Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 229
Filtrar
1.
Open Biol ; 14(6): 240041, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38835242

RESUMEN

Platelets are blood cells derived from megakaryocytes that play a central role in regulating haemostasis and vascular integrity. The microtubule cytoskeleton of megakaryocytes undergoes a critical dynamic reorganization during cycles of endomitosis and platelet biogenesis. Quiescent platelets have a discoid shape maintained by a marginal band composed of microtubule bundles, which undergoes remarkable remodelling during platelet activation, driving shape change and platelet function. Disrupting or enhancing this process can cause platelet dysfunction such as bleeding disorders or thrombosis. However, little is known about the molecular mechanisms underlying the reorganization of the cytoskeleton in the platelet lineage. Recent studies indicate that the emergence of a unique platelet tubulin code and specific pathogenic tubulin mutations cause platelet defects and bleeding disorders. Frequently, these mutations exhibit dominant negative effects, offering valuable insights into both platelet disease mechanisms and the functioning of tubulins. This review will highlight our current understanding of the role of the microtubule cytoskeleton in the life and death of platelets, along with its relevance to platelet disorders.


Asunto(s)
Plaquetas , Citoesqueleto , Megacariocitos , Microtúbulos , Humanos , Plaquetas/metabolismo , Megacariocitos/metabolismo , Megacariocitos/citología , Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/genética , Animales , Trastornos de las Plaquetas Sanguíneas/metabolismo , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/patología , Mutación
2.
Sci Rep ; 14(1): 14080, 2024 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890442

RESUMEN

Familial platelet disorder with associated myeloid malignancies (FPDMM) is an autosomal dominant disease caused by heterozygous germline mutations in RUNX1. It is characterized by thrombocytopenia, platelet dysfunction, and a predisposition to hematological malignancies. Although FPDMM is a precursor for diseases involving abnormal DNA methylation, the DNA methylation status in FPDMM remains unknown, largely due to a lack of animal models and challenges in obtaining patient-derived samples. Here, using genome editing techniques, we established two lines of human induced pluripotent stem cells (iPSCs) with different FPDMM-mimicking heterozygous RUNX1 mutations. These iPSCs showed defective differentiation of hematopoietic progenitor cells (HPCs) and megakaryocytes (Mks), consistent with FPDMM. The FPDMM-mimicking HPCs showed DNA methylation patterns distinct from those of wild-type HPCs, with hypermethylated regions showing the enrichment of ETS transcription factor (TF) motifs. We found that the expression of FLI1, an ETS family member, was significantly downregulated in FPDMM-mimicking HPCs with a RUNX1 transactivation domain (TAD) mutation. We demonstrated that FLI1 promoted binding-site-directed DNA demethylation, and that overexpression of FLI1 restored their megakaryocytic differentiation efficiency and hypermethylation status. These findings suggest that FLI1 plays a crucial role in regulating DNA methylation and correcting defective megakaryocytic differentiation in FPDMM-mimicking HPCs with a RUNX1 TAD mutation.


Asunto(s)
Diferenciación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Metilación de ADN , Células Madre Pluripotentes Inducidas , Megacariocitos , Mutación , Proteína Proto-Oncogénica c-fli-1 , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Megacariocitos/metabolismo , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/metabolismo , Trastornos de las Plaquetas Sanguíneas/patología , Activación Transcripcional , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Leucemia Mieloide Aguda , Trastornos de la Coagulación Sanguínea Heredados
3.
Blood Cancer J ; 14(1): 25, 2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38316746

RESUMEN

Germline, mono-allelic mutations in RUNX1 cause familial platelet disorder (RUNX1-FPD) that evolves into myeloid malignancy (FPD-MM): MDS or AML. FPD-MM commonly harbors co-mutations in the second RUNX1 allele and/or other epigenetic regulators. Here we utilized patient-derived (PD) FPD-MM cells and established the first FPD-MM AML cell line (GMR-AML1). GMR-AML1 cells exhibited active super-enhancers of MYB, MYC, BCL2 and CDK6, augmented expressions of c-Myc, c-Myb, EVI1 and PLK1 and surface markers of AML stem cells. In longitudinally studied bone marrow cells from a patient at FPD-MM vs RUNX1-FPD state, we confirmed increased chromatin accessibility and mRNA expressions of MYB, MECOM and BCL2 in FPD-MM cells. GMR-AML1 and PD FPD-MM cells were sensitive to homoharringtonine (HHT or omacetaxine) or mebendazole-induced lethality, associated with repression of c-Myc, EVI1, PLK1, CDK6 and MCL1. Co-treatment with MB and the PLK1 inhibitor volasertib exerted synergistic in vitro lethality in GMR-AML1 cells. In luciferase-expressing GMR-AML1 xenograft model, MB, omacetaxine or volasertib monotherapy, or co-treatment with MB and volasertib, significantly reduced AML burden and improved survival in the immune-depleted mice. These findings highlight the molecular features of FPD-MM progression and demonstrate HHT, MB and/or volasertib as effective agents against cellular models of FPD-MM.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas , Leucemia Mieloide Aguda , Humanos , Animales , Ratones , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Homoharringtonina , Plaquetas/patología , Trastornos de las Plaquetas Sanguíneas/complicaciones , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/patología , Proteínas Proto-Oncogénicas c-bcl-2
6.
Int J Mol Sci ; 22(18)2021 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-34576024

RESUMEN

Platelets are small anucleate blood cells that play vital roles in haemostasis and thrombosis, besides other physiological and pathophysiological processes. These roles are tightly regulated by a complex network of signalling pathways. Mass spectrometry-based proteomic techniques are contributing not only to the identification and quantification of new platelet proteins, but also reveal post-translational modifications of these molecules, such as acetylation, glycosylation and phosphorylation. Moreover, target proteomic analysis of platelets can provide molecular biomarkers for genetic aberrations with established or non-established links to platelet dysfunctions. In this report, we review 67 reports regarding platelet proteomic analysis and signalling on a molecular base. Collectively, these provide detailed insight into the: (i) technical developments and limitations of the assessment of platelet (sub)proteomes; (ii) molecular protein changes upon ageing of platelets; (iii) complexity of platelet signalling pathways and functions in response to collagen, rhodocytin, thrombin, thromboxane A2 and ADP; (iv) proteomic effects of endothelial-derived mediators such as prostacyclin and the anti-platelet drug aspirin; and (v) molecular protein changes in platelets from patients with congenital disorders or cardiovascular disease. However, sample sizes are still low and the roles of differentially expressed proteins are often unknown. Based on the practical and technical possibilities and limitations, we provide a perspective for further improvements of the platelet proteomic field.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/genética , Plaquetas/metabolismo , Proteoma/genética , Proteómica , Trastornos de las Plaquetas Sanguíneas/sangre , Trastornos de las Plaquetas Sanguíneas/patología , Humanos , Activación Plaquetaria/genética , Procesamiento Proteico-Postraduccional/genética , Transducción de Señal/genética
7.
Muscle Nerve ; 64(5): 567-575, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34368974

RESUMEN

INTRODUCTION/AIMS: Stromal interaction molecule 1 (STIM1) is a reticular Ca2+ sensor composed of a luminal and a cytosolic domain. Autosomal dominant mutations in STIM1 cause tubular aggregate myopathy and Stormorken syndrome or its variant York platelet syndrome. In this study we aimed to expand the features related to new variants in STIM1. METHODS: We performed a cross-sectional study of individuals harboring monoallelic STIM1 variants recruited at five tertiary centers involved in a study of inherited myopathies analyzed with a multigene-targeted panel. RESULTS: We identified seven individuals (age range, 26-57 years) harboring variants in STIM1, including five novel changes: three located in the EF-hand domain, one in the sterile α motif (SAM) domain, and one in the cytoplasmatic region of the protein. Functional evaluation of the pathogenic variants using a heterologous expression system and measuring store-operated calcium entry demonstrated their causative role and suggested a link of new variants with the clinical phenotype. Muscle contractures, found in three individuals, showed variability in body distribution and in the number of joints involved. Three patients showed cardiac and respiratory involvement. Short stature, hyposplenism, sensorineural hearing loss, hypothyroidism, and Gilbert syndrome were variably observed among the patients. Laboratory tests revealed hyperCKemia in six patients, thrombocytopenia in two patients, and hypocalcemia in one patient. Muscle biopsy showed the presence of tubular aggregates in three patients, type I fiber atrophy in one patient, and nonspecific myopathic changes in two patients. DISCUSSION: Our clinical, histological, and molecular data expand the genetic and clinical spectrum of STIM1-related diseases.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas , Miopatías Estructurales Congénitas , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/metabolismo , Trastornos de las Plaquetas Sanguíneas/patología , Calcio/metabolismo , Estudios Transversales , Humanos , Miosis/genética , Miosis/metabolismo , Miosis/patología , Miopatías Estructurales Congénitas/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
10.
Small GTPases ; 12(5-6): 440-457, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33459160

RESUMEN

Platelets are master regulators and effectors of haemostasis with increasingly recognized functions as mediators of inflammation and immune responses. The Rho family of GTPase members Rac1, Cdc42 and RhoA are known to be major components of the intracellular signalling network critical to platelet shape change and morphological dynamics, thus playing a major role in platelet spreading, secretion and thrombus formation. Initially linked to the regulation of actomyosin contraction and lamellipodia formation, recent reports have uncovered non-canonical functions of platelet RhoGTPases in the regulation of reactive oxygen species (ROS), where intrinsically generated ROS modulate platelet function and contribute to thrombus formation. Platelet RhoGTPases orchestrate oxidative processes and cytoskeletal rearrangement in an interconnected manner to regulate intracellular signalling networks underlying platelet activity and thrombus formation. Herein we review our current knowledge of the regulation of platelet ROS generation by RhoGTPases and their relationship with platelet cytoskeletal reorganization, activation and function.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/patología , Plaquetas/fisiología , Especies Reactivas de Oxígeno/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Trastornos de las Plaquetas Sanguíneas/enzimología , Plaquetas/citología , Plaquetas/enzimología , Humanos
11.
Neuropathology ; 40(6): 559-569, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33073872

RESUMEN

Tubular aggregate myopathy (TAM) is a progressive disorder characterized by muscle weakness, cramps, and myalgia. TAM clinically overlaps with Stormorken syndrome (STRMK), combining TAM with miosis, thrombocytopenia, hyposplenism, ichthyosis, short stature, and dyslexia. TAM and STRMK arise from gain-of-function mutations in STIM1 (stromal interaction molecule 1) or ORAI1, both encoding key regulators of Ca2+ homeostasis, and mutations in either gene result in excessive extracellular Ca2+ entry. The pathomechanistic similarities and differences between TAM and STRMK are only partially understood. Here we provide functional in vitro experiments demonstrating that STIM1 harboring the TAM D84G or the STRMK R304W mutation similarly cluster and exert a dominant effect on the wild-type protein. Both mutants recruit ORAI1 to the clusters, increase cytosolic Ca2+ levels, promote major nuclear import of the Ca2+ -dependent transcription factor NFAT (nuclear factor of activated T cells), and trigger the formation of circular membrane stacks. In conclusion, the analyzed TAM and STRMK mutations have a comparable impact on STIM1 protein function and downstream effects of excessive Ca2+ entry, highlighting that TAM and STRMK involve a common pathomechanism.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/genética , Dislexia/genética , Ictiosis/genética , Trastornos Migrañosos/genética , Miosis/genética , Miopatías Estructurales Congénitas/genética , Proteínas de Neoplasias/genética , Bazo/anomalías , Molécula de Interacción Estromal 1/genética , Animales , Trastornos de las Plaquetas Sanguíneas/metabolismo , Trastornos de las Plaquetas Sanguíneas/patología , Células Cultivadas , Dislexia/metabolismo , Dislexia/patología , Eritrocitos Anormales/metabolismo , Eritrocitos Anormales/patología , Humanos , Ictiosis/metabolismo , Ictiosis/patología , Ratones , Trastornos Migrañosos/metabolismo , Trastornos Migrañosos/patología , Miosis/metabolismo , Miosis/patología , Fatiga Muscular/genética , Mutación , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/patología , Factores de Transcripción NFATC/metabolismo , Proteína ORAI1/metabolismo , Bazo/metabolismo , Bazo/patología , Transfección
12.
Cytometry B Clin Cytom ; 98(6): 464-475, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32516490

RESUMEN

Inherited platelet function disorders are rare hemorrhagic diseases. The gold standard for their exploration is optical aggregometry; however, investigations by flow cytometry (FCM) are being increasingly used. In this review, the physiology of platelets is first recalled, setting the stage for the compartments of platelets that can be apprehended by specific and appropriate labeling. As this requires some pre-analytical precautions and specific analytical settings, a second part focuses on these characteristic aspects, based on literature and on the authors' experience in the field, for qualitative or quantitative explorations. Membrane labeling with antibodies to CD42a or CD41, respectively, useful to assess the genetic-related defects of Glanzmann thrombocytopenia and Bernard Soulier syndrome are then described. Platelet degranulation disorders are detailed in the next section, as they can be explored, upon platelet activation, by measuring the expression of surface P-Selectin (CD62P) or CD63. Mepacrin uptake and release after activation is another test allowing to explore the function of dense granules. Finally, the flip-flop anomaly related to Scott syndrome is depicted. Tables summarizing possible FCM assays, and characteristic histograms are provided as reference for flow laboratories interested in developing platelet exploration.


Asunto(s)
Trastornos de la Coagulación Sanguínea Heredados/sangre , Trastornos de las Plaquetas Sanguíneas/sangre , Citometría de Flujo , Inmunofenotipificación , Trastornos de la Coagulación Sanguínea Heredados/tratamiento farmacológico , Trastornos de la Coagulación Sanguínea Heredados/genética , Trastornos de la Coagulación Sanguínea Heredados/inmunología , Trastornos de las Plaquetas Sanguíneas/tratamiento farmacológico , Trastornos de las Plaquetas Sanguíneas/inmunología , Trastornos de las Plaquetas Sanguíneas/patología , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Plaquetas/patología , Humanos , Activación Plaquetaria/genética , Activación Plaquetaria/inmunología , Quinacrina/uso terapéutico
13.
Horm Res Paediatr ; 93(1): 40-45, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32388508

RESUMEN

BACKGROUND AND OBJECTIVE: Gastrointestinal (GI) polyps with unknown malignant potential and a platelet storage pool deficiency that increases the risk of severe intraoperative and other types of bleeding have been identified in McCune-Albright syndrome (MAS). The natural course of these disorders has not been well characterized. The aim of this study was to report the follow-up of GI polyps and platelet dysfunction (PD) in a cohort of 28 patients with MAS. METHODS: Twenty-eight patients with MAS (15 females) were included. Endoscopic screening for GI polyps was undertaken in 14 subjects and 19 were tested for PD. RESULTS: Six subjects (5 males) were diagnosed with GI polyps at a median age of 23 (range 15-43) years, and were monitored for a median period of 8 (range 4.5-11.5) years. At endoscopic follow-up, the 4 patients with hamartomatous polyps at first endoscopy had either normal findings (n = 2), or duodenal gastric metaplasia (n = 2). Two patients with caecal polyps were identified. Of 8 subjects with a platelet storage pool deficiency, 5 required transfusions during surgery, and subsequent platelet cover in 2 markedly reduced intraoperative blood loss. CONCLUSIONS: New polyps with uncertain malignant potential are diagnosed after long term follow-up in MAS. Platelet cover reduces the need for red blood cell transfusion during orthopaedic surgery and may be useful to reduce non-operative bleeding events. We recommend regular upper and lower endoscopy and screening for PD in all MAS patients.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/complicaciones , Displasia Fibrosa Poliostótica/complicaciones , Pólipos Intestinales/complicaciones , Adolescente , Adulto , Trastornos de las Plaquetas Sanguíneas/sangre , Trastornos de las Plaquetas Sanguíneas/patología , Femenino , Displasia Fibrosa Poliostótica/sangre , Displasia Fibrosa Poliostótica/patología , Estudios de Seguimiento , Humanos , Pólipos Intestinales/sangre , Pólipos Intestinales/patología , Masculino , Persona de Mediana Edad , Adulto Joven
14.
Pediatr Blood Cancer ; 67(2): e28078, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31724816

RESUMEN

Defects of platelet intracellular signaling can result in severe platelet dysfunction. Several mutations in each of the linked genes FERMT3 and RASGRP2 on chromosome 11 causing a Glanzmann-like bleeding phenotype have been identified so far. We report on novel variants in two unrelated pediatric patients with severe bleeding diathesis-one with leukocyte adhesion deficiency type III due to a homozygous frameshift in FERMT3 and the other with homozygous variants in both, FERMT3 and RASGRP2. We focus on the challenging genetic and functional variant assessment and aim to accentuate the risk of obtaining misleading results due to the phenomenon of genetic linkage.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/patología , Factores de Intercambio de Guanina Nucleótido/genética , Trastornos Hemorrágicos/patología , Proteínas de la Membrana/genética , Mutación , Proteínas de Neoplasias/genética , Adolescente , Trastornos de las Plaquetas Sanguíneas/genética , Niño , Femenino , Ligamiento Genético , Trastornos Hemorrágicos/genética , Homocigoto , Humanos , Masculino , Linaje , Fenotipo , Pronóstico
15.
Sci Transl Med ; 11(522)2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31826978

RESUMEN

Platelet transfusions can fail to prevent bleeding in patients with inherited platelet function disorders (IPDs), such as Glanzmann's thrombasthenia (GT; integrin αIIbß3 dysfunction), Bernard-Soulier syndrome [BSS; glycoprotein (GP) Ib/V/IX dysfunction], and the more recently identified nonsyndromic RASGRP2 variants. Here, we used IPD mouse models and real-time imaging of hemostatic plug formation to investigate whether dysfunctional platelets impair the hemostatic function of healthy donor [wild-type (WT)] platelets. In Rasgrp2-/- mice or mice with platelet-specific deficiency in the integrin adaptor protein TALIN1 ("GT-like"), WT platelet transfusion was ineffective unless the ratio between mutant and WT platelets was ~2:1. In contrast, thrombocytopenic mice or mice lacking the extracellular domain of GPIbα ("BSS-like") required very few transfused WT platelets to normalize hemostasis. Both Rasgrp2-/- and GT-like, but not BSS-like, platelets effectively localized to the injury site. Mechanistic studies identified at least two mechanisms of interference by dysfunctional platelets in IPDs: (i) delayed adhesion of WT donor platelets due to reduced access to GPIbα ligands exposed at sites of vascular injury and (ii) impaired consolidation of the hemostatic plug. We also investigated the hemostatic activity of transfused platelets in the setting of dual antiplatelet therapy (DAPT), an acquired platelet function disorder (APD). "DAPT" platelets did not prolong the time to initial hemostasis, but plugs were unstable and frequent rebleeding was observed. Thus, we propose that the endogenous platelet count and the ratio of transfused versus endogenous platelets should be considered when treating select IPD and APD patients with platelet transfusions.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/patología , Plaquetas/patología , Hemostasis , Transfusión de Plaquetas , Animales , Sitios de Unión , Terapia Antiplaquetaria Doble , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Integrinas/metabolismo , Ligandos , Ratones Endogámicos C57BL , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Trombastenia/patología , Donantes de Tejidos
16.
Stem Cell Res ; 41: 101603, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31698193

RESUMEN

Familial Platelet Disorder with associated Myeloid Malignancy (FPDMM) is a rare platelet disorder caused by mutations in RUNX1. We generated an iPSC line (GENYOi005-A) from a FPDMM patient with a non-previously reported variant p.Thr196Ala. Non-integrative Sendai viruses expressing the Yamanaka reprogramming factors were used to reprogram peripheral blood mononuclear cells from this FPDMM patient. Characterization of GENYOi005-A included genetic analysis of RUNX1 locus, Short Tandem Repeats profiling, alkaline phosphatase enzymatic activity, expression of pluripotency-associated factors and differentiation studies in vitro and in vivo. This iPSC line will provide a powerful tool to study developmental alterations of FPDMM patients.


Asunto(s)
Trastornos de la Coagulación Sanguínea Heredados/genética , Trastornos de la Coagulación Sanguínea Heredados/patología , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/patología , Diferenciación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Madre Pluripotentes Inducidas/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Leucocitos Mononucleares/patología , Mutación , Células Cultivadas , Reprogramación Celular , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Leucocitos Mononucleares/metabolismo , Persona de Mediana Edad
18.
J Thromb Haemost ; 17(9): 1430-1439, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31220402

RESUMEN

The past decade has brought unprecedented advances in our understanding of megakaryocyte (MK) biology and platelet production, processes that are strongly dependent on the cytoskeleton. Facilitated by technological innovations, such as new high-resolution imaging techniques (in vitro and in vivo) and lineage-specific gene knockout and reporter mouse strains, we are now able to visualize and characterize the molecular machinery required for MK development and proplatelet formation in live mice. Whole genome and RNA sequencing analysis of patients with rare platelet disorders, combined with targeted genetic interventions in mice, has led to the identification and characterization of numerous new genes important for MK development. Many of the genes important for proplatelet formation code for proteins that control cytoskeletal dynamics in cells, such as Rho GTPases and their downstream targets. In this review, we discuss how the final stages of MK development are controlled by the cellular cytoskeletons, and we compare changes in MK biology observed in patients and mice with mutations in cytoskeleton regulatory genes.


Asunto(s)
Plaquetas/fisiología , Citoesqueleto/fisiología , Trombopoyesis/fisiología , Actinas/metabolismo , Animales , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/patología , Plaquetas/ultraestructura , Proteínas Sanguíneas/metabolismo , Gránulos Citoplasmáticos/fisiología , Forminas/sangre , Genes Reporteros , Humanos , Ratones , Ratones Noqueados , Miosina Tipo IIA no Muscular/sangre , Biogénesis de Organelos , Trombopoyesis/genética , Tubulina (Proteína)/metabolismo
19.
Haematologica ; 104(10): 2084-2090, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30819905

RESUMEN

Primary platelet secretion defects constitute a heterogeneous group of functional defects characterized by reduced platelet granule secretion upon stimulation by different agonists. The clinical and laboratory heterogeneity of primary platelet secretion defects warrants a tailored approach. We performed a pilot study in order to develop DNA sequence analysis pipelines for gene discovery and to create a list of candidate causal genes for platelet secretion defects. Whole-exome sequencing analysis of 14 unrelated Italian patients with primary secretion defects and 16 controls was performed on Illumina HiSeq. Variant prioritization was carried out using two filtering approaches: identification of rare, potentially damaging variants in platelet candidate genes or by selecting singletons. To corroborate the results, exome sequencing was applied in a family in which platelet secretion defects and a bleeding diathesis were present. Platelet candidate gene analysis revealed gene defects in 10/14 patients, which included ADRA2A, ARHGAP1, DIAPH1, EXOC1, FCGR2A, ITPR1, LTBP1, PTPN7, PTPN12, PRKACG, PRKCD, RAP1GAP, STXBP5L, and VWF The analysis of singletons identified additional gene defects in PLG and PHACTR2 in two other patients. The family analysis confirmed a missense variant p.D1144N in the STXBP5L gene and p.P83H in the KCNMB3 gene as potentially causal. In summary, exome sequencing revealed potential causal variants in 12 of 14 patients with primary platelet secretion defects, highlighting the limitations of the genomic approaches for causal gene identification in this heterogeneous clinical and laboratory phenotype.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/genética , Secuenciación del Exoma , Adulto , Trastornos de las Plaquetas Sanguíneas/metabolismo , Trastornos de las Plaquetas Sanguíneas/patología , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto
20.
J Child Neurol ; 34(6): 321-324, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30761937

RESUMEN

Stormorken syndrome is a rare genetic disorder (MIM 185070) first reported in 1983 with thrombocytopenia, muscle weakness, asplenia, and miosis caused by a mutation of the stromal interaction molecule 1 ( STIM1) gene.1 The muscle weakness is caused by a myopathy with tubular aggregate formation. We report a family in which both child and mother presented with proximal muscle weakness and thrombocytopenia. Histologic, histochemical, and electron microscopy studies were performed on the muscle specimen. It documented accumulation of tubular aggregates and chronic myopathic changes with dystrophic features. Genetic testing revealed that both mother and son carried a missense mutation of c.326A>G in exon 3 of the STIM1 gene, which is novel for Stormorken syndrome. We suggest that patients with unexplained chronic idiopathic thrombocytopenia and proximal weakness have genetic testing for Stormorken syndrome.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/diagnóstico , Trastornos de las Plaquetas Sanguíneas/patología , Dislexia/diagnóstico , Dislexia/patología , Ictiosis/diagnóstico , Ictiosis/patología , Trastornos Migrañosos/diagnóstico , Trastornos Migrañosos/patología , Miosis/diagnóstico , Miosis/patología , Bazo/anomalías , Trastornos de las Plaquetas Sanguíneas/complicaciones , Preescolar , Dislexia/complicaciones , Eritrocitos Anormales/patología , Humanos , Ictiosis/complicaciones , Masculino , Microscopía Electrónica , Trastornos Migrañosos/complicaciones , Miosis/complicaciones , Fatiga Muscular , Debilidad Muscular/etiología , Debilidad Muscular/patología , Mutación Missense , Miopatías Estructurales Congénitas/complicaciones , Miopatías Estructurales Congénitas/diagnóstico , Miopatías Estructurales Congénitas/patología , Bazo/patología , Trombocitopenia/etiología , Trombocitopenia/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...