Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Int J Mol Sci ; 24(12)2023 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-37373084

RESUMEN

Congenital dyserythropoietic anemia type II (CDA II) is an inherited autosomal recessive blood disorder which belongs to the wide group of ineffective erythropoiesis conditions. It is characterized by mild to severe normocytic anemia, jaundice, and splenomegaly owing to the hemolytic component. This often leads to liver iron overload and gallstones. CDA II is caused by biallelic mutations in the SEC23B gene. In this study, we report 9 new CDA II cases and identify 16 pathogenic variants, 6 of which are novel. The newly reported variants in SEC23B include three missenses (p.Thr445Arg, p.Tyr579Cys, and p.Arg701His), one frameshift (p.Asp693GlyfsTer2), and two splicing variants (c.1512-2A>G, and the complex intronic variant c.1512-3delinsTT linked to c.1512-16_1512-7delACTCTGGAAT in the same allele). Computational analyses of the missense variants indicated a loss of key residue interactions within the beta sheet and the helical and gelsolin domains, respectively. Analysis of SEC23B protein levels done in patient-derived lymphoblastoid cell lines (LCLs) showed a significant decrease in SEC23B protein expression, in the absence of SEC23A compensation. Reduced SEC23B mRNA expression was only detected in two probands carrying nonsense and frameshift variants; the remaining patients showed either higher gene expression levels or no expression changes at all. The skipping of exons 13 and 14 in the newly reported complex variant c.1512-3delinsTT/c.1512-16_1512-7delACTCTGGAAT results in a shorter protein isoform, as assessed by RT-PCR followed by Sanger sequencing. In this work, we summarize a comprehensive spectrum of SEC23B variants, describe nine new CDA II cases accounting for six previously unreported variants, and discuss innovative therapeutic approaches for CDA II.


Asunto(s)
Anemia Diseritropoyética Congénita , Humanos , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/metabolismo , Mutación , Mutación Missense , Exones , Alelos , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
2.
Curr Opin Hematol ; 29(3): 126-136, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35441598

RESUMEN

PURPOSE OF REVIEW: The congenital dyserythropoietic anemias (CDA) are hereditary disorders characterized by ineffective erythropoiesis. This review evaluates newly developed CDA disease models, the latest advances in understanding the pathogenesis of the CDAs, and recently identified CDA genes. RECENT FINDINGS: Mice exhibiting features of CDAI were recently generated, demonstrating that Codanin-1 (encoded by Cdan1) is essential for primitive erythropoiesis. Additionally, Codanin-1 was found to physically interact with CDIN1, suggesting that mutations in CDAN1 and CDIN1 result in CDAI via a common mechanism. Recent advances in CDAII (which results from SEC23B mutations) have also been made. SEC23B was found to functionally overlap with its paralogous protein, SEC23A, likely explaining the absence of CDAII in SEC23B-deficient mice. In contrast, mice with erythroid-specific deletion of 3 or 4 of the Sec23 alleles exhibited features of CDAII. Increased SEC23A expression rescued the CDAII erythroid defect, suggesting a novel therapeutic strategy for the disease. Additional recent advances included the identification of new CDA genes, RACGAP1 and VPS4A, in CDAIII and a syndromic CDA type, respectively. SUMMARY: Establishing cellular and animal models of CDA is expected to result in improved understanding of the pathogenesis of these disorders, which may ultimately lead to the development of new therapies.


Asunto(s)
Anemia Diseritropoyética Congénita , ATPasas de Translocación de Protón Vacuolares , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/metabolismo , Animales , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Eritropoyesis/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Ratones , Mutación , Proteínas Nucleares/genética , ATPasas de Translocación de Protón Vacuolares/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo
3.
J Biol Chem ; 298(1): 101536, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34954140

RESUMEN

SEC23B is one of two vertebrate paralogs of SEC23, a key component of the coat protein complex II vesicles. Complete deficiency of SEC23B in mice leads to perinatal death caused by massive degeneration of professional secretory tissues. However, functions of SEC23B in postnatal mice and outside professional secretory tissues are unclear. In this study, we generated a Sec23b KO mouse and a knockin (KI) mouse with the E109K mutation, the most common human mutation in congenital dyserythropoietic anemia type II patients. We found that E109K mutation led to decreases in SEC23B levels and protein mislocalization. However, Sec23bki/ki mice showed no obvious abnormalities. Sec23b hemizygosity (Sec23bki/ko) was partially lethal, with only half of expected hemizygous mice surviving past weaning. Surviving Sec23bki/ko mice exhibited exocrine insufficiency, increased endoplasmic reticulum stress and apoptosis in the pancreas, and phenotypes consistent with chronic pancreatitis. Sec23bki/ko mice had mild to moderate anemia without other typical congenital dyserythropoietic anemia type II features, likely resulting from exocrine insufficiency. Moreover, Sec23bki/ko mice exhibited severe growth restriction accompanied by growth hormone (GH) insensitivity, reminiscent of Laron syndrome. Growth restriction is not associated with hepatocyte-specific Sec23b deletion, suggesting a nonliver origin of this phenotype. We propose that inflammation associated with chronic pancreatic deficiency may explain GH insensitivity in Sec23bki/ko mice. Our results reveal a genotype-phenotype correlation in SEC23B deficiency and indicate that pancreatic acinar is most sensitive to SEC23B deficiency in adult mice. The Sec23bki/ko mice provide a novel model of chronic pancreatitis and growth retardation with GH insensitivity.


Asunto(s)
Anemia Diseritropoyética Congénita , Mutación Missense , Pancreatitis Crónica , Proteínas de Transporte Vesicular , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/metabolismo , Animales , Ratones , Pancreatitis Crónica/genética , Pancreatitis Crónica/metabolismo , Fenotipo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
5.
Int J Mol Sci ; 22(6)2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33809261

RESUMEN

Lipin2 is a phosphatidate phosphatase that plays critical roles in fat homeostasis. Alterations in Lpin2, which encodes lipin2, cause the autoinflammatory bone disorder Majeed syndrome. Lipin2 limits lipopolysaccharide (LPS)-induced inflammatory responses in macrophages. However, little is known about the precise molecular mechanisms underlying its anti-inflammatory function. In this study, we attempted to elucidate the molecular link between the loss of lipin2 function and autoinflammatory bone disorder. Using a Lpin2 knockout murine macrophage cell line, we showed that lipin2 deficiency enhances innate immune responses to LPS stimulation through excessive activation of the NF-κB signaling pathway, partly because of TAK1 signaling upregulation. Lipin2 depletion also enhanced RANKL-mediated osteoclastogenesis and osteoclastic resorption activity accompanied by NFATc1 dephosphorylation and increased nuclear accumulation. These results suggest that lipin2 suppresses the development of autoinflammatory bone disorder by fine-tuning proinflammatory responses and osteoclastogenesis in macrophages. Therefore, this study provides insights into the molecular pathogenesis of monogenic autoinflammatory bone disorders and presents a potential therapeutic intervention.


Asunto(s)
Anemia Diseritropoyética Congénita/genética , Síndromes de Inmunodeficiencia/genética , Inflamación/genética , Quinasas Quinasa Quinasa PAM/genética , Factores de Transcripción NFATC/genética , Proteínas Nucleares/genética , Osteomielitis/genética , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Anemia Diseritropoyética Congénita/metabolismo , Anemia Diseritropoyética Congénita/patología , Animales , Resorción Ósea/genética , Resorción Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular/genética , Humanos , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/patología , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/genética , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , FN-kappa B/genética , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Osteoclastos/metabolismo , Osteogénesis/genética , Osteomielitis/metabolismo , Osteomielitis/patología , Ligando RANK/genética , Transducción de Señal/genética , Factor de Transcripción ReIA/genética
7.
Am J Hematol ; 94(11): 1227-1235, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31400017

RESUMEN

The erythroferrone (ERFE) is the erythroid regulator of hepatic iron metabolism by suppressing the expression of hepcidin. Congenital dyserythropoietic anemia type II (CDAII) is an inherited hyporegenerative anemia due to biallelic mutations in the SEC23B gene. Patients with CDAII exhibit marked clinical variability, even among individuals sharing the same pathogenic variants. The ERFE expression in CDAII is increased and related to abnormal erythropoiesis. We identified a recurrent low-frequency variant, A260S, in the ERFE gene in 12.5% of CDAII patients with a severe phenotype. We demonstrated that the ERFE-A260S variant leads to increased levels of ERFE, with subsequently marked impairment of iron regulation pathways at the hepatic level. Functional characterization of ERFE-A260S in the hepatic cell system demonstrated its modifier role in iron overload by impairing the BMP/SMAD pathway. We herein described for the first time an ERFE polymorphism as a genetic modifier variant. This was with a mild effect on disease expression, under a multifactorial-like model, in a condition of iron-loading anemia due to ineffective erythropoiesis.


Asunto(s)
Anemia Diseritropoyética Congénita/genética , Proteínas Morfogenéticas Óseas/fisiología , Sobrecarga de Hierro/etiología , Hígado/metabolismo , Hormonas Peptídicas/genética , Transducción de Señal/genética , Proteínas Smad/fisiología , Adolescente , Adulto , Anemia Diseritropoyética Congénita/complicaciones , Anemia Diseritropoyética Congénita/metabolismo , Transfusión Sanguínea , Proteína Morfogenética Ósea 6/farmacología , Línea Celular , Niño , Eritropoyesis/genética , Femenino , Estudios de Asociación Genética , Hepcidinas/biosíntesis , Hepcidinas/sangre , Hepcidinas/genética , Humanos , Masculino , Hormonas Peptídicas/sangre , Hormonas Peptídicas/farmacología , Hormonas Peptídicas/fisiología , Proteínas Recombinantes/farmacología , Índice de Severidad de la Enfermedad , Proteínas Smad/biosíntesis , Proteínas Smad/genética , Adulto Joven
8.
Exp Hematol ; 73: 25-37.e8, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30876823

RESUMEN

Krüppel-like factor 1 (KLF1), a transcription factor controlling definitive erythropoiesis, is involved in sequential control of terminal cell division and enucleation via fine regulation of key cell cycle regulator gene expression in erythroid lineage cells. Type IV congenital dyserythropoietic anemia (CDA) is caused by a monoallelic mutation at the second zinc finger of KLF1 (c.973G>A, p.E325K). We recently diagnosed a female patient with type IV CDA with the identical missense mutation. To understand the mechanism underlying the dyserythropoiesis caused by the mutation, we generated induced pluripotent stem cells (iPSCs) from the CDA patient (CDA-iPSCs). The erythroid cells that differentiated from CDA-iPSCs (CDA-erythroid cells) displayed multinucleated morphology, absence of CD44, and dysregulation of the KLF1 target gene expression. In addition, uptake of bromodeoxyuridine by CDA-erythroid cells was significantly decreased at the CD235a+/CD71+ stage, and microarray analysis revealed that cell cycle regulator genes were dysregulated, with increased expression of negative regulators such as CDKN2C and CDKN2A. Furthermore, inducible expression of the KLF1 E325K, but not the wild-type KLF1, caused a cell cycle arrest at the G1 phase in CDA-erythroid cells. Microarray analysis of CDA-erythroid cells and real-time polymerase chain reaction analysis of the KLF1 E325K inducible expression system also revealed altered expression of several KLF1 target genes including erythrocyte membrane protein band 4.1 (EPB41), EPB42, glutathione disulfide reductase (GSR), glucose phosphate isomerase (GPI), and ATPase phospholipid transporting 8A1 (ATP8A1). Our data indicate that the E325K mutation in KLF1 is associated with disruption of transcriptional control of cell cycle regulators in association with erythroid membrane or enzyme abnormalities, leading to dyserythropoiesis.


Asunto(s)
Anemia Diseritropoyética Congénita , Diferenciación Celular/genética , Células Eritroides , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Células Madre Pluripotentes Inducidas , Factores de Transcripción de Tipo Kruppel , Mutación Missense , Adulto , Sustitución de Aminoácidos , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/metabolismo , Anemia Diseritropoyética Congénita/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Eritroides/metabolismo , Células Eritroides/patología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo
9.
Br J Haematol ; 185(3): 436-449, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30836435

RESUMEN

Congenital dyserythropoietic anaemia type I (CDA-I) is one of a heterogeneous group of inherited anaemias characterised by ineffective erythropoiesis. CDA-I is caused by bi-allelic mutations in either CDAN1 or C15orf41 and, to date, 56 causative mutations have been documented. The diagnostic pathway is reviewed and the utility of genetic testing in reducing the time taken to reach an accurate molecular diagnosis and avoiding bone marrow aspiration, where possible, is described. The management of CDA-I patients is discussed, highlighting both general and specific measures which impact on disease progression. The use of interferon alpha and careful management of iron overload are reviewed and suggest the most favourable outcomes are achieved when CDA-I patients are managed with a holistic and multidisciplinary approach. Finally, the current understanding of the molecular and cellular pathogenesis of CDA-I is presented, highlighting critical questions likely to lead to improved therapy for this disease.


Asunto(s)
Alelos , Pruebas Genéticas , Glicoproteínas/genética , Interferón-alfa/uso terapéutico , Mutación , Proteínas Nucleares/genética , Anemia Diseritropoyética Congénita/diagnóstico , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/metabolismo , Anemia Diseritropoyética Congénita/terapia , Glicoproteínas/metabolismo , Humanos , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/prevención & control , Proteínas Nucleares/metabolismo
11.
Proc Natl Acad Sci U S A ; 115(33): E7748-E7757, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30065114

RESUMEN

Approximately one-third of the mammalian proteome is transported from the endoplasmic reticulum-to-Golgi via COPII-coated vesicles. SEC23, a core component of coat protein-complex II (COPII), is encoded by two paralogous genes in vertebrates (Sec23a and Sec23b). In humans, SEC23B deficiency results in congenital dyserythropoietic anemia type-II (CDAII), while SEC23A deficiency results in a skeletal phenotype (with normal red blood cells). These distinct clinical disorders, together with previous biochemical studies, suggest unique functions for SEC23A and SEC23B. Here we show indistinguishable intracellular protein interactomes for human SEC23A and SEC23B, complementation of yeast Sec23 by both human and murine SEC23A/B, and rescue of the lethality of sec23b deficiency in zebrafish by a sec23a-expressing transgene. We next demonstrate that a Sec23a coding sequence inserted into the murine Sec23b locus completely rescues the lethal SEC23B-deficient pancreatic phenotype. We show that SEC23B is the predominantly expressed paralog in human bone marrow, but not in the mouse, with the reciprocal pattern observed in the pancreas. Taken together, these data demonstrate an equivalent function for SEC23A/B, with evolutionary shifts in the transcription program likely accounting for the distinct phenotypes of SEC23A/B deficiency within and across species, a paradigm potentially applicable to other sets of paralogous genes. These findings also suggest that enhanced erythroid expression of the normal SEC23A gene could offer an effective therapeutic approach for CDAII patients.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Eritrocitos/metabolismo , Complejos Multiproteicos/biosíntesis , Proteínas de Transporte Vesicular/biosíntesis , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/metabolismo , Médula Ósea/metabolismo , Médula Ósea/patología , Vesículas Cubiertas por Proteínas de Revestimiento/genética , Eritrocitos/patología , Regulación de la Expresión Génica , Células HEK293 , Humanos , Complejos Multiproteicos/genética , Especificidad de la Especie , Proteínas de Transporte Vesicular/genética
12.
Cytometry B Clin Cytom ; 94(2): 312-326, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-27784127

RESUMEN

BACKGROUND: Bone marrow examination has been the confirmatory test for congenital dyserythropoietic anemia type II (CDAII). Occasional spherocytes on peripheral blood smear can confound the diagnosis. Since a screening test is still unavailable, we explored the feasibility of using flow cytometry as a preliminary screening method. METHODS: Thirteen monoclonal antibodies with specificities for eight erythrocyte membrane proteins were used in FACS analysis to probe the cellular features of red cells from CDAII, normal adults, hereditary spherocytosis (HS), and cord red cells. Confocal microscopy was performed on normal and CDAII to determine the overall distribution of CD44 and CD47. Their expression levels on cultured erythroblasts were also analyzed. RESULTS: The densely stained band 3 as seen in CDAII in gel electrophoresis was also obtained for Dantu phenotype. Likewise analysis of CDAII cases (n = 26) using the eosin-5'maleimide (EMA) binding test found 57% of patients giving results either positive or in the grey area for HS. Enhanced fluorescence of CD44 was detected in 96% of the CDAII patients, and anti-CD47 binding was also elevated to a lesser degree. Although RNA expressions of CD44 and CD47 in the cultured erythroblasts of normal controls and CDAII were similar, confocal microscopy revealed more CDAII red cells giving elevated fluorescence than normal red cells. CONCLUSIONS: A distinction between CDAII and HS can be made using the EMA Binding test and anti-CD44 binding. Confirmation of CDAII can subsequently be made based on clinical presentation together with either bone marrow examination or DNA sequencing of SEC23B. © 2016 International Clinical Cytometry Society.


Asunto(s)
Anemia Diseritropoyética Congénita/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular/fisiología , Receptores de Hialuranos/metabolismo , Esferocitosis Hereditaria/metabolismo , Antígeno CD47/metabolismo , Eritrocitos/metabolismo , Femenino , Citometría de Flujo/métodos , Humanos , Masculino , Tamizaje Masivo , Persona de Mediana Edad , Fenotipo
14.
Pediatr Blood Cancer ; 63(5): 917-21, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26713410

RESUMEN

We describe a child with dyserythropoietic anemia, thrombocytosis, functional platelet defect, and megakaryocyte dysplasia. We show that (i) this constellation of hematopoietic abnormalities was due to a germline mutation within the 5' untranslated region (5'UTR) of globin transcription factor 1 (GATA1); (ii) the mutation impaired a 5'UTR GATA1 splicing site, with promoted production of the shortened GATA1 isoform lacking the N-terminus; and (iii) expression of the GATA1 N-terminus is restricted to erythroblasts and megakaryocytes in normal marrow, consistent with the patient's abnormal erythropoiesis and megakaryopoiesis. Our findings provide insights into the clinically relevant in vivo function of the N-terminal domain of GATA1 in human hematopoiesis.


Asunto(s)
Regiones no Traducidas 5' , Anemia Diseritropoyética Congénita/genética , Factor de Transcripción GATA1/genética , Megacariocitos , Sitios de Empalme de ARN , Empalme Alternativo , Anemia Diseritropoyética Congénita/metabolismo , Preescolar , Factor de Transcripción GATA1/biosíntesis , Humanos , Masculino , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética
15.
Am J Hematol ; 88(11): E283-5, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23861216

RESUMEN

Chronic blood transfusions start at a very young age in subjects with transfusion-dependent anemias, the majority of whom have hereditary anemias. To understand how rapidly iron overload develops, we retrospectively reviewed 308 MRIs for evaluation of liver, pancreatic, or cardiac iron in 125 subjects less than 10 years old. Median age at first MRI evaluation was 6.0 years. Median liver iron concentrations in patients less than 3.5 years old were 14 and 13 mg/g dry weight in thalassemia major (TM) and Diamond-Blackfan anemia (DBA) patients, respectively. At time of first MRI, pancreatic iron was markedly elevated (> 100 Hz) in DBA patients, and cardiac iron ( R2* >50 Hz) was present in 5/112 subjects (4.5%), including a 2.5 years old subject with DBA. Five of 14 patients (38%) with congenital dyserythropoietic anemia (CDA) developed excess cardiac iron before their 10th birthday. Thus, clinically significant hepatic and cardiac iron accumulation occurs at an early age in patients on chronic transfusions, particularly in those with ineffective or absent erythropoiesis, such as DBA, CDA, and TM, who are at higher risk for iron cardiomyopathy. Performing MRI for iron evaluation in the liver, heart, and pancreas as early as feasible, particularly in those conditions in which there is suppressed bone marrow activity is very important in the management of iron loaded children in order to prescribe appropriate chelation to prevent long-term sequelae. .


Asunto(s)
Anemia/terapia , Sobrecarga de Hierro/etiología , Hierro/análisis , Hígado/química , Miocardio/química , Páncreas/química , Reacción a la Transfusión , Anemia/etiología , Anemia/metabolismo , Anemia de Diamond-Blackfan/metabolismo , Anemia de Diamond-Blackfan/terapia , Anemia Diseritropoyética Congénita/metabolismo , Anemia Diseritropoyética Congénita/terapia , Niño , Preescolar , Estudios de Cohortes , Eritropoyesis , Hospitales Pediátricos , Humanos , Hierro/metabolismo , Sobrecarga de Hierro/epidemiología , Hígado/metabolismo , Los Angeles/epidemiología , Imagen por Resonancia Magnética , Miocardio/metabolismo , Páncreas/metabolismo , Estudios Retrospectivos , Riesgo , Talasemia beta/metabolismo , Talasemia beta/terapia
16.
Blood Cells Mol Dis ; 51(1): 17-21, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23453696

RESUMEN

Congenital dyserythropoietic anemia type II, a recessive disorder of erythroid differentiation, is due to mutations in SEC23B, a component of the core trafficking machinery COPII. In no case homozygosity or compound heterozygosity for nonsense mutation(s) was found. This study represents the first description of molecular mechanisms underlying SEC23B hypomorphic genotypes by the analysis of five novel mutations. Our findings suggest that reduction of SEC23B gene expression is not associated with CDA II severe clinical presentation; conversely, the combination of a hypomorphic allele with one functionally altered results in more severe phenotypes. We propose a mechanism of compensation SEC23A-mediated which justifies these observations.


Asunto(s)
Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/metabolismo , Mutación , Fenotipo , Proteínas de Transporte Vesicular/genética , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Niño , Exones , Femenino , Genotipo , Humanos , Intrones , Masculino , Linaje , Polimorfismo Genético , Sitios de Empalme de ARN , Proteínas de Transporte Vesicular/metabolismo
17.
Am J Hematol ; 88(2): 135-40, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22764119

RESUMEN

Many diseases attributed to trafficking defects are primary disorders of protein folding and assembly. However, an increasing number of disease states are directly attributable to defects in trafficking machinery. In this context, the cytoplasmic coat protein (COP)II complex plays a pivotal role: it mediates the anterograde transport of correctly folded secretory cargo from the endoplasmic reticulum towards the Golgi apparatus. This review attempts to describe the involvement of COPII complex alteration in the pathogenesis of human genetic disorders; particularly, we will focus on two disorders, the Congenital Dyserythropoietic Anemia type II and the Combined Deficiency of Factor V and VIII.


Asunto(s)
Anemia Diseritropoyética Congénita/genética , Deficiencia del Factor V/genética , Hemofilia A/genética , Lectinas de Unión a Manosa/genética , Proteínas de la Membrana/genética , Mutación , Proteínas de Transporte Vesicular/genética , Anemia Diseritropoyética Congénita/metabolismo , Animales , Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/metabolismo , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Retículo Endoplásmico/metabolismo , Deficiencia del Factor V/metabolismo , Salud de la Familia , Aparato de Golgi/metabolismo , Hemofilia A/metabolismo , Humanos , Hipobetalipoproteinemias/genética , Hipobetalipoproteinemias/metabolismo , Síndromes de Malabsorción/genética , Síndromes de Malabsorción/metabolismo , Lectinas de Unión a Manosa/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas de Transporte Vesicular/metabolismo
18.
Stem Cell Res Ther ; 3(6): 55, 2012 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-23257067

RESUMEN

Erythrocytes contain oxygen-carrying hemoglobin to all body cells. Impairments in the generation of erythrocytes, a process known as erythropoiesis, or in hemoglobin synthesis alter cell function because of decreased oxygen supply and lead to anemic diseases. Thus, understanding how erythropoiesis is regulated during embryogenesis and adulthood is important to develop novel therapies for anemia. The zebrafish, Danio rerio, provides a powerful model for such study. Their small size and the ability to generate a large number of embryos enable large-scale analysis, and their transparency facilitates the visualization of erythroid cell migration. Importantly, the high conservation of hematopoietic genes among vertebrates and the ability to successfully transplant hematopoietic cells into fish have enabled the establishment of models of human anemic diseases in fish. In this review, we summarize the current progress in our understanding of erythropoiesis on the basis of zebrafish studies and highlight fish models of human anemias. These analyses could enable the discovery of novel drugs as future therapies.


Asunto(s)
Anemia/patología , Eritropoyesis/fisiología , Anemia/metabolismo , Anemia Diseritropoyética Congénita/metabolismo , Anemia Diseritropoyética Congénita/patología , Anemia Hipocrómica/metabolismo , Anemia Hipocrómica/patología , Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patología , Animales , Ancirinas/deficiencia , Ancirinas/metabolismo , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/metabolismo , Modelos Animales de Enfermedad , Eritrocitos/citología , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Hemocromatosis/metabolismo , Hemocromatosis/patología , Esferocitosis Hereditaria/metabolismo , Esferocitosis Hereditaria/patología , Pez Cebra
19.
Blood ; 120(1): 31-8, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22586181

RESUMEN

Multiple diseases, hematologic and nonhematologic, result from defects in the early secretory pathway. Congenital dyserythropoietic anemia type II (CDAII) and combined deficiency of coagulation factors V and VIII (F5F8D) are the 2 known hematologic diseases that result from defects in the endoplasmic reticulum (ER)-to-Golgi transport system. CDAII is caused by mutations in the SEC23B gene, which encodes a core component of the coat protein complex II (COPII). F5F8D results from mutations in either LMAN1 (lectin mannose-binding protein 1) or MCFD2 (multiple coagulation factor deficiency protein 2), which encode the ER cargo receptor complex LMAN1-MCFD2. These diseases and their molecular pathogenesis are the focus of this review.


Asunto(s)
Anemia Diseritropoyética Congénita , Vesículas Cubiertas por Proteínas de Revestimiento/patología , Deficiencia del Factor V , Hemofilia A , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/metabolismo , Anemia Diseritropoyética Congénita/patología , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Deficiencia del Factor V/genética , Deficiencia del Factor V/metabolismo , Deficiencia del Factor V/patología , Hemofilia A/genética , Hemofilia A/metabolismo , Hemofilia A/patología , Humanos , Lectinas de Unión a Manosa/genética , Lectinas de Unión a Manosa/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
20.
Orphanet J Rare Dis ; 6: 89, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-22208203

RESUMEN

BACKGROUND: Congenital dyserythropoietic anemia type II (CDAII), the most common form of CDA, is an autosomal recessive condition. CDAII diagnosis is based on invasive, expensive, and time consuming tests that are available only in specialized laboratories. The recent identification of SEC23B mutations as the cause of CDAII opens new possibilities for the molecular diagnosis of the disease. The aim of this study was to characterize molecular genomic SEC23B defects in 16 unrelated patients affected by CDAII and correlate the identified genetic alterations with SEC23B transcript and protein levels in erythroid precursors. METHODS: SEC23B was sequenced in 16 patients, their relatives and 100 control participants. SEC23B transcript level were studied by quantitative PCR (qPCR) in peripheral erythroid precursors and lymphocytes from the patients and healthy control participants. Sec23B protein content was analyzed by immunoblotting in samples of erythroblast cells from CDAII patients and healthy controls. RESULTS: All of the investigated cases carried SEC23B mutations on both alleles, with the exception of two patients in which a single heterozygous mutation was found. We identified 15 different SEC23B mutations, of which four represent novel mutations: p.Gln214Stop, p.Thr485Ala, p.Val637Gly, and p.Ser727Phe. The CDAII patients exhibited a 40-60% decrease of SEC23B mRNA levels in erythroid precursors when compared with the corresponding cell type from healthy participants. The largest decrease was observed in compound heterozygote patients with missense/nonsense mutations. In three patients, Sec23B protein levels were evaluated in erythroid precursors and found to be strictly correlated with the reduction observed at the transcript level. We also demonstrate that Sec23B mRNA expression levels in lymphocytes and erythroblasts are similar. CONCLUSIONS: In this study, we identified four novel SEC23B mutations associated with CDAII disease. We also demonstrate that the genetic alteration results in a significant decrease of SEC23B transcript in erythroid precursors. Similar down-regulation was observed in peripheral lymphocytes, suggesting that the use of these cells might be sufficient in the identification of Sec23B gene alterations. Finally, we demonstrate that decreased Sec23B protein levels in erythroid precursors correlate with down-regulation of the SEC23B mRNA transcript.


Asunto(s)
Anemia Diseritropoyética Congénita/genética , Células Precursoras Eritroides/metabolismo , Mutación , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Anemia Diseritropoyética Congénita/metabolismo , Células Cultivadas , Familia , Humanos , Italia , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN , Proteínas de Transporte Vesicular/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...