GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    American Society of Hematology ; 2020
    In:  Blood Vol. 136, No. 11 ( 2020-09-10), p. 1274-1283
    In: Blood, American Society of Hematology, Vol. 136, No. 11 ( 2020-09-10), p. 1274-1283
    Abstract: Congenital dyserythropoietic anemias (CDAs) are a heterogeneous group of inherited anemias that affect the normal differentiation–proliferation pathways of the erythroid lineage. They belong to the wide group of ineffective erythropoiesis conditions that mainly result in monolinear cytopenia. CDAs are classified into the 3 major types (I, II, III), plus the transcription factor-related CDAs, and the CDA variants, on the basis of the distinctive morphological, clinical, and genetic features. Next-generation sequencing has revolutionized the field of diagnosis of and research into CDAs, with reduced time to diagnosis, and ameliorated differential diagnosis in terms of identification of new causative/modifier genes and polygenic conditions. The main improvements regarding CDAs have been in the study of iron metabolism in CDAII. The erythroblast-derived hormone erythroferrone specifically inhibits hepcidin production, and its role in the mediation of hepatic iron overload has been dissected out. We discuss here the most recent advances in this field regarding the molecular genetics and pathogenic mechanisms of CDAs, through an analysis of the clinical and molecular classifications, and the complications and clinical management of patients. We summarize also the main cellular and animal models developed to date and the possible future therapies.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2011
    In:  Blood Vol. 118, No. 21 ( 2011-11-18), p. 2098-2098
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2098-2098
    Abstract: Abstract 2098 Congenital dyserythropoietic anemias (CDAs) designate a group of genetic disorders in which ineffective erythropoiesis is the predominant mechanism of anemia marked by distinct morphologic abnormalities of the majority of erythroblasts in the bone marrow. CDA type II (CDAII) is the most common type of CDA. It is characterized by a recessive model of inheritance, mild to moderate anemia, jaundice, and splenomegaly (Fukuda MN, Glycobiology 1990; Fukuda MN, Clin Haematol 1993), by the presence of bi- and multinucleated erythroblasts in bone marrow, with nuclei of equal size and DNA content, suggesting a cytokinesis disturbance (Schwarz K and Iolascon A. et al., Nat Genet. 2009). The specific hallmark of diagnosis is the presence of the more abundant protein of membrane red cell, band 3, in a hypoglycosilated state; this is thinner and migrated slightly faster than in controls on sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) (Anselstetter V et al., Br J Haematol 1977). The causative gene of CDAII, SEC23B, is a member of the SEC23 subfamily of the SEC23/SEC24 family, which is involved in vesicle trafficking. The encoded protein has similarity to yeast Sec23p (66.4%) component of COPII, the coat protein complex responsible for vesicle budding from the ER. The function of this gene product has been implicated in cargo selection and concentration. The SEC23B gene spans approximately 54 kb on human chromosome 20p11.23 and codifies for a protein of 767 aminoacids divided in five functional domains: zink finger, trunk, β-sheet, helical and gelsolin domain. Although most of the SEC23B mutations are sporadic events, 4 mutations (R14W, E109K, R497C, I318T) accounted for more than 50% of mutated alleles. The aim of this study is the in vitro characterization of the R14W mutation, the most frequent variant in Italy, particularly in South of Italy (Russo R et al., Am J Hematol. in press). We used the human erythroleukemia cell line, HEL, as in vitro model because it is more similar to mature red cell. By using in silico tool ESyPred3D Web Server 1.0, we predicted that this aminoacidic substitution alters the zink finger domain 3D structure, when compared to wild type protein. This tool implements homology modeling approach followed by a final analysis with MODELLER release 4 in order to build a 3D model of the submitted protein (Lambert et al, 2002). However, when we transfected the SEC23B-R14W we observed a strong reduction of gene expression in the mutant when compared to SEC23B-wt construct by qRT-PCR. These results have been also confirmed at the protein level. In fact the protein expression of SEC23B-R14W showed a reduction comparable to gene expression respect to SEC23B-wt construct. Immunofluorescence analyses by confocal microscopy, were used for the investigation of the cellular localization of SEC23B-R1W protein and, interestingly, the localization of mutant protein was not changed when compared to that wt. Our data allow us to hypothesize that the mutation R14W gives rise to anomalous protein product quantity, but the protein function would be like not altered. Our findings demonstrated that the most frequent mutation found in Italy, SEC23B-R14W, results in a reduced half-life of the mutated mRNA, without altering the cellular localization in HEL cell line. SEC23B belongs to a multiproteic compelx that assembles with the others complex proteins in accordance with a specific structure. Each structure establishes a cargo selectivity. Further studies are necessary in order to understand what is the role of SEC23B in selectivity of the cargo in erythroid cells and how its disruption could determines the appearance of the principal pathological phenotype in CDAII patients, for example the hypoglycosilation of band 3. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Society of Hematology ; 2008
    In:  Blood Vol. 112, No. 11 ( 2008-11-16), p. 416-416
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 416-416
    Abstract: DMT1 (Nramp2/DCT1), also called SLC11A2, is a widely expressed metal-iron transporter that is vital for iron absorption (enterocytes) and utilization (erythroid precursors). Transcription of the mammalian SLC11A2 genes coding for DMT1 gives rise to four variant mRNA transcripts that differ in their tissue distribution and regulation. A 5′-end mRNA processing gives two isoforms: the first one starts from exon 1A, and the other from exon 1B. Whereas the 1B isoform is ubiquitous, the 1A isoform is tissue-specific, expressed predominantly in the duodenum and kidney. In addition, variable 3′-end processing yields two transcripts that differ in their 3′-translated regions and UTRs (untranslated regions). One of the transcripts, denoted IRE, contains in its 3′-UTR an iron-responsive element (IRE) that may alter mRNA stability according to iron status, so that the IRE-containing and non-IRE forms may be expected to differ mainly in their regulation by iron. In humans, DMT1 mutation causes microcytic hypochromic anemia due to decreased erythroid iron utilization with liver iron overload, high transferrin saturation and mildly elevated serum ferritin. MicroRNAs (miRNAs) are single-stranded RNAs of ~22 nucleotides in length, and they constitute a novel class of gene regulators. In animals, miRNAs exert their regulatory effects by binding to imperfect complementary sites within the 3′-untranslated regions (3′-UTRs) of their mRNA targets affecting protein translation. They are involved in a variety of important biological processes, such as developmental timing and patterning, apoptosis, hematopoietic differentiation, cell proliferation, organ development and tumorigenesis. The aim of our work was to study the microRNAs regulation of the gene involved in iron metabolism, particularly the microRNAs that target the DMT1 non IRE isoform gene. In-silico analysis of the mirBase targets database (Griffiths-Jones, 2004) was directed towards the identification of miRNAs potentially targeting DMT1 (non IRE isoform). Among others, we selected microRNA LET-7D because it was downregulated during eritroid differentiation of CD34+ cells and K562 cell line while DMT1 was upregulated, furthermore it belongs to a conserved microRNAs family. The over expression of the pre-microRNA LET-7D in K562 cell line down regulated both the mRNA and the protein expression of DMT1 (non IRE isoform). On the other hand, the DMT1 (IRE isoform) was up regulated. As control, we mutated the seed region of microRNA and the over expression of mutated microRNA LET-7D in K562 cell line did not interfere with DMT1 expression. In order to evaluate whether DMT1 was effectively a target of miR-LET-7D, the DMT1 3′UTR was cloned downstream of a luciferase reporter gene vector; the K562 cells line were then transfected with the over expressing vector and the reporter construct, with the relative luciferase activity showing that miRNA LET- 7D transfection led to decreased activity of the reporter gene, thus indicating binding with the 3′UTR and destabilisation of productive translation of luciferase mRNA. As controls, DMT1 3′UTR mutated in the miRNA LET-7D binding site was not affected by miRNA LET-7D. We are performing functional study to evaluate the state of iron metabolism in K562 cell line (stable clone) overexpressing the microRNA LET-7D. Here, we identify the microRNA LET-7D as a regulator of the iron metabolism through its targeting of the DMT1 (non IRE isoform).
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 1993-1993
    Abstract: Iron loading anemias are characterized by ineffective erythropoiesis and iron overload. This group of anemias includes thalassemia syndromes, congenital dyserythropoietic anemias (CDA), and some forms of congenital hemolytic anemias. Among them pyruvate kinase deficiency (PKD) has been shown to develop iron overload also in absence of transfusions suggesting dyserythropoietic features. Moreover, severe forms can be misdiagnosed as CDA due to bone marrow abnormalities and ineffective erythropoiesis further supporting this evidences. The hormone erythroferrone (hERFE) is produced by erythroblasts in response to erythropoietin (EPO), and acts by suppressing hepcidin, thereby increasing iron absorption and mobilisation for erythropoiesis demand. The ERFE-hepcidin axis seems to play a crucial role in the pathogenesis of these disorders; an increased erythroferrone release by immature erythroid cells results in hepcidin suppression and secondary iron overload that could finally results in ineffective erythropoiesis and anemia. To investigate the pathophysiological basis of iron overload in PKD, we analysed the levels of hERFE, EPO, hepcidin, and soluble transferrin receptor (sTFR) in a large group of 41 PKD patients equally distributed by gender, age and severity. The results were analysed in comparison with two groups of patients affected by hemolytic anemia with overt dyserythropoiesis (42 patients with CDA type II) and with congenital hemolytic anemia due to RBC membrane defects (51 patients with hereditary spherocytosis [HS]), respectively. Demographic, hematologic, and biochemical features of the three groups of patients are reported in the table. Among the PKD patients, 18/41 were & lt;18 yrs, median Hb level at the time of the study was 9.05g/dL (range 5.5-14.5), 12 underwent splenectomy, 28 ever received at least three transfusions their life, 14 of them transfusion dependent ( & gt;6 tx/yrs). Mean ferritin levels at the time of the study were 546 ng/ml (range 59-4990), 15/41 patients requiring chelation therapy for iron overload developed also in absence of transfusions. As expected, CDAII patients showed decreased hepcidin levels (3.74 ng/mL; n.v. 17.25, P & lt;0.001) associated with increased erythropoietin (62.7 IU/L, n.v. 6.5, P=0.01) and hERFE (24.8 ng/mL, n.v. 1, P & lt;0.0001). On the contrary, HS showed increased hepcidin, with less marked increased of ERFE (9.9 ng/mL, P=0.02) and EPO (36.4IU/L, P=0.005). In PKD patients we observed decreased hepcidin levels (7.15 ng/mL, P=0.03)), increased hERFE (18ng/mL, P & lt;0.0001) and EPO (75.6 IU/L, P=0.009). Instead, sTFR was equally increased in the three groups of patients (Figure). Interestingly, by comparing the three groups of patients, PKD showed dyserythropoietic features as evidenced by the observation of intermediate values between HS and CDAII of hepcidin (P=0.007 PKD v CDAII and P=0.0002 PKD vs HS), hEFRE, and sTFR. This study provides the first analysis of the main regulators of systemic iron homeostasis in PK deficiency compared either with the model of a structural RBC defect (HS) or with the typical model of dyserythropoietic anemia with ineffective erythropoiesis, such as CDAII. These data provide evidence of the dyserythropoietic features of PK deficiency, underlining the need of accurate diagnosis and paving the way of novel therapeutic approaches in PK deficiency. Zaninoni A. and Russo R. equally contributed to the study Figure 1 Figure 1. Disclosures Fattizzo: Kira: Speakers Bureau; Alexion: Speakers Bureau; Novartis: Speakers Bureau; Momenta: Honoraria, Speakers Bureau; Annexon: Consultancy; Apellis: Speakers Bureau; Amgen: Honoraria, Speakers Bureau. Barcellini: Incyte: Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria; Bioverativ: Membership on an entity's Board of Directors or advisory committees; Agios: Honoraria, Research Funding; Alexion Pharmaceuticals: Honoraria. Iolascon: Bluebird Bio: Other: Advisory Board; Celgene: Other: Advisory Board. Bianchi: Agios pharmaceutics: Consultancy, Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 128, No. 14 ( 2016-10-06), p. 1899-1902
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 133, No. 3 ( 2019-01-17), p. 252-265
    Abstract: Resolvins (Rvs), endogenous lipid mediators, play a key role in the resolution of inflammation. Sickle cell disease (SCD), a genetic disorder of hemoglobin, is characterized by inflammatory and vaso-occlusive pathologies. We document altered proresolving events following hypoxia/reperfusion in humanized SCD mice. We demonstrate novel protective actions of 17R-resolvin D1 (17R-RvD1; 7S, 8R, 17R-trihydroxy-4Z, 9E, 11E, 13Z, 15E, 19Z-docosahexaenoic acid) in reducing ex vivo human SCD blood leukocyte recruitment by microvascular endothelial cells and in vivo neutrophil adhesion and transmigration. In SCD mice exposed to hypoxia/reoxygenation, oral administration of 17R-RvD1 reduces systemic/local inflammation and vascular dysfunction in lung and kidney. The mechanism of action of 17R-RvD1 involves (1) enhancement of SCD erythrocytes and polymorphonuclear leukocyte efferocytosis, (2) blunting of NF-κB activation, and (3) a reduction in inflammatory cytokines, vascular activation markers, and E-selectin expression. Thus, 17R-RvD1 might represent a new therapeutic strategy for the inflammatory vasculopathy of SCD.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Hematology ; 2020
    In:  Hematology Vol. 2020, No. 1 ( 2020-12-4), p. 465-470
    In: Hematology, American Society of Hematology, Vol. 2020, No. 1 ( 2020-12-4), p. 465-470
    Abstract: Inherited microcytic anemias can be broadly classified into 3 subgroups: (1) defects in globin chains (hemoglobinopathies or thalassemias), (2) defects in heme synthesis, and (3) defects in iron availability or iron acquisition by the erythroid precursors. These conditions are characterized by a decreased availability of hemoglobin (Hb) components (globins, iron, and heme) that in turn causes a reduced Hb content in red cell precursors with subsequent delayed erythroid differentiation. Iron metabolism alterations remain central to the diagnosis of microcytic anemia, and, in general, the iron status has to be evaluated in cases of microcytosis. Besides the very common microcytic anemia due to acquired iron deficiency, a range of hereditary abnormalities that result in actual or functional iron deficiency are now being recognized. Atransferrinemia, DMT1 deficiency, ferroportin disease, and iron-refractory iron deficiency anemia are hereditary disorders due to iron metabolism abnormalities, some of which are associated with iron overload. Because causes of microcytosis other than iron deficiency should be considered, it is important to evaluate several other red blood cell and iron parameters in patients with a reduced mean corpuscular volume (MCV), including mean corpuscular hemoglobin, red blood cell distribution width, reticulocyte hemoglobin content, serum iron and serum ferritin levels, total iron-binding capacity, transferrin saturation, hemoglobin electrophoresis, and sometimes reticulocyte count. From the epidemiological perspective, hemoglobinopathies/thalassemias are the most common forms of hereditary microcytic anemia, ranging from inconsequential changes in MCV to severe anemia syndromes.
    Type of Medium: Online Resource
    ISSN: 1520-4391 , 1520-4383
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    detail.hit.zdb_id: 2084287-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3184-3184
    Abstract: Abstract 3184 Isolated Familial Pseudohyperkalemia (FP) is a dominant red cell trait characterized by cold-induced slow ‘passive leak’ of red cell K+ into plasma, first described in a large Scottish family from Edinburgh (Stewart GW, et al., 1979). Although in freshly obtained blood samples plasma [K+] was normal, it was increased when measured in blood stored at or below room temperature. This trait was unaccompanied by clinical symptoms or signs except for mild abnormalities of red cell shape. FP Lille was later described in a large Flemish kindred with morphologically normal red cells (Dagher G, et al., 1989; Vantyghem MC, et al., 1991). In this family, red cell K+ efflux measured in the presence of ouabain and bumetanide was normal at 37°C, but greatly increased at 22°C and 9°C. FP Lille mapped to 2q35-q36 (Carella M, et al., 2004), whereas FP Edinburgh mapped to 16q23-qter (Iolascon A, et al., 1999). Subsequently, asymptomatic cases FP Chiswick and FP Falkirk with remarkable increased MCV were reported (Haines PG, et al., 2001). Functional gene mapping and sequencing analysis of the candidate genes within the 2q35-q36 critical interval in three multigenerational FP families with 20 affected individuals identified two novel heterozygous missense mutations in the ABCB6 gene that cosegregated with disease phenotype. The two genomic substitutions altered two adjacent nucleotides within codon 375 of ABCB6, a porphyrin transporter that in erythrocyte membranes bears the Langereis blood group antigen system (Krishnamurthy PC, et al., 2006; Helias V, et al., 2012). Structural modeling predicts subtle changes in protein structure associated with either mutation. ABCB6 mRNA and protein levels increased during erythroid differentiation of CD34+ erythroid precursors (at 7 and 14 days of EPO induced differentiation), and of HEL and K562 erythroleukemia cells. However, the ABCB6 R375Q mutation altered neither levels of ABCB6 mRNA or protein, nor protein localization in mature erythrocytes or erythroid precursor cells. These data strongly suggest that missense mutations in residue 375 of the ABCB6 polypeptide either mediate the cold-induced K+ leak of chromosome 2-linked FP, or activate an independent, cold-induced cation permeability pathway of the red cell. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3009-3009
    Abstract: Abstract 3009 Poster Board II-985 Congenital dyserythropoietic anemia type II (CDA II) is an autosomal recessive disorder affecting the normal differentiation-proliferation pathway of the erythroid lineage. It comprises an anemia of variable severity, jaundice, and variable splenomegaly. Erythroid hyperplasia with binuclearity or multinuclearity involving late erythroblasts in the bone marrow (BM) is a key feature of the diagnosis (Iolascon, 2001). In addition, on electron microscopy, vesicles of endoplasmic reticulum (ER) appear to be running beneath the plasma membrane (Alloisio, 1996). The principal biochemical feature is the hypoglycosilation of several proteins, such as transferrin and band 3 (Anselstetter, 1977). Recently, we identified SEC23B as the CDA II causative gene (Schwarz, 2009). The SEC23B gene encodes the SEC23B component which is part of the cytoplasmic coat protein (COP)II complex. COPII coated vesicles bud from the endoplasmic reticulum to export newly synthesized proteins to the trans Golgi. In yeast, COPII coated vesicles form by the sequential binding of Sar1-GTP, the inner complex proteins Sec23-Sec24 and the outer complex components Sec13-Sec31 on the endoplasmic reticulum (ER) (Fromme, 2008). Our aim was to characterize the COPII complex in CD34+ progenitor cells during erythroid differentiation by gene expression analysis. Mononuclear cells from the peripheral blood of normal subjects were isolated on a Ficoll-Hypaque gradient and CD34+ progenitors were separated on immuno-affinity columns. For erythroid differentiation, CD34+ cells of three different pools were separately plated on plastic culture dishes in methylcellulose medium containing 3U/mL erythropoietin (EPO) (Pinho, 2008). The cells have been cultured for 7 and 14 days after EPO treatment. Quantitative real time (qRT)-PCR on CD34+ during erythroid differentiation was performed to assess the gene expression level. The relative gene expression was calculated by 2*(-ΔCt) method (Livak, 2001), using GAPDH gene as internal control (figure 1). Mammalian orthologues have been identified for each of the five proteins involved in COPII coat formation. In humans, two isoforms of Sec23, Sar1 and Sec31 and four mammalian isoforms of Sec24 have been reported (Kuge, 1994; Paccaud, 1996; Wendeler, 2007; Mancias, 2008; Shugrue, 1999; Tang, 2000; Stankewich, 2005). We already demonstrated that during normal erythropoiesis a SEC23A down regulation is associated to SEC23B upregulation. These data suggest that SEC23B mutants could disrupt the COPII complex in erythroid lineage, and consequently induce CDA II (Schwarz, 2009). On the contrary, the two isoforms of SAR1 showed the same trend during erythroid differentiation: however, SAR1A isoforms has an higher expression when compared to SAR1B isoform. Among the four SEC24 isoforms, SEC24A, B and C showed the same increased expression after EPO treatment; SEC24D, indeed, showed a decreasing trend during differentiation time. Only one form of mammalian SEC13 has been described (Shaywitz, 1995), and it showed an increased expression after EPO treatment. Between SEC31 mammalian isoforms, SEC31A showed overall an higher gene expression compared to SEC31B gene. The gene expression analysis of SEC12, the transmembrane guanine nucleotide exchange factor that catalyzes the COPII vesicle formation by GDP-GTP exchange on Sar1 (Sato, 2004), revealed an higher mRNA level at 14 days when compared to 7 days after EPO treatment. Here, we have identified the isoforms of COPII complex most expressed during erythroid differentiation. This study could allow us to clarify the role of each COPII gene in erythroid lineage, and to identify other genes potentially involved in CDA II pathogenesis.Figure 1.Gene expression profile during CD34+ erythroid differentiation. Data are presented as mean ± standard deviation. *p value 〈 0.05 calculated on day 0 by Student t test corrected by Bonferroni method.Figure 1. Gene expression profile during CD34+ erythroid differentiation. Data are presented as mean ± standard deviation. *p value 〈 0.05 calculated on day 0 by Student t test corrected by Bonferroni method. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2132-2132
    Abstract: Introduction Isolated Familial Pseudohyperkalemia (FP) is a dominant red cell trait characterized by cold-induced slow 'passive leak' of red cell K+ into plasma, first described in a large Scottish family from Edinburgh (Stewart GW, et al., 1979). Although in freshly obtained blood samples plasma [K+] was normal, it was increased when measured in blood stored at or below room temperature. This trait was accompanied by mild abnormalities of red cell shape. Functional gene mapping and sequencing analysis of the candidate genes within the 2q35-q36 critical interval in three multigenerational FP families with 20 affected individuals identified two novel heterozygous missense mutations in the ABCB6 gene that cosegregated with disease phenotype (Andolfo I. et al., 2013). The two genomic substitutions altered two adjacent nucleotides within codon 375 of ABCB6, a porphyrin transporter that in erythrocyte membranes bears the Langereis blood group antigen system (Krishnamurthy PC, et al., 2006; Helias V, et al., 2012). Recently, the ABCB6 mutation R723Q was found in two patients with FP (Bawazir W, et al.,2014). Of note, both patients presented as blood donors, and increased cold-induced potassium leak was demonstrated. The transfusion of pseudohyperkalemic blood has clinical implications especially for neonates and infants receiving large-volume RBC transfusions. In this study we analyzed one additional family and report the first functional characterization of an ABCB6 mutation, towards understanding the pathogenic mechanism of FP. Moreover, we screened an Italian blood donor population for the R276W variant of the ABCB6 gene. Methods DNA was obtained for genetic analysis from affected and unaffected family members, after signed informed consent, according to the Declaration of Helsinki. The search for mutations was performed by direct sequencing of the ABCB6 gene. cDNAs encoding full-length wildtype ABCB6 were cloned into pcDNA3.1. Our patients' novel point mutation (c.826C 〉 T, p.R276W) was introduced into pcDNA3.1-ABCB6 by site-directed mutagenesis. WT and mutant constructs were transfected into HEK-293 cells and the cells were maintained at 30°C for 72 hrs to evaluate the effects of reduced temperature. After transfection, the cells were incubated in a medium containing 86rubidium (86Rb+) as a surrogate for K+. 86Rb+ was determined in cell lysate, and K content of extracellular medium was determined by atomic absorption spectrometry. Results We found the heterozygous mutation c.826G 〉 T, p. R276W in an Irish family. This mutation is annotated in public databases as single nucleotide variants (SNVs), and is predicted by PolyPhen2 and SIFT to be damaging. Variant R276W showed a minor allele frequency (MAF) of 1.3:100 confirming that many patients with FP could be present in the blood donor population. R276W and previously identified ABCB6 variants R375Q and R375Wwere overexpressed in HEK-293 cells to characterize the functional properties of these variants. Expression of ABCB6 mutants showed no change in RNA or polypeptide levels. However, measurement of ouabain- and bumetanide-resistant net cation flux demonstrated a greater loss of cell K from mutant ABCB6-expressing cells than from WT ABCB6-expressing cells. The high allele frequency of ABCB6 variant R276W prompted a genetic screen of 327 blood donors. The variant was present in 0.3% (1/327) of the donor cohort, and this single subject with ABCB6 R276W exhibited slightly increased MCV and variably increased plasma K+ concentrations. Conclusions: Our findings demonstrate that missense mutations in ABCB6 lead to increased K+ efflux from RBCs in FP patients. Storage of FP blood can cause a significant increase in blood K+ levels, with especially serious clinical implications for neonates and infants receiving large-volume transfusions of whole blood. Furthermore, the prevalence of FP might be underestimated, since patients with FP can be asymptomatic and thus undetected in the donor population. Screening for the most frequent ABCB6 variation, R276W, confirmed that FP patients are present in the Italian blood donor population. In the future, genetic tests for FP could be added to blood donor prescreening to improve the quality and safety of donor units. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...