Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Investig Med High Impact Case Rep ; 11: 23247096231196697, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37649376

RESUMEN

Gray platelet syndrome (GPS) is a rare hereditary hemorrhagic disorder characterized by macrothrombocytopenia and the absence of alpha-granules in platelets. Clinically, mild-to-moderate bleeding is the main manifestation, often accompanied by thrombocytopenia, splenomegaly, and myelofibrosis. Here, we present a case of a 15-year-old male patient with a history of hepatosplenomegaly, and thrombocytopenia for 8 years, who presented with sudden generalized abdominal pain. Despite initial suspicion of gastroenteritis, diagnostic imaging revealed an extensive hemoperitoneum. Subsequent genetic testing confirmed the diagnosis of GPS, which had not been previously identified. This case highlights the importance of considering inherited platelet disorders should be considered in adolescents with long-standing thrombocytopenia, and emphasizes the need for thorough evaluation in patients with suggestive symptoms.


Asunto(s)
Síndrome de Plaquetas Grises , Rotura del Bazo , Trombocitopenia , Masculino , Adolescente , Humanos , Síndrome de Plaquetas Grises/complicaciones , Síndrome de Plaquetas Grises/diagnóstico , Síndrome de Plaquetas Grises/genética , Plaquetas , Trombocitopenia/etiología , Esplenomegalia/etiología , Rotura del Bazo/diagnóstico , Rotura del Bazo/etiología , Hemorragia
2.
Nat Commun ; 14(1): 3728, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37349339

RESUMEN

Loss of NBEAL2 function leads to grey platelet syndrome (GPS), a bleeding disorder characterized by macro-thrombocytopenia and α-granule-deficient platelets. A proportion of patients with GPS develop autoimmunity through an unknown mechanism, which might be related to the proteins NBEAL2 interacts with, specifically in immune cells. Here we show a comprehensive interactome of NBEAL2 in primary T cells, based on mass spectrometry identification of altogether 74 protein association partners. These include LRBA, a member of the same BEACH domain family as NBEAL2, recessive mutations of which cause autoimmunity and lymphocytic infiltration through defective CTLA-4 trafficking. Investigating the potential association between NBEAL2 and CTLA-4 signalling suggested by the mass spectrometry results, we confirm by co-immunoprecipitation that CTLA-4 and NBEAL2 interact with each other. Interestingly, NBEAL2 deficiency leads to low CTLA-4 expression in patient-derived effector T cells, while their regulatory T cells appear unaffected. Knocking-down NBEAL2 in healthy primary T cells recapitulates the low CTLA-4 expression observed in the T cells of GPS patients. Our results thus show that NBEAL2 is involved in the regulation of CTLA-4 expression in conventional T cells and provide a rationale for considering CTLA-4-immunoglobulin therapy in patients with GPS and autoimmune disease.


Asunto(s)
Síndrome de Plaquetas Grises , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Plaquetas/metabolismo , Proteínas Sanguíneas/genética , Antígeno CTLA-4/genética , Antígeno CTLA-4/metabolismo , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/metabolismo
3.
J Thromb Haemost ; 21(6): 1409-1419, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37028650

RESUMEN

Since the description of the first case with gray platelet syndrome (GPS) in 1971, this rare inherited platelet disorder has been the focus of extensive clinical and basic research. These studies have not only increased our knowledge about the clinical manifestations of GPS but also deepened our understanding of the biogenesis of platelet α-granules and their pathophysiology in hemostasis and thrombosis. The discovery of the causal gene, neurobeachin-like 2, in 2011 was a milestone in hematology. Following this was the rapid diagnosis and phenotyping of many new patients and the further development of experimental models to characterize the pathophysiological relevance of neurobeachin-like 2 in hemostasis and immunity. The impact of altered protein function on cells other than platelets became apparent, including defects in the granules of neutrophils and monocytes and changes in the transcriptomic and proteomic profiles of other immune cells such as T lymphocytes. Besides the previously recognized clinical manifestations of macrothrombocytopenia, splenomegaly, and early-onset bone marrow fibrosis, we now recognize that immunologic abnormalities, including autoimmune diseases and recurrent infections, affect a proportion of patients with GPS. There is a proinflammatory signature of the plasma in GPS, with quantitative alterations of multiple proteins, including many produced by the liver. This review will cover the classical features of GPS and then focus on additional clinical manifestations of immune dysregulation and cellular defects beyond platelets in patients with this rare disorder.


Asunto(s)
Enfermedades Autoinmunes , Síndrome de Plaquetas Grises , Humanos , Megacariocitos/metabolismo , Síndrome de Plaquetas Grises/diagnóstico , Síndrome de Plaquetas Grises/genética , Autoinmunidad , Proteómica , Plaquetas/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/metabolismo , Gránulos Citoplasmáticos/metabolismo
5.
Am J Clin Pathol ; 156(2): 253-258, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-33586768

RESUMEN

OBJECTIVES: Gray platelet syndrome (GPS) is a rare platelet storage pool disorder associated with a marked decrease or absence of platelet α-granules and their contents. It is characterized clinically by mild to moderate bleeding; moderate macrothrombocytopenia with large, agranular platelets; splenomegaly; and bone marrow fibrosis. Electron microscopy confirms markedly reduced or absent α-granules in platelets and megakaryocytes. The classic description of GPS is caused by homozygous mutations in NBEAL2 (neurobeachinlike 2). METHODS: A 28-year-old Hispanic man with a history of easy bruising and occasional episodes of epistaxis sought treatment for pancytopenia and splenomegaly. Peripheral blood smear and bone marrow analysis, electron microscopy, and next-generation sequencing were performed. RESULTS: Large and agranular platelets were present in the peripheral blood. There was bone marrow fibrosis. Electron microscopy of the platelets showed absence of α-granules. Next-generation sequencing revealed a germline apparently homozygous nonsense variant in the NBEAL2 gene: c.5674C>T, p.Gln1892X (p.Q1829X). CONCLUSIONS: The differential diagnosis of GPS includes a myeloid neoplasm such as myelodysplastic syndrome with bone marrow fibrosis. The availability of diagnostic genetic panels for hereditable platelet disorders can assist in the recognition of GPS and other platelet disorders. We also describe a previously unreported pathogenic germline homozygous nonsense variant in the NBEAL2 gene: c.5674C>T, p.Gln1892X (p.Q1829X) in a patient with GPS.


Asunto(s)
Proteínas Sanguíneas/genética , Síndrome de Plaquetas Grises/diagnóstico , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/patología , Adulto , Humanos , Masculino , Mutación , Pancitopenia/etiología , Pancitopenia/patología , Mielofibrosis Primaria/etiología , Mielofibrosis Primaria/patología , Esplenomegalia/etiología , Esplenomegalia/patología
6.
Blood Adv ; 5(2): 549-564, 2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33496751

RESUMEN

Gray platelet syndrome (GPS) is an autosomal recessive bleeding disorder characterized by a lack of α-granules in platelets and progressive myelofibrosis. Rare loss-of-function variants in neurobeachin-like 2 (NBEAL2), a member of the family of beige and Chédiak-Higashi (BEACH) genes, are causal of GPS. It is suggested that BEACH domain containing proteins are involved in fusion, fission, and trafficking of vesicles and granules. Studies in knockout mice suggest that NBEAL2 may control the formation and retention of granules in neutrophils. We found that neutrophils obtained from the peripheral blood from 13 patients with GPS have a normal distribution of azurophilic granules but show a deficiency of specific granules (SGs), as confirmed by immunoelectron microscopy and mass spectrometry proteomics analyses. CD34+ hematopoietic stem cells (HSCs) from patients with GPS differentiated into mature neutrophils also lacked NBEAL2 expression but showed similar SG protein expression as control cells. This is indicative of normal granulopoiesis in GPS and identifies NBEAL2 as a potentially important regulator of granule release. Patient neutrophil functions, including production of reactive oxygen species, chemotaxis, and killing of bacteria and fungi, were intact. NETosis was absent in circulating GPS neutrophils. Lack of NETosis is suggested to be independent of NBEAL2 expression but associated with SG defects instead, as indicated by comparison with HSC-derived neutrophils. Since patients with GPS do not excessively suffer from infections, the consequence of the reduced SG content and lack of NETosis for innate immunity remains to be explored.


Asunto(s)
Síndrome de Plaquetas Grises , Animales , Plaquetas , Proteínas Sanguíneas , Gránulos Citoplasmáticos , Síndrome de Plaquetas Grises/genética , Humanos , Ratones , Neutrófilos
7.
Haematologica ; 106(11): 2947-2959, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-33054111

RESUMEN

In X-linked thrombocytopenia with thalassemia (XLTT; OMIM 314050), caused by the mutation p.R216Q in exon 4 of the GATA1 gene, male hemizygous patients display macrothrombocytopenia, bleeding diathesis and a ß-thalassemia trait. Herein, we describe findings in two unrelated Swedish XLTT families with a bleeding tendency exceeding what is expected from the thrombocytopenia. Blood tests revealed low P-PAI-1 and P-factor 5, and elevated S-thrombopoietin levels. Transmission electron microscopy showed diminished numbers of platelet α- and dense granules. The proteomes of isolated blood platelets from 5 male XLTT patients, compared to 5 gender- and age matched controls, were explored. Quantitative mass spectrometry showed alterations of 83 proteins (fold change ≥±1.2, q< .05). Of 46 downregulated proteins, 39 were previously reported to be associated with platelet granules. Reduced protein levels of PTGS1 and SLC35D3 were validated in megakaryocytes of XLTT bone marrow biopsies by immunohistochemistry. Platelet function testing by flow cytometry revealed low dense- and α-granule release and fibrinogen binding in response to ligation of receptors for ADP, the thrombin receptor PAR4 and the collagen receptor GPVI. Significant reductions of a number of α-granule proteins overlapped with a previous platelet proteomics investigation in the inherited macrothrombocytopenia gray platelet syndrome (GPS). In contrast, Ca2+ transporter proteins that facilitate dense granule release were downregulated in XLTT but upregulated in GPS. Ingenuity Pathway Analysis showed altered Coagulation System and Protein Ubiquitination pathways in the XLTT platelets. Collectively, the results revealed protein and functional alterations affecting platelet α- and dense granules in XLTT, probably contributing to bleeding.


Asunto(s)
Síndrome de Plaquetas Grises , Talasemia , Trombocitopenia , Plaquetas , Simulación por Computador , Gránulos Citoplasmáticos , Enfermedades Genéticas Ligadas al Cromosoma X , Síndrome de Plaquetas Grises/genética , Humanos , Masculino , Proteoma
10.
Blood ; 136(17): 1956-1967, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-32693407

RESUMEN

Gray platelet syndrome (GPS) is a rare recessive disorder caused by biallelic variants in NBEAL2 and characterized by bleeding symptoms, the absence of platelet α-granules, splenomegaly, and bone marrow (BM) fibrosis. Due to the rarity of GPS, it has been difficult to fully understand the pathogenic processes that lead to these clinical sequelae. To discern the spectrum of pathologic features, we performed a detailed clinical genotypic and phenotypic study of 47 patients with GPS and identified 32 new etiologic variants in NBEAL2. The GPS patient cohort exhibited known phenotypes, including macrothrombocytopenia, BM fibrosis, megakaryocyte emperipolesis of neutrophils, splenomegaly, and elevated serum vitamin B12 levels. Novel clinical phenotypes were also observed, including reduced leukocyte counts and increased presence of autoimmune disease and positive autoantibodies. There were widespread differences in the transcriptome and proteome of GPS platelets, neutrophils, monocytes, and CD4 lymphocytes. Proteins less abundant in these cells were enriched for constituents of granules, supporting a role for Nbeal2 in the function of these organelles across a wide range of blood cells. Proteomic analysis of GPS plasma showed increased levels of proteins associated with inflammation and immune response. One-quarter of plasma proteins increased in GPS are known to be synthesized outside of hematopoietic cells, predominantly in the liver. In summary, our data show that, in addition to the well-described platelet defects in GPS, there are immune defects. The abnormal immune cells may be the drivers of systemic abnormalities such as autoimmune disease.


Asunto(s)
Gránulos Citoplasmáticos/patología , Heterogeneidad Genética , Síndrome de Plaquetas Grises , Sistema Inmunológico/patología , Fenotipo , Biopsia , Proteínas Sanguíneas/genética , Estudios de Casos y Controles , Estudios de Cohortes , Gránulos Citoplasmáticos/metabolismo , Diagnóstico Diferencial , Frecuencia de los Genes , Estudios de Asociación Genética , Síndrome de Plaquetas Grises/clasificación , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/inmunología , Síndrome de Plaquetas Grises/patología , Humanos , Sistema Inmunológico/fisiología , Enfermedades del Sistema Inmune/sangre , Enfermedades del Sistema Inmune/diagnóstico , Enfermedades del Sistema Inmune/genética , Enfermedades del Sistema Inmune/patología , Mutación
11.
Blood ; 136(6): 715-725, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32384141

RESUMEN

Studies of inherited platelet disorders have provided many insights into platelet development and function. Loss of function of neurobeachin-like 2 (NBEAL2) causes gray platelet syndrome (GPS), where the absence of platelet α-granules indicates NBEAL2 is required for their production by precursor megakaryocytes. The endoplasmic reticulum is a dynamic network that interacts with numerous intracellular vesicles and organelles and plays key roles in their development. The megakaryocyte endoplasmic reticulum is extensive, and in this study we investigated its role in the biogenesis of α-granules by focusing on the membrane-resident trafficking protein SEC22B. Coimmunoprecipitation (co-IP) experiments using tagged proteins expressed in human HEK293 and megakaryocytic immortalized megakaryocyte progenitor (imMKCL) cells established binding of NBEAL2 with SEC22B, and demonstrated that NBEAL2 can simultaneously bind SEC22B and P-selectin. NBEAL2-SEC22B binding was also observed for endogenous proteins in human megakaryocytes using co-IP, and immunofluorescence microscopy detected substantial overlap. SEC22B binding was localized to a region of NBEAL2 spanning amino acids 1798 to 1903, where 2 GPS-associated missense variants have been reported: E1833K and R1839C. NBEAL2 containing either variant did not bind SEC22B coexpressed in HEK293 cells. CRISPR/Cas9-mediated knockout of SEC22B in imMKCL cells resulted in decreased NBEAL2, but not vice versa. Loss of either SEC22B or NBEAL2 expression resulted in failure of α-granule production and reduced granule proteins in imMKCL cells. We conclude that SEC22B is required for α-granule biogenesis in megakaryocytes, and that interactions with SEC22B and P-selectin facilitate the essential role of NBEAL2 in granule development and cargo stability.


Asunto(s)
Proteínas Sanguíneas/fisiología , Gránulos Citoplasmáticos/fisiología , Retículo Endoplásmico/fisiología , Megacariocitos/ultraestructura , Biogénesis de Organelos , Proteínas R-SNARE/fisiología , Sitios de Unión , Proteínas Sanguíneas/deficiencia , Proteínas Sanguíneas/genética , Células Cultivadas , Técnicas de Inactivación de Genes , Síndrome de Plaquetas Grises/genética , Células HEK293 , Humanos , Inmunoprecipitación , Células Progenitoras de Megacariocitos , Megacariocitos/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Mutación Missense , Selectina-P/fisiología , Mapeo de Interacción de Proteínas , Proteínas Recombinantes/metabolismo
12.
Sci Rep ; 9(1): 8333, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31171812

RESUMEN

Zinc (Zn2+) can modulate platelet and coagulation activation pathways, including fibrin formation. Here, we studied the (patho)physiological consequences of abnormal platelet Zn2+ storage and release. To visualize Zn2+ storage in human and mouse platelets, the Zn2+ specific fluorescent dye FluoZin3 was used. In resting platelets, the dye transiently accumulated into distinct cytosolic puncta, which were lost upon platelet activation. Platelets isolated from Unc13d-/- mice, characterized by combined defects of α/δ granular release, showed a markedly impaired Zn2+ release upon activation. Platelets from Nbeal2-/- mice mimicking Gray platelet syndrome (GPS), characterized by primarily loss of the α-granule content, had strongly reduced Zn2+ levels, which was also confirmed in primary megakaryocytes. In human platelets isolated from patients with GPS, Hermansky-Pudlak Syndrome (HPS) and Storage Pool Disease (SPD) altered Zn2+ homeostasis was detected. In turbidity and flow based assays, platelet-dependent fibrin formation was impaired in both Nbeal2-/- and Unc13d-/- mice, and the impairment could be partially restored by extracellular Zn2+. Altogether, we conclude that the release of ionic Zn2+ store from secretory granules upon platelet activation contributes to the procoagulant role of Zn2+ in platelet-dependent fibrin formation.


Asunto(s)
Plaquetas/citología , Proteínas Sanguíneas/genética , Proteínas de la Membrana/genética , Deficiencia de Almacenamiento del Pool Plaquetario/genética , Zinc/metabolismo , Adolescente , Adulto , Animales , Coagulación Sanguínea , Niño , Citosol/metabolismo , Femenino , Fibrina/química , Síndrome de Plaquetas Grises/genética , Voluntarios Sanos , Síndrome de Hermanski-Pudlak/genética , Homeostasis , Humanos , Masculino , Ratones , Ratones Noqueados , Microscopía Confocal , Microscopía Fluorescente , Nefelometría y Turbidimetría , Activación Plaquetaria
13.
Transfusion ; 59(1): 32-38, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30394544

RESUMEN

BACKGROUND: Beyond their role in hemostasis and thrombosis, platelets are also important mediators of inflammation by the release of hundreds of factors stored in their α-granules. Mutations in Nbeal2 cause gray platelet syndrome (GPS), characterized by the lack of platelet α-granules. This study aims to evaluate the immunological (proinflammatory) effects of platelet α-granules. STUDY DESIGN AND METHODS: We performed an experiment using Nbeal2-/- mice, the mouse model of GPS. Systemic inflammation was induced by intravenous injection of lipopolysaccharide (LPS). Inflammatory response was assessed by quantification of inflammatory soluble factors and platelet biological response modifiers. RESULTS: The lack of Nbeal2 (in Nbeal2 -/- mice, compared with controls) significantly reduced the recruitment of circulating neutrophils and monocytes. Moreover, after LPS injection, there was a significant increase in neutrophil and monocyte counts in control animals, compared with Nbeal2 -/- mice. The control of inflammation, evaluated by the production of anti-inflammatory cytokines, appeared to be greater in Nbeal2-/- mice compared with controls. Conversely, the production of certain inflammatory-soluble mediators known to characterize normal platelet secretion, such as soluble CD40 ligand (sCD40L), was decreased under experimental inflammation in Nbeal2 -/- mice. CONCLUSIONS: These results show that α-granules play a direct role in platelet-mediated inflammation balance, confirming the need to further investigate platelet-associated inflammatory pathophysiology and inflammatory adverse events related to blood transfusion.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Síndrome de Plaquetas Grises/inmunología , Lipopolisacáridos/toxicidad , Animales , Proteínas Sanguíneas/genética , Ligando de CD40/genética , Ligando de CD40/metabolismo , Modelos Animales de Enfermedad , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/metabolismo , Inmunoensayo , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética
14.
Arterioscler Thromb Vasc Biol ; 38(8): 1772-1784, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29930006

RESUMEN

Objective- Nbeal2-/- mice, a model of human gray platelet syndrome, have reduced neutrophil granularity and impaired host defense against systemic Staphylococcus aureus infection. We here aimed to study the role of Nbeal2 deficiency in both leukocytes and platelets during gram-negative pneumonia and sepsis. Approach and Results- We studied the role of Nbeal2 in platelets and leukocytes during murine pneumonia and sepsis by Klebsiella pneumoniae. Apart from platelet α-granule deficiency and reduced neutrophil granularity, also monocyte granularity was reduced in Nbeal2-/- mice, whereas plasma levels of MPO (myeloperoxidase), elastase, NGAL (neutrophil gelatinase-associated lipocalin), and MMP-9 (matrix metalloproteinase 9), and leukocyte CD11b expression were increased. Nbeal2-/- leukocytes showed unaltered in vitro antibacterial response and phagocytosis capacity against Klebsiella, and unchanged reactive nitrogen species and cytokine production. Also during Klebsiella pneumonia and sepsis, Nbeal2-/- mice had similar bacterial growth in lung and distant body sites, with enhanced leukocyte migration to the bronchoalveolar space. Despite similar infection-induced inflammation, organ damage was increased in Nbeal2-/- mice, which was also seen during endotoxemia. Platelet-specific Nbeal2 deficiency did not influence leukocyte functions, indicating that Nbeal2 directly modifies leukocytes. Transfusion of Nbeal2-/- but not of Nbeal2+/+ platelets into thrombocytopenic mice was associated with bleeding in the lung but similar host defense, pointing at a role for platelet α-granules in maintaining vascular integrity but not host defense during Klebsiella pneumosepsis. Conclusions- These data show that Nbeal2 deficiency-resulting in gray platelet syndrome-affects platelets, neutrophils, and monocytes, with intact host defense but increased organ damage during gram-negative pneumosepsis.


Asunto(s)
Plaquetas/metabolismo , Proteínas Sanguíneas/deficiencia , Síndrome de Plaquetas Grises/metabolismo , Infecciones por Klebsiella/metabolismo , Klebsiella pneumoniae/patogenicidad , Insuficiencia Multiorgánica/metabolismo , Neumonía Bacteriana/metabolismo , Sepsis/metabolismo , Animales , Plaquetas/microbiología , Proteínas Sanguíneas/genética , Antígeno CD11b/sangre , Modelos Animales de Enfermedad , Femenino , Síndrome de Plaquetas Grises/sangre , Síndrome de Plaquetas Grises/genética , Interacciones Huésped-Patógeno , Infecciones por Klebsiella/sangre , Infecciones por Klebsiella/genética , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/crecimiento & desarrollo , Lipocalina 2/sangre , Masculino , Metaloproteinasa 9 de la Matriz/sangre , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Monocitos/microbiología , Insuficiencia Multiorgánica/sangre , Insuficiencia Multiorgánica/genética , Insuficiencia Multiorgánica/microbiología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Elastasa Pancreática/sangre , Peroxidasa/sangre , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Transfusión de Plaquetas , Neumonía Bacteriana/sangre , Neumonía Bacteriana/genética , Neumonía Bacteriana/microbiología , Sepsis/sangre , Sepsis/genética , Sepsis/microbiología
15.
Platelets ; 29(6): 632-635, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29869935

RESUMEN

Homozygosity/compound heterozygosity for loss of function mutations in neurobeachin-like 2 (NBEAL2) is causative for Gray platelet syndrome (GPS; MIM #139090), characterized by thrombocytopenia and large platelets lacking α-granules and cargo. Most GPS-associated NBEAL2 mutations generate nonsense codons; frameshifts causing premature translation termination and/or changes in mRNA splicing have also been observed. Data regarding NBEAL2 protein expression in GPS patients is limited. We observed absence of NBEAL2 in platelets from GPS patients with 3 different genotypes, and reduced/truncated platelet NBEAL2 has been reported for others. GPS is commonly associated with mild bleeding, but lifethreatening bleeding has been reported in some cases. A common long-term complication in GPS patients is myelofibrosis; splenomegaly is less common but sometimes of sufficient severity to merit splenectomy. Like GPS patients, mice lacking NBEAL2 expression exhibit macrothrombocytopenia, deficiency of platelet α-granules, splenomegaly, myelofibrosis, impaired platelet function and abnormalities in megakaryocyte development. Animal studies have also reported impaired platelet function in vivo using laser injury and thrombo-inflammation models. NBEAL2 is a large gene with 54 exons, and several putative functional domains have been identified in NBEAL2, including PH (pleckstrin homology) and BEACH (beige and Chediak-Higashi) domains shared with other members of a protein family that includes LYST and LRBA, also expressed by hematopoietic cells. Potential NBEAL2-interacting proteins have recently been identified, and it is expected that current and future efforts will reveal the cellular mechanisms by which NBEAL2 facilitates platelet development and supports hemostatic function.


Asunto(s)
Plaquetas/metabolismo , Proteínas Sanguíneas/genética , Síndrome de Plaquetas Grises/genética , Mutación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Proteínas Sanguíneas/metabolismo , Niño , Preescolar , Femenino , Síndrome de Plaquetas Grises/sangre , Síndrome de Plaquetas Grises/metabolismo , Hemorragia/sangre , Hemorragia/genética , Hemorragia/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
16.
Platelets ; 29(3): 288-291, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-28504079

RESUMEN

Gray platelet syndrome (GPS) is a rare, inherited bleeding disorder characterized by the defect of platelet α-granule. Up to date, these are only four studies identifying NBEAL2 gene correlated with GPS. In the current report, we present a Chinese GPS patient who had severe bleeding tendency, abnormalities of platelet functions, and absence of platelet α-granules. Genomic DNA sequencing for the patient identified a nonsense mutation (g.27713C>A) of NBEAL2 gene (g.NG__031914.1) resulting in a premature protein (p.Glu1726*). In comparison with the reported patients, we conclude that homozygotes with nonsense or deletion mutation leading to a premature stop codon exhibit more serious bleeding problem than those with missense mutations.


Asunto(s)
Proteínas Sanguíneas/genética , Codón sin Sentido , Síndrome de Plaquetas Grises/complicaciones , Síndrome de Plaquetas Grises/genética , Hemorragia/diagnóstico , Hemorragia/etiología , Adulto , Biomarcadores , Plaquetas/metabolismo , Plaquetas/ultraestructura , Gránulos Citoplasmáticos/metabolismo , Análisis Mutacional de ADN , Femenino , Homocigoto , Humanos , Linaje , Agregación Plaquetaria/efectos de los fármacos , Pruebas de Función Plaquetaria , Análisis de Secuencia de ADN , Índice de Severidad de la Enfermedad
17.
J Immunol ; 199(8): 2948-2957, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28887433

RESUMEN

The neurobeachin-like 2 protein (Nbeal2) belongs to the family of beige and Chediak-Higashi (BEACH) domain proteins. Loss-of-function mutations in the human NBEAL2 gene or Nbeal2 deficiency in mice cause gray platelet syndrome, a bleeding disorder characterized by macrothrombocytopenia, splenomegaly, and paucity of α-granules in megakaryocytes and platelets. We found that in mast cells, Nbeal2 regulates the activation of the Shp1-STAT5 signaling axis and the composition of the c-Kit/STAT signalosome. Furthermore, Nbeal2 mediates granule formation and restricts the expression of the transcription factors, IRF8, GATA2, and MITF as well as of the cell-cycle inhibitor p27, which are essential for mast cell differentiation, proliferation, and cytokine production. These data demonstrate the relevance of Nbeal2 in mast cells above and beyond granule biosynthesis.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Síndrome de Plaquetas Grises/genética , Mastocitos/fisiología , Megacariocitos/fisiología , Animales , Proteínas Sanguíneas/genética , Ciclo Celular , Células Cultivadas , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Hemorragia , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Ratones , Ratones Noqueados , Mutación/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Esplenomegalia , Trombocitopenia
18.
J Clin Invest ; 127(9): 3521-3526, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28783043

RESUMEN

Mutations in the human NBEAL2 gene cause gray platelet syndrome (GPS), a bleeding diathesis characterized by a lack of α granules in platelets. The functions of the NBEAL2 protein have not been explored outside platelet biology, but there are reports of increased frequency of infection and abnormal neutrophil morphology in patients with GPS. We therefore investigated the role of NBEAL2 in immunity by analyzing the phenotype of Nbeal2-deficient mice. We found profound abnormalities in the Nbeal2-deficient immune system, particularly in the function of neutrophils and NK cells. Phenotyping of Nbeal2-deficient neutrophils showed a severe reduction in granule contents across all granule subsets. Despite this, Nbeal2-deficient neutrophils had an enhanced phagocyte respiratory burst relative to Nbeal2-expressing neutrophils. This respiratory burst was associated with increased expression of cytosolic components of the NADPH oxidase complex. Nbeal2-deficient NK cells were also dysfunctional and showed reduced degranulation. These abnormalities were associated with increased susceptibility to both bacterial (Staphylococcus aureus) and viral (murine CMV) infection in vivo. These results define an essential role for NBEAL2 in mammalian immunity.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Células Asesinas Naturales/metabolismo , Mutación , Neutrófilos/metabolismo , Animales , Plaquetas/metabolismo , Proteínas Sanguíneas/genética , Citosol/metabolismo , Síndrome de Plaquetas Grises/genética , Humanos , Sistema Inmunológico , Inmunofenotipificación , Ratones , Ratones Noqueados , NADPH Oxidasas/metabolismo , Fenotipo , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus
20.
Sci Rep ; 6: 23213, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26987485

RESUMEN

The Gray Platelet Syndrome (GPS) is a rare inherited bleeding disorder characterized by deficiency of platelet α-granules, macrothrombocytopenia and marrow fibrosis. The autosomal recessive form of GPS is linked to loss of function mutations in NBEAL2, which is predicted to regulate granule trafficking in megakaryocytes, the platelet progenitors. We report the first analysis of cultured megakaryocytes from GPS patients with NBEAL2 mutations. Megakaryocytes cultured from peripheral blood or bone marrow hematopoietic progenitor cells from four patients were used to investigate megakaryopoiesis, megakaryocyte morphology and platelet formation. In vitro differentiation of megakaryocytes was normal, whereas we observed deficiency of megakaryocyte α-granule proteins and emperipolesis. Importantly, we first demonstrated that platelet formation by GPS megakaryocytes was severely affected, a defect which might be the major cause of thrombocytopenia in patients. These results demonstrate that cultured megakaryocytes from GPS patients provide a valuable model to understand the pathogenesis of GPS in humans.


Asunto(s)
Plaquetas/citología , Proteínas Sanguíneas/genética , Síndrome de Plaquetas Grises/patología , Megacariocitos/citología , Anciano , Plaquetas/metabolismo , Calcio/metabolismo , Diferenciación Celular , Células Cultivadas , Niño , Emperipolesis , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/metabolismo , Humanos , Masculino , Megacariocitos/metabolismo , Megacariocitos/patología , Modelos Biológicos , Mutación , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...