Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 389(6): 527-539, 2023 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-37342957

RESUMEN

BACKGROUND: Increasing evidence links genetic defects affecting actin-regulatory proteins to diseases with severe autoimmunity and autoinflammation, yet the underlying molecular mechanisms are poorly understood. Dedicator of cytokinesis 11 (DOCK11) activates the small Rho guanosine triphosphatase (GTPase) cell division cycle 42 (CDC42), a central regulator of actin cytoskeleton dynamics. The role of DOCK11 in human immune-cell function and disease remains unknown. METHODS: We conducted genetic, immunologic, and molecular assays in four patients from four unrelated families who presented with infections, early-onset severe immune dysregulation, normocytic anemia of variable severity associated with anisopoikilocytosis, and developmental delay. Functional assays were performed in patient-derived cells, as well as in mouse and zebrafish models. RESULTS: We identified rare, X-linked germline mutations in DOCK11 in the patients, leading to a loss of protein expression in two patients and impaired CDC42 activation in all four patients. Patient-derived T cells did not form filopodia and showed abnormal migration. In addition, the patient-derived T cells, as well as the T cells from Dock11-knockout mice, showed overt activation and production of proinflammatory cytokines that were associated with an increased degree of nuclear translocation of nuclear factor of activated T cell 1 (NFATc1). Anemia and aberrant erythrocyte morphologic features were recapitulated in a newly generated dock11-knockout zebrafish model, and anemia was amenable to rescue on ectopic expression of constitutively active CDC42. CONCLUSIONS: Germline hemizygous loss-of-function mutations affecting the actin regulator DOCK11 were shown to cause a previously unknown inborn error of hematopoiesis and immunity characterized by severe immune dysregulation and systemic inflammation, recurrent infections, and anemia. (Funded by the European Research Council and others.).


Asunto(s)
Actinas , Anemia , Factores de Intercambio de Guanina Nucleótido , Inflamación , Animales , Humanos , Ratones , Actinas/genética , Actinas/metabolismo , Anemia/etiología , Anemia/genética , Modelos Animales de Enfermedad , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Hematopoyesis , Inflamación/etiología , Inflamación/genética , Pez Cebra/genética , Pez Cebra/metabolismo
2.
Blood ; 141(20): 2520-2536, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-36735910

RESUMEN

Metabolic programs contribute to hematopoietic stem and progenitor cell (HSPC) fate, but it is not known whether the metabolic regulation of protein synthesis controls HSPC differentiation. Here, we show that SLC7A1/cationic amino acid transporter 1-dependent arginine uptake and its catabolism to the polyamine spermidine control human erythroid specification of HSPCs via the activation of the eukaryotic translation initiation factor 5A (eIF5A). eIF5A activity is dependent on its hypusination, a posttranslational modification resulting from the conjugation of the aminobutyl moiety of spermidine to lysine. Notably, attenuation of hypusine synthesis in erythroid progenitors, by the inhibition of deoxyhypusine synthase, abrogates erythropoiesis but not myeloid cell differentiation. Proteomic profiling reveals mitochondrial translation to be a critical target of hypusinated eIF5A, and accordingly, progenitors with decreased hypusine activity exhibit diminished oxidative phosphorylation. This affected pathway is critical for eIF5A-regulated erythropoiesis, as interventions augmenting mitochondrial function partially rescue human erythropoiesis under conditions of attenuated hypusination. Levels of mitochondrial ribosomal proteins (RPs) were especially sensitive to the loss of hypusine, and we find that the ineffective erythropoiesis linked to haploinsufficiency of RPS14 in chromosome 5q deletions in myelodysplastic syndrome is associated with a diminished pool of hypusinated eIF5A. Moreover, patients with RPL11-haploinsufficient Diamond-Blackfan anemia as well as CD34+ progenitors with downregulated RPL11 exhibit a markedly decreased hypusination in erythroid progenitors, concomitant with a loss of mitochondrial metabolism. Thus, eIF5A-dependent protein synthesis regulates human erythropoiesis, and our data reveal a novel role for RPs in controlling eIF5A hypusination in HSPCs, synchronizing mitochondrial metabolism with erythroid differentiation.


Asunto(s)
Proteómica , Espermidina , Humanos , Espermidina/metabolismo , Factores de Iniciación de Péptidos/genética , Diferenciación Celular , Factor 5A Eucariótico de Iniciación de Traducción
3.
Blood ; 139(21): 3111-3126, 2022 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-35213692

RESUMEN

The congenital bone marrow failure syndrome Diamond-Blackfan anemia (DBA) is typically associated with variants in ribosomal protein (RP) genes impairing erythroid cell development. Here we report multiple individuals with biallelic HEATR3 variants exhibiting bone marrow failure, short stature, facial and acromelic dysmorphic features, and intellectual disability. These variants destabilize a protein whose yeast homolog is known to synchronize the nuclear import of RPs uL5 (RPL11) and uL18 (RPL5), which are both critical for producing ribosomal subunits and for stabilizing the p53 tumor suppressor when ribosome biogenesis is compromised. Expression of HEATR3 variants or repression of HEATR3 expression in primary cells, cell lines of various origins, and yeast models impairs growth, differentiation, pre-ribosomal RNA processing, and ribosomal subunit formation reminiscent of DBA models of large subunit RP gene variants. Consistent with a role of HEATR3 in RP import, HEATR3-depleted cells or patient-derived fibroblasts display reduced nuclear accumulation of uL18. Hematopoietic progenitor cells expressing HEATR3 variants or small-hairpin RNAs knocking down HEATR3 synthesis reveal abnormal acceleration of erythrocyte maturation coupled to severe proliferation defects that are independent of p53 activation. Our study uncovers a new pathophysiological mechanism leading to DBA driven by biallelic HEATR3 variants and the destabilization of a nuclear import protein important for ribosome biogenesis.


Asunto(s)
Anemia de Diamond-Blackfan , Proteínas , Transporte Activo de Núcleo Celular/genética , Anemia de Diamond-Blackfan/metabolismo , Humanos , Mutación , Proteínas/genética , Proteínas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
4.
Prenat Diagn ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38923613

RESUMEN

INTRODUCTION: Acute fetal leukemia is rare and characterized by a very poor prognosis. The aims of this study were to identify cases of acute fetal leukemia and to describe ultrasound and fetopathological findings that should lead to a suspicion of this diagnosis, as well as the investigations required to confirm it. METHODS: A national retrospective study was conducted. Clinical data, prenatal ultrasounds and postmortem findings of fetal acute leukemia cases were collected and analyzed. RESULTS: We collected seven cases: four in utero fetal deaths, two neonatal deaths and one termination of pregnancy. Prenatal ultrasounds showed fetal hydrops (42.9%) associated with hepatosplenomegaly (100%). In addition, post-mortem examination (n = 6) suggested a Down syndrome in one case and showed other organomegaly (83.3%) due to blastic infiltration, mainly in the liver, along with extrahepatic multivisceral hematopoiesis. Immunostainings allowed to specify the type of leukemia (71.4%). In one case, diagnosis was made on blood smear and flow cytometry was performed on fresh blood samples. All cases corresponded to acute myeloid leukemia. Karyotype was abnormal in 4 cases (66.7%), including one free trisomy 21, two mosaic trisomy 21 and one chromosome 15 deletion. GATA1 gene mutations were identified in two cases: one mosaic trisomy 21 and one with normal karyotype. CONCLUSION: Any hepatosplenomegaly associated with fetal hydrops and a negative immune, infectious, and metabolic work-up, should suggest acute fetal leukemia and prompt additional investigations.

5.
Blood Cells Mol Dis ; 103: 102780, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37516005

RESUMEN

We report here an instructive case referred at 16 months-old for exploration of hemolysis without anemia (compensated anemia with reticulocytosis). The biology tests confirmed the hemolysis with increased total and indirect bilirubin. The usual hemolysis diagnosis tests were normal (DAT, G6PD, PK, Hb electrophoresis) except cytology and ektacytometry suggesting an association of multiple red blood cell (RBC) membrane disorders. This led us to propose a molecular screening analysis using targeted-Next Generation Sequencing (t-NGS) with a capture technique on 93 genes involved in RBC and erythropoiesis defects. We identified 4 missense heterozygous allelic variations, all of them were described without any significance (VUS) in the SLC4A1, RhAG, PIEZO1 and SPTB genes. The study of the familial cosegregation and research functional tests allowed to decipher the role of at least two by two genes in the phenotype and the hemolytic disease of this young patient. Specialized t-NGS panel (or virtual exome/genome sequencing) in a disease-referent laboratory and the motivated collaboration of clinicians, biologists and scientists should be the gold standard for improving the diagnosis of the patients affected with RBC diseases or rare inherited anemias.


Asunto(s)
Enfermedades Hematológicas , Esferocitosis Hereditaria , Humanos , Esferocitosis Hereditaria/diagnóstico , Esferocitosis Hereditaria/genética , Espectrina/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Hemólisis , Mutación , Eritrocitos , Fenotipo , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Canales Iónicos/genética
6.
Am J Hum Genet ; 105(5): 1040-1047, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31630789

RESUMEN

Variants in genes encoding ribosomal proteins have thus far been associated with Diamond-Blackfan anemia, a rare inherited bone marrow failure, and isolated congenital asplenia. Here, we report one de novo missense variant and three de novo splice variants in RPL13, which encodes ribosomal protein RPL13 (also called eL13), in four unrelated individuals with a rare bone dysplasia causing severe short stature. The three splice variants (c.477+1G>T, c.477+1G>A, and c.477+2 T>C) result in partial intron retention, which leads to an 18-amino acid insertion. In contrast to observations from Diamond-Blackfan anemia, we detected no evidence of significant pre-rRNA processing disturbance in cells derived from two affected individuals. Consistently, we showed that the insertion-containing protein is stably expressed and incorporated into 60S subunits similar to the wild-type protein. Erythroid proliferation in culture and ribosome profile on sucrose gradient are modified, suggesting a change in translation dynamics. We also provide evidence that RPL13 is present at high levels in chondrocytes and osteoblasts in mouse growth plates. Taken together, we show that the identified RPL13 variants cause a human ribosomopathy defined by a rare skeletal dysplasia, and we highlight the role of this ribosomal protein in bone development.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Enanismo/genética , Mutación Missense/genética , Proteínas de Neoplasias/genética , Proteínas Ribosómicas/genética , Anemia de Diamond-Blackfan/genética , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
7.
Blood ; 136(11): 1262-1273, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32702755

RESUMEN

Diamond-Blackfan anemia (DBA) was the first ribosomopathy described and is a constitutional inherited bone marrow failure syndrome. Erythroblastopenia is the major characteristic of the disease, which is a model for ribosomal diseases, related to a heterozygous allelic variation in 1 of the 20 ribosomal protein genes of either the small or large ribosomal subunit. The salient feature of classical DBA is a defect in ribosomal RNA maturation that generates nucleolar stress, leading to stabilization of p53 and activation of its targets, resulting in cell-cycle arrest and apoptosis. Although activation of p53 may not explain all aspects of DBA erythroid tropism, involvement of GATA1/HSP70 and globin/heme imbalance, with an excess of the toxic free heme leading to reactive oxygen species production, account for defective erythropoiesis in DBA. Despite significant progress in defining the molecular basis of DBA and increased understanding of the mechanistic basis for DBA pathophysiology, progress in developing new therapeutic options has been limited. However, recent advances in gene therapy, better outcomes with stem cell transplantation, and discoveries of putative new drugs through systematic drug screening using large chemical libraries provide hope for improvement.


Asunto(s)
Anemia de Diamond-Blackfan , Anomalías Múltiples/genética , Adenosina Desaminasa/sangre , Adenosina Desaminasa/genética , Anemia de Diamond-Blackfan/diagnóstico , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/metabolismo , Anemia de Diamond-Blackfan/terapia , Preescolar , Anomalías Congénitas/genética , Diagnóstico Diferencial , Manejo de la Enfermedad , Resistencia a Medicamentos , Eritrocitos/enzimología , Retardo del Crecimiento Fetal/etiología , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/fisiología , Heterogeneidad Genética , Terapia Genética , Glucocorticoides/uso terapéutico , Proteínas HSP70 de Choque Térmico/metabolismo , Trasplante de Células Madre Hematopoyéticas , Humanos , Lactante , Recién Nacido , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/genética , Modelos Biológicos , Mutación , Síndromes Neoplásicos Hereditarios/genética , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/fisiología , Proteína p53 Supresora de Tumor/fisiología
8.
Haematologica ; 107(4): 958-965, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33882639

RESUMEN

Despite its high prevalence in children with sickle cell anemia (SCA), the pathophysiology of silent cerebral infarcts (SCI) remains elusive. The main objective of this study was to explore the respective roles of major determinants of brain perfusion in SCA children with no past or current history of intracranial or extracranial vasculopathy. We used a multimodal approach based notably on perfusion imaging arterial spin labeling (ASL) magnetic resonance imaging (MRI) and near infra-red spectroscopy (NIRS), as well as biomarkers reflecting blood rheology and endothelial activation. Out of 59 SCA patients (mean age 11.4±3.9 yrs), eight (13%) had a total of 12 SCI. Children with SCI had a distinctive profile characterized by decreased blood pressure, impaired blood rheology, increased P-selectin levels, and marked anemia. Although ASL perfusion and oximetry values did not differ between groups, comparison of biological and clinical parameters according to the level of perfusion categorized in terciles showed an independent association between high perfusion and increased sP-selectin, decreased red blood cell deformability, low hemoglobin F level, increased blood viscosity and no a-thalassemia deletion. NIRS measurements did not yield additional novel results. Altogether, these findings argue for early MRI detection of SCI in children with no identified vasculopathy and suggest a potential role for ASL as an additional screening tool. Early treatment targeting hemolysis, anemia and endothelial dysfunction should reduce the risk of this under diagnosed and serious complication.


Asunto(s)
Anemia de Células Falciformes , Lesiones Encefálicas , Adolescente , Lesiones Encefálicas/complicaciones , Infarto Cerebral , Niño , Hemólisis , Humanos , Imagen por Resonancia Magnética
9.
Nucleic Acids Res ; 48(2): 770-787, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31799629

RESUMEN

Variants in ribosomal protein (RP) genes drive Diamond-Blackfan anemia (DBA), a bone marrow failure syndrome that can also predispose individuals to cancer. Inherited and sporadic RP gene variants are also linked to a variety of phenotypes, including malignancy, in individuals with no anemia. Here we report an individual diagnosed with DBA carrying a variant in the 5'UTR of RPL9 (uL6). Additionally, we report two individuals from a family with multiple cancer incidences carrying a RPL9 missense variant. Analysis of cells from these individuals reveals that despite the variants both driving pre-rRNA processing defects and 80S monosome reduction, the downstream effects are remarkably different. Cells carrying the 5'UTR variant stabilize TP53 and impair the growth and differentiation of erythroid cells. In contrast, ribosomes incorporating the missense variant erroneously read through UAG and UGA stop codons of mRNAs. Metabolic profiles of cells carrying the 5'UTR variant reveal an increased metabolism of amino acids and a switch from glycolysis to gluconeogenesis while those of cells carrying the missense variant reveal a depletion of nucleotide pools. These findings indicate that variants in the same RP gene can drive similar ribosome biogenesis defects yet still have markedly different downstream consequences and clinical impacts.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Procesamiento Postranscripcional del ARN/genética , Proteínas Ribosómicas/genética , Ribosomas/genética , Regiones no Traducidas 5'/genética , Adolescente , Adulto , Anemia de Diamond-Blackfan/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Niño , Células Eritroides , Femenino , Humanos , Masculino , Mutación/genética , Precursores del ARN/genética , ARN Mensajero/genética , Secuenciación del Exoma
10.
Am J Hum Genet ; 103(6): 930-947, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30503522

RESUMEN

Diamond-Blackfan anemia (DBA) is a rare bone marrow failure disorder that affects 7 out of 1,000,000 live births and has been associated with mutations in components of the ribosome. In order to characterize the genetic landscape of this heterogeneous disorder, we recruited a cohort of 472 individuals with a clinical diagnosis of DBA and performed whole-exome sequencing (WES). We identified relevant rare and predicted damaging mutations for 78% of individuals. The majority of mutations were singletons, absent from population databases, predicted to cause loss of function, and located in 1 of 19 previously reported ribosomal protein (RP)-encoding genes. Using exon coverage estimates, we identified and validated 31 deletions in RP genes. We also observed an enrichment for extended splice site mutations and validated their diverse effects using RNA sequencing in cell lines obtained from individuals with DBA. Leveraging the size of our cohort, we observed robust genotype-phenotype associations with congenital abnormalities and treatment outcomes. We further identified rare mutations in seven previously unreported RP genes that may cause DBA, as well as several distinct disorders that appear to phenocopy DBA, including nine individuals with biallelic CECR1 mutations that result in deficiency of ADA2. However, no new genes were identified at exome-wide significance, suggesting that there are no unidentified genes containing mutations readily identified by WES that explain >5% of DBA-affected case subjects. Overall, this report should inform not only clinical practice for DBA-affected individuals, but also the design and analysis of rare variant studies for heterogeneous Mendelian disorders.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Adolescente , Niño , Preescolar , Estudios de Cohortes , Exoma/genética , Exones/genética , Femenino , Eliminación de Gen , Estudios de Asociación Genética/métodos , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Mutación/genética , Fenotipo , Proteínas Ribosómicas/genética , Ribosomas/genética , Análisis de Secuencia de ARN/métodos , Secuenciación del Exoma/métodos
11.
Blood ; 133(12): 1358-1370, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30700418

RESUMEN

Diamond-Blackfan anemia (DBA) is a congenital erythroblastopenia that is characterized by a blockade in erythroid differentiation related to impaired ribosome biogenesis. DBA phenotype and genotype are highly heterogeneous. We have previously identified 2 in vitro erythroid cell growth phenotypes for primary CD34+ cells from DBA patients and following short hairpin RNA knockdown of RPS19, RPL5, and RPL11 expression in normal human CD34+ cells. The haploinsufficient RPS19 in vitro phenotype is less severe than that of 2 other ribosomal protein (RP) mutant genes. We further documented that proteasomal degradation of HSP70, the chaperone of GATA1, is a major contributor to the defect in erythroid proliferation, delayed erythroid differentiation, increased apoptosis, and decreased globin expression, which are all features of the RPL5 or RPL11 DBA phenotype. In the present study, we explored the hypothesis that an imbalance between globin and heme synthesis may be involved in pure red cell aplasia of DBA. We identified disequilibrium between the globin chain and the heme synthesis in erythroid cells of DBA patients. This imbalance led to accumulation of excess free heme and increased reactive oxygen species production that was more pronounced in cells of the RPL5 or RPL11 phenotype. Strikingly, rescue experiments with wild-type HSP70 restored GATA1 expression levels, increased globin synthesis thereby reducing free heme excess and resulting in decreased apoptosis of DBA erythroid cells. These results demonstrate the involvement of heme in DBA pathophysiology and a major role of HSP70 in the control of balanced heme/globin synthesis.


Asunto(s)
Anemia de Diamond-Blackfan/patología , Diferenciación Celular , Células Eritroides/patología , Factor de Transcripción GATA1/metabolismo , Globinas/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Hemo/metabolismo , Anemia de Diamond-Blackfan/metabolismo , Proliferación Celular , Células Cultivadas , Células Eritroides/metabolismo , Femenino , Estudios de Seguimiento , Haploinsuficiencia , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Fenotipo , Pronóstico , ARN Interferente Pequeño , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo
12.
Haematologica ; 106(5): 1303-1310, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32241839

RESUMEN

Diamond Blackfan anemia (DBA) is predominantly an autosomal dominant inherited red cell aplasia primarily caused by pathogenic germline variants in ribosomal protein genes. DBA due to pathogenic RPL35A variants has been associated with large 3q29 deletions and phenotypes not common in DBA. We conducted a multi-institutional genotype-phenotype study of 45 patients with DBA associated with pathogenic RPL35A germline variants and curated the variant data on 21 additional cases from the literature. Genotype-phenotype analyses were conducted comparing patients with large deletions versus all other pathogenic variants in RPL35A. Twenty-two of the 45 cases had large deletions in RPL35A. After adjusting for multiple tests, a statistically significant association was observed between patients with a large deletion and steroid-resistant anemia, neutropenia, craniofacial abnormalities, chronic gastrointestinal problems, and intellectual disabilities (p<0.01) compared with all other pathogenic variants. Non-large deletion pathogenic variants were spread across RPL35A with no apparent hot spot and 56% of the individual family variants were observed more than once. In this, the largest known study of DBA patients with pathogenic RPL35A variants, we determined that patients with large deletions have a more severe phenotype that is clinically different from those with non-large deletion variants. Genes of interest also deleted in the 3q29 region that could be associated with some of these phenotypic features include LMLN and IQCG. Management of DBA due to large RPL35A deletions may be challenging due to complex problems and require comprehensive assessments by multiple specialists including immunologic, gastrointestinal, and developmental evaluations to provide optimal multidisciplinary care.


Asunto(s)
Anemia de Diamond-Blackfan , Anemia de Diamond-Blackfan/genética , Estudios de Asociación Genética , Humanos , Mutación , Fenotipo , Proteínas Ribosómicas/genética
13.
Fetal Diagn Ther ; 47(2): 156-164, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31505487

RESUMEN

BACKGROUND: Rare causes of fetal anemia requiring intrauterine transfusion (IUT) are challenging for fetal medicine specialists. OBJECTIVES: The aim of this study was to describe the perinatal patterns and prognosis in a consecutive series of fetuses transfused for fetal anemia of rare or unknown etiology, and to propose a protocol of investigation for fetal anemia of undetermined cause and for the management of subsequent pregnancies. METHOD: We conducted a retrospective descriptive study on fetuses transfused for severe anemia of rare or unknown etiology managed in our national referral center (Centre National de Référence d'Hémobiologie Périnatale) and born between 2010 and 2017. RESULTS: During the study period, 584 IUT were performed in 253 fetuses. Among those IUT, 23 (3.9%) were performed for a rare or unknown cause of anemia in 13 fetuses (5.1% of transfused fetuses). The median gestational age at diagnosis was 26 weeks of gestation (WG; range 21-33). Hemoglobin levels ranged from 1.6 to 9.1 g/dL (0.18-0.83 multiples of median) before the first IUT. The fetuses received between 1 and 6 IUT (39% received at least 2 IUT). The definitive etiologies for central anemia were: congenital syphilis, neonatal poikilocytosis, type II congenital dyserythropoietic anemia (CDA), and neonatal hemochromatosis. There was 1 case with suspected type I CDA and 1 with suspected Diamond-Blackfan anemia. There was 1 case of peripheral anemia, secondary to cerebral hemorrhages of different ages, related to a variant of the COL4A1 gene. In 6 fetuses corresponding to 4 mothers, no precise diagnosis was found despite a complete workup. In our series, there were 8 live births, 4 terminations of pregnancy, and 1 intrauterine fetal death. CONCLUSIONS: Fetal anemia of rare or unknown diagnosis represents 5% of all transfused fetuses in our cohort. Fetal and neonatal anemias can be recurrent in further pregnancies, with variable expressivity.


Asunto(s)
Anemia/terapia , Transfusión de Sangre Intrauterina , Enfermedades Fetales/terapia , Aborto Inducido , Anemia/sangre , Anemia/diagnóstico , Anemia/etiología , Biomarcadores/sangre , Transfusión de Sangre Intrauterina/efectos adversos , Femenino , Muerte Fetal/etiología , Enfermedades Fetales/sangre , Enfermedades Fetales/diagnóstico , Enfermedades Fetales/etiología , Hemoglobina Fetal/metabolismo , Edad Gestacional , Humanos , Nacimiento Vivo , Embarazo , Estudios Retrospectivos , Factores de Riesgo , Resultado del Tratamiento
14.
Am J Hematol ; 94(6): 667-677, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30916803

RESUMEN

MYH9-related disease (MYH9-RD) is a rare, autosomal dominant disorder caused by mutations in MYH9, the gene encoding the actin-activated motor protein non-muscle myosin IIA (NMIIA). MYH9-RD patients suffer from bleeding syndromes, progressive kidney disease, deafness, and/or cataracts, but the impact of MYH9 mutations on other NMIIA-expressing tissues remains unknown. In human red blood cells (RBCs), NMIIA assembles into bipolar filaments and binds to actin filaments (F-actin) in the spectrin-F-actin membrane skeleton to control RBC biconcave disk shape and deformability. Here, we tested the effects of MYH9 mutations in different NMIIA domains (motor, coiled-coil rod, or non-helical tail) on RBC NMIIA function. We found that MYH9-RD does not cause clinically significant anemia and that patient RBCs have normal osmotic deformability as well as normal membrane skeleton composition and micron-scale distribution. However, analysis of complete blood count data and peripheral blood smears revealed reduced hemoglobin content and elongated shapes, respectively, of MYH9-RD RBCs. Patients with mutations in the NMIIA motor domain had the highest numbers of elongated RBCs. Patients with mutations in the motor domain also had elevated association of NMIIA with F-actin at the RBC membrane. Our findings support a central role for motor domain activity in NMIIA regulation of RBC shape and define a new sub-clinical phenotype of MYH9-RD.


Asunto(s)
Actinas , Membrana Eritrocítica , Eritrocitos Anormales , Pérdida Auditiva Sensorineural , Mutación , Cadenas Pesadas de Miosina , Trombocitopenia/congénito , Actinas/genética , Actinas/metabolismo , Membrana Eritrocítica/genética , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/patología , Eritrocitos Anormales/metabolismo , Eritrocitos Anormales/patología , Femenino , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/metabolismo , Pérdida Auditiva Sensorineural/patología , Humanos , Masculino , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombocitopenia/patología
15.
Mol Ther ; 26(10): 2523-2532, 2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30077612

RESUMEN

Macrophage migration inhibitory factor (MIF) is elevated in patients with acute kidney injury (AKI) and is suggested as a potential predictor for renal replacement therapy in AKI. In this study, we found that MIF also plays a pathogenic role and is a therapeutic target for AKI. In a cisplatin-induced AKI mouse model, elevated plasma MIF correlated with increased serum creatinine and the severity of renal inflammation and tubular necrosis, whereas deletion of MIF protected the kidney from cisplatin-induced AKI by largely improving renal functional and histological injury, and suppressing renal inflammation including upregulation of cytokines such as interleukin (IL)-1ß, tumor necrosis factor-alpha (TNF-α), IL-6, inducible nitric oxide synthase (iNOS), MCP-1, IL-8, and infiltration of macrophages, neutrophils, and T cells. We next developed a novel therapeutic strategy for AKI by blocking the endogenous MIF with an MIF inhibitor, ribosomal protein S19 (RPS19). Similar to the MIF-knockout mice, treatment with RPS19, but not the mutant RPS19, suppressed cisplatin-induced AKI. Mechanistically, we found that both genetic knockout and pharmacological inhibition of MIF protected against AKI by inactivating the CD74-nuclear factor κB (NF-κB) signaling. In conclusion, MIF is pathogenic in cisplatin-induced AKI. Targeting MIF with an MIF inhibitor RPS19 could be a promising therapeutic potential for AKI.


Asunto(s)
Lesión Renal Aguda/terapia , Inflamación/terapia , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Proteínas Ribosómicas/administración & dosificación , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Animales , Antígenos de Diferenciación de Linfocitos B/genética , Apoptosis/efectos de los fármacos , Cisplatino/efectos adversos , Terapia Genética , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/patología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Riñón/efectos de los fármacos , Riñón/patología , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Ratones , Ratones Noqueados , FN-kappa B/genética , Necrosis , Proteínas Ribosómicas/genética , Transducción de Señal/efectos de los fármacos
16.
Ann Neurol ; 82(1): 133-138, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28556183

RESUMEN

Glucose transporter type 1 (GLUT1) deficiency syndrome (GLUT1-DS) leads to a wide range of neurological symptoms. Ketogenic diets are very efficient to control epilepsy and movement disorders. We tested a novel simple and rapid blood test in 30 patients with GLUT1-DS with predominant movement disorders, 18 patients with movement disorders attributed to other genetic defects, and 346 healthy controls. We detected significantly reduced GLUT1 expression only on red blood cells from patients with GLUT1-DS (23 patients; 78%), including patients with inconclusive genetic analysis. This test opens perspectives for the screening of GLUT1-DS in children and adults with cognitive impairment, movement disorder, or epilepsy. Ann Neurol 2017;82:133-138.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/diagnóstico , Transportador de Glucosa de Tipo 1/biosíntesis , Pruebas Hematológicas , Proteínas de Transporte de Monosacáridos/deficiencia , Adolescente , Adulto , Errores Innatos del Metabolismo de los Carbohidratos/sangre , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Transporte de Monosacáridos/sangre , Trastornos del Movimiento/sangre , Trastornos del Movimiento/diagnóstico , Adulto Joven
18.
Haematologica ; 103(6): 949-958, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29599205

RESUMEN

Diamond-Blackfan anemia (DBA) is a rare inherited bone marrow failure disorder linked predominantly to ribosomal protein gene mutations. Here the European DBA consortium reports novel mutations identified in the RPL15 gene in 6 unrelated individuals diagnosed with DBA. Although point mutations have not been previously reported for RPL15, we identified 4 individuals with truncating mutations p.Tyr81* (in 3 of 4) and p.Gln29*, and 2 with missense variants p.Leu10Pro and p.Lys153Thr. Notably, 75% (3 of 4) of truncating mutation carriers manifested with severe hydrops fetalis and required intrauterine transfusions. Even more remarkable is the observation that the 3 carriers of p.Tyr81* mutation became treatment-independent between four and 16 months of life and maintained normal blood counts until their last follow up. Genetic reversion at the DNA level as a potential mechanism of remission was not observed in our patients. In vitro studies revealed that cells carrying RPL15 mutations have pre-rRNA processing defects, reduced 60S ribosomal subunit formation, and severe proliferation defects. Red cell culture assays of RPL15-mutated primary erythroblast cells also showed a severe reduction in cell proliferation, delayed erythroid differentiation, elevated TP53 activity, and increased apoptosis. This study identifies a novel subgroup of DBA with mutations in the RPL15 gene with an unexpected high rate of hydrops fetalis and spontaneous, long-lasting remission.


Asunto(s)
Anemia de Diamond-Blackfan/complicaciones , Anemia de Diamond-Blackfan/genética , Hidropesía Fetal/diagnóstico , Hidropesía Fetal/etiología , Mutación , Complicaciones Hematológicas del Embarazo , Proteínas Ribosómicas/genética , Anemia de Diamond-Blackfan/diagnóstico , Anemia de Diamond-Blackfan/terapia , Apoptosis/genética , Biomarcadores , Diferenciación Celular/genética , Línea Celular , Proliferación Celular , Análisis Mutacional de ADN , Índices de Eritrocitos , Femenino , Genes p53 , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Linaje , Fenotipo , Embarazo , Biosíntesis de Proteínas
19.
Eur J Haematol ; 101(4): 566-569, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29969830

RESUMEN

Severe iron overload is frequent in dehydrated hereditary stomatocytosis (DHSt) despite well-compensated hemolysis and no or little transfusion requirement. We investigated 4 patients with proven DHSt, in whom the degree of hemolysis was closely related to iron status. Genetic modifiers increasing iron stores (HFE:pCys282Tyr, HAMP:c-153C>T mutations) were accompanied with high liver iron concentrations and increased hemolysis, whereas therapeutic phlebotomies alleviated the hemolytic phenotype. There were no manifestations of hemolysis in one patient with low iron stores. Hemolysis reappeared when iron supplementation was given. The search for genetic or acquired modifiers of iron status and the modulation of iron stores may help in the management of these patients.


Asunto(s)
Anemia Hemolítica Congénita/diagnóstico , Anemia Hemolítica Congénita/metabolismo , Hidropesía Fetal/diagnóstico , Hidropesía Fetal/metabolismo , Hierro/metabolismo , Fenotipo , Adulto , Alelos , Anemia Hemolítica Congénita/sangre , Anemia Hemolítica Congénita/genética , Biomarcadores , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Proteína de la Hemocromatosis/genética , Humanos , Hidropesía Fetal/sangre , Hidropesía Fetal/genética , Masculino , Persona de Mediana Edad , Mutación , Radiografía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA