Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Br J Haematol ; 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38815995

RESUMEN

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.

2.
Br J Haematol ; 204(6): 2453-2457, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38594875

RESUMEN

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.


Asunto(s)
Actinina , Homocigoto , Fenotipo , Trombocitopenia , Humanos , Trombocitopenia/genética , Actinina/genética , Femenino , Masculino , Linaje , Mutación , Adulto
3.
Haematologica ; 108(7): 1909-1919, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-36519321

RESUMEN

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.


Asunto(s)
Pruebas Genéticas , Trombocitopenia , Humanos , Secuenciación del Exoma , Fenotipo , Pruebas Genéticas/métodos , Genotipo , Trombocitopenia/diagnóstico , Trombocitopenia/genética
5.
Int J Mol Sci ; 22(19)2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34638529

RESUMEN

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.


Asunto(s)
Síndrome de Bernard-Soulier/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Trombocitopenia/genética , Adulto , Síndrome de Bernard-Soulier/sangre , Síndrome de Bernard-Soulier/patología , Plaquetas/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Linaje , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Dominios Proteicos/genética , Trombocitopenia/patología , Factor de von Willebrand/metabolismo
6.
Elife ; 102021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34059198

RESUMEN

Thrombocytopenic disorders have been treated with the Thrombopoietin-receptor agonist Eltrombopag. Patients with the same apparent form of thrombocytopenia may respond differently to the treatment. We describe a miniaturized bone marrow tissue model that provides a screening bioreactor for personalized, pre-treatment response prediction to Eltrombopag for individual patients. Using silk fibroin, a 3D bone marrow niche was developed that reproduces platelet biogenesis. Hematopoietic progenitors were isolated from a small amount of peripheral blood of patients with mutations in ANKRD26 and MYH9 genes, who had previously received Eltrombopag. The ex vivo response was strongly correlated with the in vivo platelet response. Induced Pluripotent Stem Cells (iPSCs) from one patient with mutated MYH9 differentiated into functional megakaryocytes that responded to Eltrombopag. Combining patient-derived cells and iPSCs with the 3D bone marrow model technology allows having a reproducible system for studying drug mechanisms and for individualized, pre-treatment selection of effective therapy in Inherited Thrombocytopenias.


Platelets are tiny cell fragments essential for blood to clot. They are created and released into the bloodstream by megakaryocytes, giant cells that live in the bone marrow. In certain genetic diseases, such as Inherited Thrombocytopenia, the bone marrow fails to produce enough platelets: this leaves patients extremely susceptible to bruising, bleeding, and poor clotting after an injury or surgery. Certain patients with Inherited Thrombocytopenia respond well to treatments designed to boost platelet production, but others do not. Why these differences exist could be investigated by designing new test systems that recreate the form and function of bone marrow in the laboratory. However, it is challenging to build the complex and poorly understood bone marrow environment outside of the body. Here, Di Buduo et al. have developed an artificial three-dimensional miniature organ bioreactor system that recreates the key features of bone marrow. In this system, megakaryocytes were grown from patient blood samples, and hooked up to a tissue scaffold made of silk. The cells were able to grow as if they were in their normal environment, and they could shed platelets into an artificial bloodstream. After treating megakaryocytes with drugs to stimulate platelet production, Di Buduo et al. found that the number of platelets recovered from the bioreactor could accurately predict which patients would respond to these drugs in the clinic. This new test system enables researchers to predict how a patient will respond to treatment, and to tailor therapy options to each individual. This technology could also be used to test new drugs for Inherited Thrombocytopenias and other blood-related diseases; if scaled-up, it could also, one day, generate large quantities of lab-grown blood cells for transfusion.


Asunto(s)
Benzoatos/farmacología , Plaquetas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Hidrazinas/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Megacariocitos/efectos de los fármacos , Pirazoles/farmacología , Receptores de Trombopoyetina/agonistas , Nicho de Células Madre , Trombocitopenia/tratamiento farmacológico , Trombopoyesis/efectos de los fármacos , Adulto , Anciano , Reactores Biológicos , Plaquetas/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Femenino , Fibroínas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Megacariocitos/metabolismo , Persona de Mediana Edad , Miniaturización , Mutación , Cadenas Pesadas de Miosina/genética , Receptores de Trombopoyetina/metabolismo , Trombocitopenia/sangre , Trombocitopenia/genética , Adulto Joven
9.
Haematologica ; 105(3): 820-828, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31273088

RESUMEN

Patients with inherited thrombocytopenias often require platelet transfusions to raise their platelet count before surgery or other invasive procedures; moreover, subjects with clinically significant spontaneous bleeding may benefit from an enduring improvement of thrombocytopenia. The hypothesis that thrombopoietin-mimetics can increase platelet count in inherited thrombocytopenias is appealing, but evidence is scarce. We conducted a prospective, phase II clinical trial to investigate the efficacy of the oral thrombopoietin-mimetic eltrombopag in different forms of inherited thrombocytopenia. We enrolled 24 patients affected by MYH9-related disease, ANKRD26-related thrombocytopenia, X-linked thrombocytopenia/ Wiskott-Aldrich syndrome, monoallelic Bernard-Soulier syndrome, or ITGB3-related thrombocytopenia. The average pre-treatment platelet count was 40.4 ×109/L. Patients received a 3- to 6-week course of eltrombopag in a dose-escalated manner. Of 23 patients evaluable for response, 11 (47.8%) achieved a major response (platelet count >100 ×109/L), ten (43.5%) had a minor response (platelet count at least twice the baseline value), and two patients (8.7%) did not respond. The average increase of platelet count compared to baseline was 64.5 ×109/L (P<0.001). Four patients with clinically significant spontaneous bleeding entered a program of long-term eltrombopag administration (16 additional weeks): all of them obtained remission of mucosal hemorrhages, with the remission persisting throughout the treatment period. Treatment was globally well tolerated: five patients reported mild adverse events and one patient a moderate adverse event. In conclusion, eltrombopag was safe and effective in increasing platelet count and reducing bleeding symptoms in different forms of inherited thrombocytopenia. Despite these encouraging results, caution is recommended when using thrombopoietinmimetics in inherited thrombocytopenias predisposing to leukemia. ClinicalTrials.gov identifier: NCT02422394.


Asunto(s)
Hidrazinas , Trombocitopenia , Benzoatos/efectos adversos , Humanos , Hidrazinas/efectos adversos , Estudios Prospectivos , Pirazoles , Trombocitopenia/tratamiento farmacológico
11.
Hamostaseologie ; 39(1): 87-94, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29996171

RESUMEN

MYH9-related disease (MYH9-RD) is an autosomal-dominant thrombocytopenia caused by mutations in the gene for non-muscle myosin heavy chain IIA (NMMHC-IIA). Patients present congenital macrothrombocytopenia and inclusions of NMMHC-IIA in leukocytes, and have a variable risk of developing kidney damage, sensorineural deafness, presenile cataracts and/or liver enzymes abnormalities. The spectrum of mutations found in MYH9-RD patients is limited and the incidence and severity of the non-congenital features are predicted by the causative MYH9 variant. In particular, different alterations of the C-terminal tail domain of NMMHC-IIA associate with remarkably different disease evolution. We report four novel MYH9 mutations affecting the tail domain of NMMHC-IIA and responsible for MYH9-RD in four families. Two variants cause amino acid substitutions in the coiled-coil region of NMMHC-IIA, while the other two are a splicing variant and a single nucleotide deletion both resulting in frameshift alterations of the short non-helical tailpiece. Characterization of phenotypes of affected individuals shows that all of these novel variants are associated with a mild clinical evolution of the disease.


Asunto(s)
Trastornos de los Cromosomas/genética , Proteínas Motoras Moleculares/genética , Mutación , Cadenas Pesadas de Miosina/genética , Trombocitopenia/congénito , Adolescente , Adulto , Anciano , Sustitución de Aminoácidos , Rotura Cromosómica , Trastornos de los Cromosomas/patología , Femenino , Mutación del Sistema de Lectura , Humanos , Masculino , Persona de Mediana Edad , Proteínas Motoras Moleculares/química , Cadenas Pesadas de Miosina/química , Linaje , Fenotipo , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Trombocitopenia/genética , Trombocitopenia/patología , Adulto Joven
12.
Blood ; 133(12): 1346-1357, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30591527

RESUMEN

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.


Asunto(s)
Plaquetas/patología , Predisposición Genética a la Enfermedad , Megacariocitos/patología , Mutación , Trombocitopenia/patología , Adolescente , Adulto , Animales , Plaquetas/metabolismo , Sistemas CRISPR-Cas , Niño , Femenino , Estudios de Seguimiento , Hematopoyesis , Humanos , Masculino , Megacariocitos/metabolismo , Persona de Mediana Edad , Linaje , Pronóstico , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Trombocitopenia/etiología , Trombocitopenia/genética , Pez Cebra
13.
EMBO Mol Med ; 10(1): 63-75, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29191945

RESUMEN

Congenital amegakaryocytic thrombocytopenia (CAMT) is an inherited disorder characterized at birth by thrombocytopenia with reduced megakaryocytes, which evolves into generalized bone marrow aplasia during childhood. Although CAMT is genetically heterogeneous, mutations of MPL, the gene encoding for the receptor of thrombopoietin (THPO), are the only known disease-causing alterations. We identified a family with three children affected with CAMT caused by a homozygous mutation (p.R119C) of the THPO gene. Functional studies showed that p.R119C affects not only ability of the cytokine to stimulate MPL but also its release, which is consistent with the relatively low serum THPO levels measured in patients. In all the three affected children, treatment with the THPO-mimetic romiplostim induced trilineage hematological responses, remission of bleeding and infections, and transfusion independence, which were maintained after up to 6.5 years of observation. Recognizing patients with THPO mutations among those with juvenile bone marrow failure is essential to provide them with appropriate substitutive therapy and prevent the use of invasive and unnecessary treatments, such as hematopoietic stem cell transplantation or immunosuppression.


Asunto(s)
Mutación Puntual , Receptores Fc/uso terapéutico , Receptores de Trombopoyetina/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/genética , Trombopoyetina/genética , Línea Celular , Preescolar , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Femenino , Células HEK293 , Humanos , Lactante , Masculino , Linaje , Mutación Puntual/efectos de los fármacos , Trombopoyetina/uso terapéutico
14.
Haematologica ; 101(11): 1333-1342, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27365488

RESUMEN

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.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiología , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Represoras/genética , Trombocitopenia/genética , Adolescente , Adulto , Transformación Celular Neoplásica/genética , Niño , Preescolar , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Familia , Humanos , Lactante , Recién Nacido , Péptidos y Proteínas de Señalización Intercelular , Persona de Mediana Edad , Mutación , Proteínas Nucleares/genética , Linaje , Trombocitopenia/patología , Adulto Joven , Proteína ETS de Variante de Translocación 6
16.
J Hematol Oncol ; 8: 71, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26081257

RESUMEN

BACKGROUND: Increased plasma levels of proteasome have been associated with various neoplasms, especially myeloid malignancies. Little is known of the cellular origin and release mechanisms of such proteasome. We recently identified and characterized a novel particulate cytoplasmic structure (PaCS) showing selective accumulation of ubiquitin-proteasome system (UPS) components. PaCSs have been reported in some epithelial neoplasms and in two genetic disorders characterized by hematopoietic cell dysplasia and increased risk of leukemia. However, no information is available about PaCSs in hematopoietic neoplasms. METHODS: PaCSs were investigated by ultrastructural, immunogold, and immunofluorescence analysis of bone marrow (BM) biopsies and peripheral blood (PB) cell preparations of 33 consecutive, untreated, or relapsed patients affected by different hematopoietic neoplasms. BM and PB samples from individuals with non-neoplastic BM or healthy donors were studied as controls. Granulocytes and platelet proteasome content was measured by immunoblotting and plasma proteasome levels by ELISA. RESULTS: PaCSs with typical, selective immunoreactivity for polyubiquitinated proteins and proteasome were widespread in granulocytic cells, megakaryocytes, and platelets of patients with myeloproliferative neoplasms (MPN). In acute myeloid leukemia and myelodysplastic syndromes (MDS), PaCSs were only occasionally detected in blast cells and were found consistently in cells showing granulocytic and megakaryocytic maturation. Conversely, PaCSs were poorly represented or absent in non-neoplastic hematopoietic tissue or lymphoid neoplasms. In MPN granulocytes and platelets, the presence of PaCSs was associated with increased amounts of proteasome in cell lysates. PaCSs were often localized in cytoplasmic blebs generating PaCSs-filled plasma membrane vesicles observable in the BM intercellular space. In MPN and MDS, accumulation of PaCSs was associated with significant increase in plasma proteasome. Immunogold analysis showed that PaCSs of myeloid neoplasia selectively concentrated the chaperone proteins Hsp40, Hsp70, and Hsp90. CONCLUSIONS: PaCSs accumulate in cells of myeloid neoplasms in a lineage- and maturation-restricted manner; in particular, they are widespread in granulocytic and megakaryocytic lineages of MPN patients. PaCSs development was associated with excess accumulation of polyubiquitinated proteins, proteasome, and chaperone molecules, indicating impairment of the UPS-dependent protein homeostasis and a possible link with Hsp90-related leukemogenesis. A mechanism of PaCSs discharge by leukemic cells could contribute to increased plasma proteasome of MPN and MDS.


Asunto(s)
Trastornos Mieloproliferativos/genética , Complejo de la Endopetidasa Proteasomal/genética , Ubiquitina/genética , Adulto , Anciano , Anciano de 80 o más Años , Preescolar , Estructuras Citoplasmáticas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Trastornos Mieloproliferativos/patología
17.
Histochem Cell Biol ; 144(2): 179-84, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25952156

RESUMEN

Ubiquitin-proteasome system (UPS) proteins and proteolytic activity are localized in a recently identified cytoplasmic structure characterized by accumulation of barrel-like particles, which is known as the particulate cytoplasmic structure (PaCS). PaCSs have been detected in neoplastic, preneoplastic, chronically infected, and fetal cells, which produce high amounts of misfolded proteins to be degraded by the UPS. Chaperone molecules are crucial in the early stages of handling misfolded proteins; therefore, we searched for these molecules in PaCSs. Heat shock proteins (Hsp), Hsp90, Hsp70, Hsp40, and Bcl-2-associated athanogene (Bag)3 chaperones, although not Bag6, were selectively concentrated into PaCSs of several cell lines and ex vivo fetal or neoplastic cells. Present findings point to PaCSs as an integrated, active UPS center well equipped for metabolism of misfolded proteins, especially in cells under physiological (fetal development) or pathological (neoplasia or inflammation) stress.


Asunto(s)
Estructuras Citoplasmáticas/metabolismo , Proteínas de Choque Térmico/análisis , Chaperonas Moleculares/análisis , Complejo de la Endopetidasa Proteasomal/metabolismo , Pliegue de Proteína , Ubiquitina/metabolismo , Línea Celular Tumoral , Estructuras Citoplasmáticas/química , Proteínas de Choque Térmico/metabolismo , Humanos , Inmunohistoquímica , Lactante , Chaperonas Moleculares/metabolismo
18.
Blood ; 124(6): e4-e10, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-24990887

RESUMEN

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.


Asunto(s)
Plaquetas/patología , Trombocitopenia/sangre , Trombocitopenia/genética , Adolescente , Adulto , Estudios de Casos y Controles , Tamaño de la Célula , Niño , Preescolar , Diagnóstico Diferencial , Femenino , Pérdida Auditiva Sensorineural/sangre , Pérdida Auditiva Sensorineural/clasificación , Pérdida Auditiva Sensorineural/genética , Humanos , Lactante , Masculino , Persona de Mediana Edad , Proteínas Motoras Moleculares/genética , Mutación , Cadenas Pesadas de Miosina/genética , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/diagnóstico , Trombocitopenia/clasificación , Trombocitopenia/congénito , Adulto Joven
19.
Eur J Med Genet ; 56(1): 7-12, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23123319

RESUMEN

MYH9-related disease (MYH9-RD) is a rare autosomal dominant syndromic disorder caused by mutations in MYH9, the gene encoding for the heavy chain of non-muscle myosin IIA (myosin-9). MYH9-RD is characterized by congenital macrothrombocytopenia and typical inclusion bodies in neutrophils associated with a variable risk of developing sensorineural deafness, presenile cataract, and/or progressive nephropathy. The spectrum of mutations responsible for MYH9-RD is limited. We report five families, each with a novel MYH9 mutation. Two mutations, p.Val34Gly and p.Arg702Ser, affect the motor domain of myosin-9, whereas the other three, p.Met847_Glu853dup, p.Lys1048_Glu1054del, and p.Asp1447Tyr, hit the coiled-coil tail domain of the protein. The motor domain mutations were associated with more severe clinical phenotypes than those in the tail domain.


Asunto(s)
Proteínas Motoras Moleculares/genética , Mutación , Cadenas Pesadas de Miosina/genética , Trombocitopenia/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Secuencia de Bases , Niño , Preescolar , Exones , Femenino , Genes Dominantes , Estudios de Asociación Genética , Humanos , Masculino , Persona de Mediana Edad , Modelos Moleculares , Proteínas Motoras Moleculares/química , Datos de Secuencia Molecular , Cadenas Pesadas de Miosina/química , Linaje , Conformación Proteica , Alineación de Secuencia , Síndrome , Trombocitopenia/diagnóstico , Adulto Joven
20.
Thromb Haemost ; 109(2): 263-71, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23223974

RESUMEN

ANKRD26-related thrombocytopenia (ANKRD26-RT) is an autosomal-dominant thrombocytopenia caused by mutations in the 5'UTR of the ANKRD26 gene. ANKRD26-RT is characterised by dysmegakaryopoiesis and an increased risk of leukaemia. PaCSs are novel particulate cytoplasmic structures with selective immunoreactivity for polyubiquitinated proteins and proteasome that have been detected in a number of solid cancers, in the epithelia of Helicobacter pylori gastritis and related preneoplastic lesions, and in the neutrophils of Schwachman-Diamond syndrome, a genetic disease with neutropenia and increased leukaemia risk. We searched for PaCSs in blood cells from 14 consecutive patients with ANKRD26-RT. Electron microscopy combined with immunogold staining for polyubiquitinated proteins, 20S and 19S proteasome showed PaCSs in most ANKRD26-RT platelets, as in a restricted minority of platelets from healthy controls and from subjects with other inherited or immune thrombocytopenias. In ANKRD26-RT platelets, the PaCS amount exceeded that of control platelets by a factor of 5 (p<0.0001). Immunoblotting showed that the higher PaCS number was associated with increased amounts of polyubiquitinated proteins and proteasome in ANKRD26-RT platelets. PaCSs were also extensively represented in ANKRD26-RT megakaryocytes, but not in healthy control megakaryocytes, and were absent in other ANKRD26-RT and control blood cells. Therefore, large amounts of PaCSs are a characteristic feature of ANKRD26-RT platelets and megakaryocytes, although these novel cell components are also present in a small subpopulation of normal platelets. The widespread presence of PaCSs in inherited diseases with increased leukaemia risk, as well as in solid neoplasms and their preneoplastic lesions, suggests a link of these structures with oncogenesis.


Asunto(s)
Plaquetas/enzimología , Estructuras Citoplasmáticas/enzimología , Megacariocitos/enzimología , Mutación , Complejo de la Endopetidasa Proteasomal/sangre , Trombocitopenia/enzimología , Adulto , Anciano , Plaquetas/ultraestructura , Western Blotting , Estudios de Casos y Controles , Estructuras Citoplasmáticas/ultraestructura , Femenino , Predisposición Genética a la Enfermedad , Humanos , Inmunohistoquímica , Masculino , Megacariocitos/ultraestructura , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Fenotipo , Trombocitopenia/sangre , Trombocitopenia/genética , Trombocitopenia/patología , Ubiquitinación , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...