consolidated role of the Fanconi anemia proteins in DNA repair, their involvement in mitochondrial function is emerging. The purpose of this work was to assess whether the mitochondrial phenotype, independent of genomic integrity, could correlate with patient phenotype. We evaluated mitochondrial and clinical features of 11 affected individuals homozygous or compound heterozygous for p.His913Pro and p.Arg951Gln/Trp, the two residues of FANCA that are more frequently affected in our cohort of patients. Although p.His913Pro and p.Arg951Gln proteins are stably expressed in cytoplasm, they are unable to migrate in the nucleus, preventing cells from repairing DNA. In these cells, the electron transfer between respiring complex I-III is reduced and the ATP/AMP ratio is impaired with defective ATP production and AMP accumulation. These activities are intermediate between those observed in wild-type and FANCA-/- cells, suggesting that the variants at residues His913 and Arg951 are hypomorphic mutations. Consistent with these findings, the clinical phenotype of most of the patients carrying these mutations is mild. These data further support the recent finding that the Fanconi anemia proteins play a role in mitochondria, and open up possibilities for genotype/phenotype studies based on novel mitochondrial criteria.


Subject(s)
Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia/genetics , Mitochondria , Mutation, Missense , Adenosine Triphosphate/biosynthesis , Adolescent , Cell Nucleus/metabolism , Child , Child, Preschool , DNA Repair/genetics , Electron Transport , Fanconi Anemia Complementation Group A Protein/metabolism , Female , Humans , Loss of Function Mutation , Male , Phenotype
14.
Blood ; 125(5): 869-72, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25361813

ABSTRACT

Inherited thrombocytopenias (ITs) are a heterogeneous group of syndromic and nonsyndromic diseases caused by mutations affecting different genes. Alterations of ACTN1, the gene encoding for α-actinin 1, have recently been identified in a few families as being responsible for a mild form of IT (ACTN1-related thrombocytopenia; ACTN1-RT). To better characterize this disease, we screened ACTN1 in 128 probands and found 10 (8 novel) missense heterozygous variants in 11 families. Combining bioinformatics, segregation, and functional studies, we demonstrated that all but 1 amino acid substitution had deleterious effects. The clinical and laboratory findings of 31 affected individuals confirmed that ACTN1-RT is a mild macrothrombocytopenia with low risk for bleeding. Low reticulated platelet counts and only slightly increased serum thrombopoietin levels indicated that the latest phases of megakaryopoiesis were affected. Given its relatively high frequency in our cohort (4.2%), ACTN1-RT has to be taken into consideration in the differential diagnosis of ITs.


Subject(s)
Actinin/genetics , Blood Platelets/metabolism , Mutation, Missense , Phenotype , Thrombocytopenia/genetics , Actinin/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Blood Platelets/pathology , Case-Control Studies , Child , Child, Preschool , Female , Gene Expression , Genotype , Heterozygote , Humans , Male , Middle Aged , Pedigree , Platelet Count , Severity of Illness Index , Thrombocytopenia/metabolism , Thrombocytopenia/pathology , Thrombocytopenia/physiopathology , Thrombopoiesis/genetics , Thrombopoietin/blood
17.
Curr Opin Hematol ; 23(5): 486-92, 2016 09.
Article in English | MEDLINE | ID: mdl-27438527

ABSTRACT

PURPOSE OF REVIEW: Inherited thrombocytopenias are a heterogeneous group of diseases caused by mutations in many genes. They account for approximately only 50% of cases, suggesting that novel genes have yet to be identified for a comprehensive understanding of platelet biogenesis defects. This review provides an update of the last year of discoveries on inherited thrombocytopenias focusing on the molecular basis and potential pathogenic mechanisms affecting megakaryopoiesis and platelet production. RECENT FINDINGS: Most of the novel discoveries are related to identification of mutations in novel inherited thrombocytopenia genes using a next-generation sequencing approach. They include MECOM, DIAPH1, TRPM7, SRC, FYB, and SLFN14, playing different roles in megakaryopoiesis and platelet production. Moreover, it is worth mentioning data on hypomorphic mutations of FLI1 and the association of single nucleotide polymorphisms, such as that identified in ACTN1, with thrombocytopenia. SUMMARY: Thanks to the application of next-generation sequencing, the number of inherited thrombocytopenia genes is going to increase rapidly. Considering the wide genetic heterogeneity (more than 30 genes), these technologies can also be used for diagnostic purpose. Whatever is the aim, extreme caution should be taken in interpreting data, as inherited thrombocytopenias are mainly autosomal dominant diseases caused by variants of apparent unknown significance.


Subject(s)
Genetic Predisposition to Disease , Thrombocytopenia/genetics , Animals , Cytoskeleton/genetics , Cytoskeleton/metabolism , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Humans , Inheritance Patterns , Mutation , Signal Transduction , Thrombocytopenia/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Blood ; 124(6): e4-e10, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-24990887

ABSTRACT

Abnormalities of platelet size are one of the distinguishing features of inherited thrombocytopenias (ITs), and evaluation of blood films is recommended as an essential step for differential diagnosis of these disorders. Nevertheless, what we presently know about this subject is derived mainly from anecdotal evidence. To improve knowledge in this field, we evaluated platelet size on blood films obtained from 376 patients with all 19 forms of IT identified so far and found that these conditions differ not only in mean platelet diameter, but also in platelet diameter distribution width and the percentage of platelets with increased or reduced diameters. On the basis of these findings, we propose a new classification of ITs according to platelet size. It distinguishes forms with giant platelets, with large platelets, with normal or slightly increased platelet size, and with normal or slightly decreased platelet size. We also measured platelet diameters in 87 patients with immune thrombocytopenia and identified cutoff values for mean platelet diameter and the percentage of platelets with increased or reduced size that have good diagnostic accuracy in differentiating ITs with giant platelets and with normal or slightly decreased platelet size from immune thrombocytopenia and all other forms of IT.


Subject(s)
Blood Platelets/pathology , Thrombocytopenia/blood , Thrombocytopenia/genetics , Adolescent , Adult , Case-Control Studies , Cell Size , Child , Child, Preschool , Diagnosis, Differential , Female , Hearing Loss, Sensorineural/blood , Hearing Loss, Sensorineural/classification , Hearing Loss, Sensorineural/genetics , Humans , Infant , Male , Middle Aged , Molecular Motor Proteins/genetics , Mutation , Myosin Heavy Chains/genetics , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/diagnosis , Thrombocytopenia/classification , Thrombocytopenia/congenital , Young Adult
19.
Haematologica ; 101(11): 1333-1342, 2016 11.
Article in English | MEDLINE | ID: mdl-27365488

ABSTRACT

ETV6-related thrombocytopenia is an autosomal dominant thrombocytopenia that has been recently identified in a few families and has been suspected to predispose to hematologic malignancies. To gain further information on this disorder, we searched for ETV6 mutations in the 130 families with inherited thrombocytopenia of unknown origin from our cohort of 274 consecutive pedigrees with familial thrombocytopenia. We identified 20 patients with ETV6-related thrombocytopenia from seven pedigrees. They have five different ETV6 variants, including three novel mutations affecting the highly conserved E26 transformation-specific domain. The relative frequency of ETV6-related thrombocytopenia was 2.6% in the whole case series and 4.6% among the families with known forms of inherited thrombocytopenia. The degree of thrombocytopenia and bleeding tendency of the patients with ETV6-related thrombocytopenia were mild, but four subjects developed B-cell acute lymphoblastic leukemia during childhood, resulting in a significantly higher incidence of this condition compared to that in the general population. Clinical and laboratory findings did not identify any particular defects that could lead to the suspicion of this disorder from the routine diagnostic workup. However, at variance with most inherited thrombocytopenias, platelets were not enlarged. In vitro studies revealed that the maturation of the patients' megakaryocytes was defective and that the patients have impaired proplatelet formation. Moreover, platelets from patients with ETV6-related thrombocytopenia have reduced ability to spread on fibrinogen. Since the dominant thrombocytopenias due to mutations in RUNX1 and ANKRD26 are also characterized by normal platelet size and predispose to hematologic malignancies, we suggest that screening for ETV6, RUNX1 and ANKRD26 mutations should be performed in all subjects with autosomal dominant thrombocytopenia and normal platelet size.


Subject(s)
Genetic Predisposition to Disease/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Thrombocytopenia/genetics , Adolescent , Adult , Cell Transformation, Neoplastic/genetics , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit/genetics , Family , Humans , Infant , Infant, Newborn , Intercellular Signaling Peptides and Proteins , Middle Aged , Mutation , Nuclear Proteins/genetics , Pedigree , Thrombocytopenia/pathology , Young Adult , ETS Translocation Variant 6 Protein
20.
Am J Hematol ; 91(7): 666-71, 2016 07.
Article in English | MEDLINE | ID: mdl-27013026

ABSTRACT

We analyzed 97 Fanconi anemia patients from a clinic/biological database for genotype, somatic, and hematologic phenotype, adverse hematological events, solid tumors, and treatment. Seventy-two patients belonged to complementation group A. Eighty percent of patients presented with mild/moderate somatic phenotype and most with cytopenia. No correlation was seen between somatic/hematologic phenotype and number of missense mutations of FANCA alleles. Over follow-up, 33% of patients improved or maintained mild/moderate cytopenia or normal blood count, whereas remaining worsened cytopenia. Eleven patients developed a hematological adverse event (MDS, AML, pathological cytogenetics) and three developed solid tumors. 10 years cumulative risk of death of the whole cohort was 25.6% with median follow-up 5.8 years. In patients eligible to hematopoietic stem cell transplantation because of moderate cytopenia, mortality was significantly higher in subjects transplanted from matched unrelated donor over nontransplanted subjects, whereas there was no significant difference between matched sibling donor transplants and nontransplanted patients. In patients eligible to transplant because of severe cytopenia and clonal disease, mortality risk was not significantly different in transplanted from matched unrelated versus matched sibling donor versus nontransplanted subjects. The decision to transplant should rely on various elements including, type of donor, HLA matching, patient comorbidities, impairment, and clonal evolution of hematopoiesis. Am. J. Hematol. 91:666-671, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Fanconi Anemia/therapy , Hematopoietic Stem Cell Transplantation/methods , Adolescent , Adult , Child , Child, Preschool , Decision Making , Fanconi Anemia/mortality , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/mortality , Histocompatibility Testing , Humans , Infant , Infant, Newborn , Italy , Male , Pancytopenia/chemically induced , Phenotype , Siblings , Tissue Donors , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL