Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Pediatr Hematol Oncol ; 46(5): e338-e347, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38857202

RESUMEN

Primary mitochondrial disorders (PMDs) are known for their pleiotropic manifestations in humans, affecting almost any organ or system at any time. Hematologic manifestations, such as cytopenias and sideroblastic anemia, occur in 10% to 30% of patients with confirmed PMDs. These can be the initial presenting features or complications that develop over time. Surveillance for these manifestations allows for prompt identification and treatment. This article provides an overview of the pathophysiology underpinning the hematologic effects of mitochondrial dysfunction, discussing the 3 key roles of the mitochondria in hematopoiesis: providing energy for cell differentiation and function, synthesizing heme, and generating iron-sulfur clusters. Subsequently, the diagnosis and management of mitochondrial disorders are discussed, focusing on hematologic manifestations and the specific conditions commonly associated with them. Through this, we aimed to provide a concise point of reference for those considering a mitochondrial cause for a patient's hematologic abnormality, or for those considering a hematologic manifestation in a patient with known or suspected mitochondrial disease.


Asunto(s)
Enfermedades Hematológicas , Enfermedades Mitocondriales , Humanos , Enfermedades Mitocondriales/complicaciones , Enfermedades Hematológicas/sangre , Enfermedades Hematológicas/complicaciones , Enfermedades Hematológicas/patología , Mitocondrias/patología , Hematopoyesis , Anemia Sideroblástica/diagnóstico , Anemia Sideroblástica/terapia
2.
Intern Med J ; 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38064543

RESUMEN

BACKGROUND: Sickle cell disease (SCD) is the most common monogenic disorder worldwide. In deoxygenated conditions, the altered beta chain (haemoglobin S [HbS]) polymerises and distorts the erythrocyte, resulting in pain crises, vasculopathy and end-organ damage. Clinical complications of SCD cause substantial morbidity, and therapy demands expertise and resources. Optimising care for patients and planning resource allocation for the future requires an understanding of the disease in the Australian population. The Australian Haemoglobinopathy Registry (HbR) is a collaborative initiative of specialist centres collating and analysing data on patients with haemoglobin disorders. AIMS: To provide a snapshot of SCD in Australia over a 12-month period based on data from the HbR. METHODS: Patients with a clinically significant sickling disorder across 12 clinical sites were included for analysis. Data include demographic and diagnostic details, as well as details of the clinical management of the condition over a 12-month period. RESULTS: Data on 359 SCD patients demonstrate a shift in the demographic of patients in Australia, with a growing proportion of sub-Saharan African ethnicities associated with the HbSS genotype. Acute and chronic complications are common, and patients require significant outpatient and inpatient support. Prevalence of disease complications and therapeutic trends are in keeping with other high-income countries. CONCLUSIONS: This study provides the first national picture of SCD in Australia, describing the characteristics and needs of SCD patients, elucidating demand for current and novel therapy and facilitating the planning of services for this vulnerable population.

3.
Hum Mutat ; 42(11): 1367-1383, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34298585

RESUMEN

The congenital sideroblastic anemias (CSAs) are a heterogeneous group of inherited disorders of erythropoiesis characterized by pathologic deposits of iron in the mitochondria of developing erythroblasts. Mutations in the mitochondrial glycine carrier SLC25A38 cause the most common recessive form of CSA. Nonetheless, the disease is still rare, there being fewer than 70 reported families. Here we describe the clinical phenotype and genotypes of 31 individuals from 24 families, including 11 novel mutations. We also review the spectrum of reported mutations and genotypes associated with the disease, describe the unique localization of missense mutations in transmembrane domains and account for the presence of several alleles in different populations.


Asunto(s)
Anemia Sideroblástica/congénito , Genotipo , Proteínas de Transporte de Membrana Mitocondrial/genética , Mutación , Fenotipo , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino
4.
EJHaem ; 2(2): 157-166, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-35845273

RESUMEN

Telomere biology disorders (TBDs), including dyskeratosis congenita (DC), are a group of rare inherited diseases characterized by very short telomeres. Mutations in the components of the enzyme telomerase can lead to insufficient telomere maintenance in hematopoietic stem cells, resulting in the bone marrow failure that is characteristic of these disorders. While an increasing number of genes are being linked to TBDs, the causative mutation remains unidentified in 30-40% of patients with DC. There is therefore a need for whole genome sequencing (WGS) in these families to identify novel genes, or mutations in regulatory regions of known disease-causing genes. Here we describe a family in which a partial deletion of the 3' untranslated region (3' UTR) of DKC1, encoding the protein dyskerin, was identified by WGS, despite being missed by whole exome sequencing. The deletion segregated with disease across the family and resulted in reduced levels of DKC1 mRNA in the proband. We demonstrate that the DKC1 3' UTR contains two polyadenylation signals, both of which were removed by this deletion, likely causing mRNA instability. Consistent with the major function of dyskerin in stabilization of the RNA subunit of telomerase, hTR, the level of hTR was also reduced in the proband, providing a molecular basis for his very short telomeres. This study demonstrates that the terminal region of the 3' UTR of the DKC1 gene is essential for gene function and illustrates the importance of analyzing regulatory regions of the genome for molecular diagnosis of inherited disease.

5.
Nat Immunol ; 20(10): 1299-1310, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31534238

RESUMEN

Resisting and tolerating microbes are alternative strategies to survive infection, but little is known about the evolutionary mechanisms controlling this balance. Here genomic analyses of anatomically modern humans, extinct Denisovan hominins and mice revealed a TNFAIP3 allelic series with alterations in the encoded immune response inhibitor A20. Each TNFAIP3 allele encoded substitutions at non-catalytic residues of the ubiquitin protease OTU domain that diminished IκB kinase-dependent phosphorylation and activation of A20. Two TNFAIP3 alleles encoding A20 proteins with partial phosphorylation deficits seemed to be beneficial by increasing immunity without causing spontaneous inflammatory disease: A20 T108A;I207L, originating in Denisovans and introgressed in modern humans throughout Oceania, and A20 I325N, from an N-ethyl-N-nitrosourea (ENU)-mutagenized mouse strain. By contrast, a rare human TNFAIP3 allele encoding an A20 protein with 95% loss of phosphorylation, C243Y, caused spontaneous inflammatory disease in humans and mice. Analysis of the partial-phosphorylation A20 I325N allele in mice revealed diminished tolerance of bacterial lipopolysaccharide and poxvirus inoculation as tradeoffs for enhanced immunity.


Asunto(s)
Infecciones por Poxviridae/inmunología , Poxviridae/fisiología , Dominios Proteicos/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Alelos , Animales , Extinción Biológica , Humanos , Inmunidad , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense/genética , Fosforilación
7.
Eur J Haematol ; 98(2): 97-105, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27537786

RESUMEN

OBJECTIVES: To assess the effect of iron chelation therapy with deferasirox on cardiac iron and function in patients with transfusion-dependent thalassemia major, sickle cell disease (SCD), and myelodysplastic syndromes (MDS). METHODS: This phase IV, single-arm, open-label study over 53 wk evaluated the change in cardiac and liver iron load with deferasirox (up to 40 mg/kg/d), measured by magnetic resonance imaging (MRI). RESULTS: Cardiac iron load (myocardial T2*) significantly improved (P = 0.002) overall (n = 46; n = 36 thalassemia major, n = 4 SCD, n = 6 MDS). Results were significant for patients with normal and moderate baseline cardiac iron (P = 0.017 and P = 0.015, respectively), but not in the five patients with severe cardiac iron load. Liver iron concentration (LIC) significantly decreased overall [mean LIC 10.4 to 8.2 mg Fe/g dry tissue (dw); P = 0.024], particularly in those with baseline LIC >7 mg Fe/g dw (19.9 to 15.6 mg Fe/g dw; P = 0.002). Furthermore, myocardial T2* significantly increased in patients with LIC <7 mg Fe/g dw, but not in those with a higher LIC. Safety was consistent with previous reports. CONCLUSIONS: Once-daily deferasirox over 1 yr significantly increased myocardial T2* and reduced LIC. This confirms that single-agent deferasirox is effective in the management of cardiac iron, especially for patients with myocardial T2* >10 ms (Clinicaltrials.gov identifier: NCT00673608).


Asunto(s)
Benzoatos/uso terapéutico , Transfusión Sanguínea , Terapia por Quelación , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Hierro/metabolismo , Miocardio/metabolismo , Triazoles/uso terapéutico , Adolescente , Adulto , Anciano , Benzoatos/farmacología , Deferasirox , Femenino , Ferritinas/sangre , Pruebas de Función Cardíaca , Hemoglobinopatías/complicaciones , Hemoglobinopatías/terapia , Humanos , Quelantes del Hierro/farmacología , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/etiología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Reacción a la Transfusión , Resultado del Tratamiento , Triazoles/farmacología , Adulto Joven
8.
Br J Haematol ; 175(2): 318-330, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27432187

RESUMEN

Accurate diagnosis of rare inherited anaemias is challenging, requiring a series of complex and expensive laboratory tests. Targeted next-generation-sequencing (NGS) has been used to investigate these disorders, but the selection of genes on individual panels has been narrow and the validation strategies used have fallen short of the standards required for clinical use. Clinical-grade validation of negative results requires the test to distinguish between lack of adequate sequencing reads at the locations of known mutations and a real absence of mutations. To achieve a clinically-reliable diagnostic test and minimize false-negative results we developed an open-source tool (CoverMi) to accurately determine base-coverage and the 'discoverability' of known mutations for every sample. We validated our 33-gene panel using Sanger sequencing and microarray. Our panel demonstrated 100% specificity and 99·7% sensitivity. We then analysed 57 clinical samples: molecular diagnoses were made in 22/57 (38·6%), corresponding to 32 mutations of which 16 were new. In all cases, accurate molecular diagnosis had a positive impact on clinical management. Using a validated NGS-based platform for routine molecular diagnosis of previously undiagnosed congenital anaemias is feasible in a clinical diagnostic setting, improves precise diagnosis and enhances management and counselling of the patient and their family.


Asunto(s)
Anemia/diagnóstico , Anemia/genética , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Biología Computacional/métodos , Manejo de la Enfermedad , Estudios de Asociación Genética , Pruebas Genéticas/métodos , Pruebas Genéticas/normas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Mutación , Polimorfismo de Nucleótido Simple , Enfermedades Raras , Reproducibilidad de los Resultados , Flujo de Trabajo
9.
JIMD Rep ; 28: 49-57, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26537577

RESUMEN

Pathogenic variants in mitochondrial aminoacyl-tRNA synthetases result in a broad range of mitochondrial respiratory chain disorders despite their shared role in mitochondrial protein synthesis. LARS2 encodes the mitochondrial leucyl-tRNA synthetase, which attaches leucine to its cognate tRNA. Sequence variants in LARS2 have previously been associated with Perrault syndrome, characterized by premature ovarian failure and hearing loss (OMIM #615300). In this study, we report variants in LARS2 that are associated with a severe multisystem metabolic disorder. The proband was born prematurely with severe lactic acidosis, hydrops, and sideroblastic anemia. She had multisystem complications with hyaline membrane disease, impaired cardiac function, a coagulopathy, pulmonary hypertension, and progressive renal disease and succumbed at 5 days of age. Whole exome sequencing of patient DNA revealed compound heterozygous variants in LARS2 (c.1289C>T; p.Ala430Val and c.1565C>A; p.Thr522Asn). The c.1565C>A (p.Thr522Asn) LARS2 variant has previously been associated with Perrault syndrome and both identified variants are predicted to be damaging (SIFT, PolyPhen). Muscle and liver samples from the proband did not display marked mitochondrial respiratory chain enzyme deficiency. Immunoblotting of patient muscle and liver showed LARS2 levels were reduced in liver and complex I protein levels were reduced in patient muscle and liver. Aminoacylation assays revealed p.Ala430Val LARS2 had an 18-fold loss of catalytic efficiency and p.Thr522Asn a 9-fold loss compared to wild-type LARS2. We suggest that the identified LARS2 variants are responsible for the severe multisystem clinical phenotype seen in this baby and that mutations in LARS2 can result in variable phenotypes.

10.
Blood ; 124(18): 2767-74, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25205116

RESUMEN

Telomerase is a ribonucleoprotein enzyme that is necessary for overcoming telomere shortening in human germ and stem cells. Mutations in telomerase or other telomere-maintenance proteins can lead to diseases characterized by depletion of hematopoietic stem cells and bone marrow failure (BMF). Telomerase localization to telomeres requires an interaction with a region on the surface of the telomere-binding protein TPP1 known as the TEL patch. Here, we identify a family with aplastic anemia and other related hematopoietic disorders in which a 1-amino-acid deletion in the TEL patch of TPP1 (ΔK170) segregates with disease. All family members carrying this mutation, but not those with wild-type TPP1, have short telomeres. When introduced into 293T cells, TPP1 with the ΔK170 mutation is able to localize to telomeres but fails to recruit telomerase to telomeres, supporting a causal relationship between this TPP1 mutation and bone marrow disorders. ACD/TPP1 is thus a newly identified telomere-related gene in which mutations cause aplastic anemia and related BMF disorders.


Asunto(s)
Enfermedades de la Médula Ósea/genética , Médula Ósea/patología , Mutación de Línea Germinal/genética , Patrón de Herencia/genética , Serina Proteasas/genética , Telomerasa/genética , Telómero/metabolismo , Adulto , Alelos , Anemia/genética , Niño , Exoma/genética , Femenino , Genoma Humano/genética , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Neoplasias/genética , Linaje , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Complejo Shelterina , Telomerasa/metabolismo , Homeostasis del Telómero/genética , Proteínas de Unión a Telómeros
11.
Am J Physiol Cell Physiol ; 299(5): C1153-61, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20686069

RESUMEN

The proton-coupled folate transporter (PCFT-SLC46A1) is required for intestinal folate absorption and is mutated in the autosomal recessive disorder, hereditary folate malabsorption (HFM). This report characterizes properties and requirements of the R376 residue in PCFT function, including a R376Q mutant associated with HFM. Gln, Cys, and Ala substitutions resulted in markedly impaired transport of 5-formyltetrahydrofolate (5-FTHF) and 5-methyltetrahydrofolate (5-MTHF) due to an increase in K(m) and decrease in V(max) in HeLa R1-11 transfectants lacking endogenous folate transport function. In contrast, although the influx K(m) for pemetrexed was increased, transport was fully preserved at saturating concentrations and enhanced for the like-charged R376K- and R376H-PCFT. Pemetrexed and 5-FTHF influx mediated by R376Q-PCFT was markedly decreased at pH 5.5 compared with wild-type PCFT. However, while pemetrexed transport was substantially preserved at low pH (4.5-5.0), 5-FTHF transport remained very low. Electrophysiological studies in Xenopus oocytes demonstrated that 1) the R376Q mutant, like wild-type PCFT, transports protons in the absence of folate substrate, and in this respect has channel-like properties; and 2) the influx K(m) mediated by R376Q-PCFT is increased for 5-MTHF, 5-FTHF, and pemetrexed. The data suggest that mutation of the R376 residue to Gln impairs proton binding which, in turn, modulates the folate-binding pocket and depresses the rate of conformational alteration of the carrier, a change that appears to be, in part, substrate dependent.


Asunto(s)
Sustitución de Aminoácidos , Arginina/metabolismo , Ácido Fólico/metabolismo , Glutamina/metabolismo , Síndromes de Malabsorción , Transportador de Folato Acoplado a Protón/genética , Animales , Antagonistas del Ácido Fólico , Glutamatos/metabolismo , Glutamina/genética , Guanina/análogos & derivados , Guanina/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Síndromes de Malabsorción/genética , Síndromes de Malabsorción/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Oocitos/citología , Oocitos/fisiología , Técnicas de Placa-Clamp , Pemetrexed , Transportador de Folato Acoplado a Protón/metabolismo , Protones , Tetrahidrofolatos/genética , Tetrahidrofolatos/metabolismo , Xenopus laevis
12.
Pediatr Nephrol ; 24(9): 1749-52, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19399522

RESUMEN

Thrombotic thrombocytopenic purpura (TTP) is a rare disease among pediatric patients, in whom it may be mistaken for hemolytic uremic syndrome (HUS) and idiopathic thrombocytopenic purpura (ITP). Familial forms are caused by mutations in the ADAMTS13 gene, whereas acquired forms may result from an inhibitory antibody directed against ADAMTS13, a metalloprotease that cleaves very large multimers of Von Willebrand factor (VWF), thereby preventing platelet aggregation in blood vessels. We report two cases of TTP. The first was a 15-year-old girl with her first episode of TTP that failed to respond after 10 days of plasmapheresis and was treated with rituximab; she has remained in remission at 12 months of follow-up. The second was a 6-year-old boy with acquired relapsing TTP previously managed with plasmapheresis and prednisolone, who presented with a third relapse that was treated with plasmapheresis and rituximab; he remains in remission 17 months after treatment. Rituximab has been used by pediatricians for treating B cell malignancy, autoimmune diseases and antibody-mediated diseases, such as the Factor VIII inhibitors in hemophilia A, and may also have a promising role in children with acute refractory or chronic relapsing TTP.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Factores Inmunológicos/uso terapéutico , Plasmaféresis , Púrpura Trombocitopénica Idiopática/terapia , Adolescente , Anticuerpos Monoclonales de Origen Murino , Preescolar , Terapia Combinada , Femenino , Humanos , Masculino , Púrpura Trombocitopénica Idiopática/patología , Recurrencia , Inducción de Remisión , Rituximab , Resultado del Tratamiento
14.
Pediatrics ; 122(1): e139-48, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18595958

RESUMEN

OBJECTIVE: Unclassified inherited bone marrow failure syndromes are a heterogeneous group of genetic disorders that represent either new syndromes or atypical clinical courses of known inherited bone marrow failure syndromes. The relative prevalence of the unclassified inherited bone marrow failure syndromes and their characteristics and the clinical and economic challenges that they create have never been studied. METHODS: We analyzed cases of inherited bone marrow failure syndrome in the Canadian Inherited Marrow Failure Registry that were deemed unclassifiable at study entry. RESULTS: From October 2001 to March 2006, 39 of the 162 patients enrolled in the Canadian Inherited Marrow Failure Registry were registered as having unclassified inherited bone marrow failure syndromes. These patients presented at a significantly older age (median: 9 months) than the patients with classified inherited bone marrow failure syndrome (median: 1 month) and had substantial variation in the clinical presentations. The hematologic phenotype, however, was similar to the classified inherited bone marrow failure syndromes and included single- or multiple-lineage cytopenia, severe aplastic anemia, myelodysplasia, and malignancy. Grouping patients according to the affected blood cell lineage(s) and to the presence of associated physical malformations was not always sufficient to characterize a condition, because affected members from several families fit into different phenotypic groups. Compared with the classified inherited bone marrow failure syndromes, the patients with unclassified inherited bone marrow failure syndromes had 3.2 more specific diagnostic tests at 4.5 times higher cost per evaluated patient to attempt to categorize their syndrome. At last follow-up, only 20% of the unclassified inherited bone marrow failure syndromes were ultimately diagnosed with a specific syndrome on the basis of the development of new clinical findings or positive genetic tests. CONCLUSIONS: Unclassified inherited bone marrow failure syndromes are relatively common among the inherited bone marrow failure syndromes and present a major diagnostic and therapeutic dilemma.


Asunto(s)
Enfermedades de la Médula Ósea/genética , Anemia Aplásica/epidemiología , Enfermedades de la Médula Ósea/clasificación , Enfermedades de la Médula Ósea/epidemiología , Canadá/epidemiología , Análisis Citogenético , Femenino , Enfermedades Hematológicas , Humanos , Lactante , Masculino , Defectos del Tubo Neural/epidemiología , Neutropenia/epidemiología , Pancitopenia/epidemiología , Fenotipo , Sistema de Registros , Síndrome , Trombocitopenia/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA