Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Vox Sang ; 119(5): 496-504, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38326223

RESUMEN

BACKGROUND AND OBJECTIVES: Polymorphic molecules expressed on the surface of certain blood cells are traditionally categorized as blood groups and human platelet or neutrophil antigens. CD36 is widely considered a platelet antigen (Naka) and anti-CD36 can cause foetal/neonatal alloimmune thrombocytopenia (FNAIT) in CD36-negative pregnant women. CD36 is used as a marker of differentiation in early erythroid culture. During the experimental culture of CD34+ cells from random blood donors, we observed that one individual lacked CD36. We sought to investigate this observation further and determine if CD36 fulfils the International Society of Blood Transfusion criteria for becoming a blood group. MATERIALS AND METHODS: Surface markers were monitored by flow cytometry on developing cells during the erythroid culture of CD34+ cells. Genetic and flow cytometric analyses on peripheral blood cells were performed. Proteomic datasets were analysed, and clinical case reports involving anti-CD36 and foetal anaemia were scrutinized. RESULTS: Sequencing of CD36-cDNA identified homozygosity for c.1133G>T/p.Gly378Val in the CD36-negative donor. The minor allele frequency of rs146027667:T is 0.1% globally and results in abolished CD36 expression. CD36 has been considered absent from mature red blood cells (RBCs); however, we detected CD36 expression on RBCs and reticulocytes from 20 blood donors. By mining reticulocyte and RBC datasets, we found evidence for CD36-derived peptides enriched in the membrane fractions. Finally, our literature review revealed severe cases of foetal anaemia attributed to anti-CD36. CONCLUSIONS: Based on these findings, we conclude that CD36 fulfils the criteria for becoming a new blood group system and that anti-CD36 is implicated not only in FNAIT but also foetal anaemia.


Asunto(s)
Antígenos CD36 , Eritrocitos , Antígenos CD36/genética , Antígenos CD36/sangre , Humanos , Femenino , Eritrocitos/metabolismo , Embarazo , Antígenos de Grupos Sanguíneos/genética , Masculino , Recién Nacido , Trombocitopenia Neonatal Aloinmune/sangre , Trombocitopenia Neonatal Aloinmune/genética , Relevancia Clínica
4.
Transfusion ; 63(12): 2297-2310, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37921035

RESUMEN

BACKGROUND: Accurate blood type data are essential for blood bank management, but due to costs, few of 43 blood group systems are routinely determined in Danish blood banks. However, a more comprehensive dataset of blood types is useful in scenarios such as rare blood type allocation. We aimed to investigate the viability and accuracy of predicting blood types by leveraging an existing dataset of imputed genotypes for two cohorts of approximately 90,000 each (Danish Blood Donor Study and Copenhagen Biobank) and present a more comprehensive overview of blood types for our Danish donor cohort. STUDY DESIGN AND METHODS: Blood types were predicted from genome array data using known variant determinants. Prediction accuracy was confirmed by comparing with preexisting serological blood types. The Vel blood group was used to test the viability of using genetic prediction to narrow down the list of candidate donors with rare blood types. RESULTS: Predicted phenotypes showed a high balanced accuracy >99.5% in most cases: A, B, C/c, Coa /Cob , Doa /Dob , E/e, Jka /Jkb , Kna /Knb , Kpa /Kpb , M/N, S/s, Sda , Se, and Yta /Ytb , while some performed slightly worse: Fya /Fyb , K/k, Lua /Lub , and Vel ~99%-98% and CW and P1 ~96%. Genetic prediction identified 70 potential Vel negatives in our cohort, 64 of whom were confirmed correct using polymerase chain reaction (negative predictive value: 91.5%). DISCUSSION: High genetic prediction accuracy in most blood groups demonstrated the viability of generating blood types using preexisting genotype data at no cost and successfully narrowed the pool of potential individuals with the rare Vel-negative phenotype from 180,000 to 70.


Asunto(s)
Antígenos de Grupos Sanguíneos , Humanos , Antígenos de Grupos Sanguíneos/genética , Genotipo , Fenotipo , Donantes de Sangre , Reacción en Cadena de la Polimerasa
5.
Nat Commun ; 14(1): 5001, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37591894

RESUMEN

Genetic determinants underlying most human blood groups are now clarified but variation in expression levels remains largely unexplored. By developing a bioinformatics pipeline analyzing GATA1/Chromatin immunoprecipitation followed by sequencing (ChIP-seq) datasets, we identify 193 potential regulatory sites in 33 blood-group genes. As proof-of-concept, we aimed to delineate the low-expressing complement receptor 1 (CR1) Helgeson phenotype on erythrocytes, which is correlated with several diseases and protects against severe malaria. We demonstrate that two candidate CR1 enhancer motifs in intron 4 bind GATA1 and drive transcription. Both are functionally abolished by naturally-occurring SNVs. Erythrocyte CR1-mRNA and CR1 levels correlate dose-dependently with genotype of one SNV (rs11117991) in two healthy donor cohorts. Haplotype analysis of rs11117991 with previously proposed markers for Helgeson shows high linkage disequilibrium in Europeans but explains the poor prediction reported for Africans. These data resolve the longstanding debate on the genetic basis of inherited low CR1 and form a systematic starting point to investigate the blood group regulome.


Asunto(s)
Células Eritroides , Factor de Transcripción GATA1 , Receptores de Complemento 3b , Humanos , Pueblo Africano , Biología Computacional , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Genotipo , Intrones , Fenotipo , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/metabolismo , Secuenciación de Inmunoprecipitación de Cromatina , Células Eritroides/metabolismo , Pueblo Europeo
6.
Vox Sang ; 118(8): 690-694, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37265146

RESUMEN

BACKGROUND AND OBJECTIVES: The extremely rare Rhnull phenotype is characterized by the absence of all Rh antigens on erythrocytes. It is divided into the regulator and amorph types based on the underlying genetic background. The more common regulator type depends on critical variants silencing RHAG, which encodes RhAG glycoprotein, necessary for RhD/RhCE expression. Rhnull cells have altered expression of glycophorin B and LW glycoprotein. MATERIALS AND METHODS: Four unrelated Rhnull individuals were investigated. Serological testing was performed according to standard blood bank practice. RHD/RHCE and S/s allele-specific Polymerase chain reaction (PCR) genotyping was done on genomic DNA using in-house PCR assays. RHAG, and in some cases also RHD/RHCE, were sequenced. Initial s phenotyping results triggered additional serological investigation. RESULTS: Anti-Rh29 was identified in all four individuals. Extended typing with anti-S and anti-s showed that the three samples predicted to type as s+ failed to react with 2 of 5 anti-s. Sequence analysis of all 10 RHAG exons and the immediate intron/exon boundaries revealed a single nucleotide variant in the 3'-end of intron 6, c.946 -2a>g in all samples. RHD/RHCE showed no alterations. CONCLUSION: A novel Nordic Rhnull allele was identified. In addition, it was shown that s+ Rhnull red blood cells are not only U- but also have qualitative changes in their s antigen expression.


Asunto(s)
Antígenos de Grupos Sanguíneos , Sistema del Grupo Sanguíneo Rh-Hr , Sistema del Grupo Sanguíneo Rh-Hr/genética , Fenotipo , Secuencia de Bases , Reacción en Cadena de la Polimerasa
7.
Pathogens ; 12(6)2023 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-37375493

RESUMEN

Babesia is spread to humans via ticks or blood transfusions. Severity of Plasmodium falciparum malaria is strongly correlated to the ABO blood group of the patient. Babesia divergens is an intraerythrocytic parasite with many similarities to malaria, but the impact of ABO on the susceptibility to and progression of the infection in humans is unknown. We have now cultured B. divergens in human group A, B and O erythrocytes in vitro and measured rates of multiplication. The predilection for the different erythrocyte types was also determined using an in vitro erythrocyte preference assay when the parasites were grown in group A, B or O erythrocytes over time and then offered to invade differently stained erythrocytes of all the blood types at the same time. The results showed no difference in multiplication rates for the different blood types, and the parasite exhibited no obvious morphological differences in the different blood types. When cultured first in one blood type and then offered to grow in the others, the preference assay showed that there was no difference between the A, B or O blood groups. In conclusion, this indicates that individuals of the different ABO blood types are likely to be equally susceptible to B. divergens infections.

8.
Microbes Infect ; 25(3): 105060, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36270601

RESUMEN

Plasmodium falciparum malaria can cause severe anemia. Even after treatment, hematocrit can decrease. The role of autoantibodies against erythrocytes is not clearly elucidated and how common they are, or what they are directed against, is still largely unknown. We have investigated antibodies against erythrocytes in healthy adult men living in a highly malaria endemic area in Uganda. We found antibodies in more than half of the individuals, which is significantly more than in a non-endemic area (Sweden). Some of the Ugandan samples had a broad reactivity where it was not possible to determine the exact target of the autoantibodies, but we also found specific antibodies directed against erythrocyte surface antigens known to be of importance for merozoite invasion such as glycophorin A (anti-Ena, anti-M) and glycophorin B (anti-U, anti-S). In addition, several autoantibodies had partial specificities against glycophorin C and the blood group systems Rh, Diego (located on Band 3), Duffy (located on ACKR1), and Cromer (located on CD55), all of which have been described to be important for malaria and therefore of interest for understanding how autoantibodies could potentially stop parasites from entering the erythrocyte. In conclusion, specific autoantibodies against erythrocytes are common in a malaria endemic area.


Asunto(s)
Malaria Falciparum , Malaria , Masculino , Humanos , Autoanticuerpos , Plasmodium falciparum , Eritrocitos , Antígenos de Protozoos , Proteínas Protozoarias , Malaria Falciparum/epidemiología , Malaria Falciparum/metabolismo
11.
Vox Sang ; 117(11): 1332-1344, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36121188

RESUMEN

BACKGROUND AND OBJECTIVES: Under the ISBT, the Working Party (WP) for Red Cell Immunogenetics and Blood Group Terminology is charged with ratifying blood group systems, antigens and alleles. This report presents the outcomes from four WP business meetings, one located in Basel in 2019 and three held as virtual meetings during the COVID-19 pandemic in 2020 and 2021. MATERIALS AND METHODS: As in previous meetings, matters pertaining to blood group antigen nomenclature were discussed. New blood group systems and antigens were approved and named according to the serologic, genetic, biochemical and cell biological evidence presented. RESULTS: Seven new blood group systems, KANNO (defined numerically as ISBT 037), SID (038), CTL2 (039), PEL (040), MAM (041), EMM (042) and ABCC1 (043) were ratified. Two (039 and 043) were de novo discoveries, and the remainder comprised reported antigens where the causal genes were previously unknown. A further 15 blood group antigens were added to the existing blood group systems: MNS (002), RH (004), LU (005), DI (010), SC (013), GE (020), KN (022), JMH (026) and RHAG (030). CONCLUSION: The ISBT now recognizes 378 antigens, of which 345 are clustered within 43 blood group systems while 33 still have an unknown genetic basis. The ongoing discovery of new blood group systems and antigens underscores the diverse and complex biology of the red cell membrane. The WP continues to update the blood group antigen tables and the allele nomenclature tables. These can be found on the ISBT website (http://www.isbtweb.org/working-parties/red-cell-immunogenetics-and-blood-group-terminology/).


Asunto(s)
Antígenos de Grupos Sanguíneos , COVID-19 , Eritrocitos , Humanos , Antígenos de Grupos Sanguíneos/genética , Transfusión Sanguínea , Inmunogenética , Pandemias , Eritrocitos/inmunología
12.
Transfus Med ; 32(2): 168-174, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33987889

RESUMEN

OBJECTIVE: To evaluate the effect of platelet:erythrocyte (P:E) ratios on Plasmodium falciparum erythrocyte invasion. BACKGROUND: Recent reports have shown that platelets are directly involved in the immune response towards P. falciparum during erythrocyte invasion. However, the literature both supports and conflicts with a role for platelets in limiting invasion. Also, the effect of platelet numbers on invasion (parasitemia) has not been thoroughly investigated. METHODS/MATERIALS: The P. falciparum strains FCR3S1.2 and W2mef were cultured with group O erythrocytes. The cultures were synchronised and supplemented with pooled platelets at P:E ratios ranging from 1:100 to 1:2. Parasitemia was measured at 40 h by flow cytometry and by microscopy of blood smears. RESULTS: A linear relationship was observed between reduced invasion and increased platelet numbers at P:E ratios ranging from 1:100 to 1:20. However, this effect was reversed at lower ratios (1:10-1:2). Microscopic evaluation revealed aggregation and attachment of platelets to erythrocytes, but not specifically to parasitised erythrocytes. CONCLUSION: We have shown that under physiological P:E ratios (approx. 1:10-1:40), platelets inhibited P. falciparum invasion in a dose-dependent manner. At ratios of 1:10 and below, platelets did not further increase the inhibitory effect and, although the trend was reversed, inhibition was still maintained.


Asunto(s)
Malaria Falciparum , Plasmodium falciparum , Plaquetas , Eritrocitos , Humanos , Parasitemia
13.
Transfus Med Rev ; 35(2): 67-69, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33975731
15.
Free Radic Biol Med ; 162: 149-159, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32092412

RESUMEN

During red blood cell (RBC) lysis hemoglobin and heme leak out of the cells and cause damage to the endothelium and nearby tissue. Protective mechanisms exist; however, these systems are not sufficient in diseases with increased extravascular hemolysis e.g. hemolytic anemia. α1-microglobulin (A1M) is a ubiquitous reductase and radical- and heme-binding protein with antioxidation properties. Although present in the circulation in micromolar concentrations, its function in blood is unclear. Here, we show that A1M provides RBC stability. A1M-/- mice display abnormal RBC morphology, reminiscent of macrocytic anemia conditions, i.e. fewer, larger and more heterogeneous cells. Recombinant human A1M (rA1M) reduced in vitro hemolysis of murine RBC against spontaneous, osmotic and heme-induced stress. Moreover, A1M is taken up by human RBCs both in vitro and in vivo. Similarly, rA1M also protected human RBCs against in vitro spontaneous, osmotic, heme- and radical-induced hemolysis as shown by significantly reduced leakage of hemoglobin and LDH. Addition of rA1M resulted in decreased hemolysis compared to addition of the heme-binding protein hemopexin and the radical-scavenging and reducing agents ascorbic acid and Trolox (vitamin E). Furthermore, rA1M significantly reduced spontaneous and heme-induced fetal RBC cell death. Addition of A1M to human whole blood resulted in a significant reduction of hemolysis, whereas removal of A1M from whole blood resulted in increased hemolysis. We conclude that A1M has a protective function in reducing hemolysis which is neither specific to the origin of hemolytic insult, nor species specific.


Asunto(s)
Anemia Macrocítica , Hemólisis , alfa-Globulinas , Animales , Muerte Celular , Eritrocitos , Humanos , Ratones , Ratones Noqueados , Fenotipo
16.
Int J Mol Sci ; 21(19)2020 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-33008134

RESUMEN

α1-microglobulin (A1M) is a small protein present in vertebrates including humans. It has several physiologically relevant properties, including binding of heme and radicals as well as enzymatic reduction, that are used in the protection of cells and tissue. Research has revealed that A1M can ameliorate heme and ROS-induced injuries in cell cultures, organs, explants and animal models. Recently, it was shown that A1M could reduce hemolysis in vitro, observed with several different types of insults and sources of RBCs. In addition, in a recently published study, it was observed that mice lacking A1M (A1M-KO) developed a macrocytic anemia phenotype. Altogether, this suggests that A1M may have a role in RBC development, stability and turnover. This opens up the possibility of utilizing A1M for therapeutic purposes in pathological conditions involving erythropoietic and hemolytic abnormalities. Here, we provide an overview of A1M and its potential therapeutic effect in the context of the following erythropoietic and hemolytic conditions: Diamond-Blackfan anemia (DBA), 5q-minus myelodysplastic syndrome (5q-MDS), blood transfusions (including storage), intraventricular hemorrhage (IVH), preeclampsia (PE) and atherosclerosis.


Asunto(s)
alfa-Globulinas/genética , Eritrocitos/metabolismo , Eritropoyesis/genética , Síndromes Mielodisplásicos/genética , alfa-Globulinas/metabolismo , Animales , Femenino , Hemo/genética , Hemo/metabolismo , Hemólisis/genética , Homeostasis , Humanos , Ratones , Ratones Noqueados , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/terapia
17.
Transfus Med Hemother ; 47(4): 326-336, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32884505

RESUMEN

The U antigen (MNS5) is one of 49 antigens belonging to the MNS blood group system (ISBT002) carried on glycophorins A (GPA) and B (GPB). U is present on the red blood cells in almost all Europeans and Asians but absent in approximately 1.0% of Black Africans. U negativity coincides with negativity for S (MNS3) and s (MNS4) on GPB, thus be called S-s-U-, and is thought to arise from homozygous deletion of GYPB. Little is known about the molecular background of these deletions. Bioinformatic analysis of the 1000 Genomes Project data revealed several candidate regions with apparent deletions in GYPB. Highly specific Gap-PCRs, only resulting in positive amplification from DNAs with deletions present, allowed for the exact genetic localization of 3 different breakpoints; 110.24- and 103.26-kb deletions were proven to be the most frequent in Black Americans and Africans. Among 157 CEPH DNAs, deletions in 6 out of 8 African ethnicities were present. Allele frequencies of the deletions within African ethnicities varied greatly and reached a cumulative 23.3% among the Mbuti Pygmy people from the Congo. Similar observations were made for U+var alleles, known to cause strongly reduced GPB expression. The 110- and 103-kb deletional GYPB haplotypes were found to represent the most prevalent hereditary factors causative of the MNS blood group phenotype S-s-U-. Respective GYPB deletions are now accessible by molecular detection of homo- and hemizygous transmission.

18.
Int J Mol Sci ; 21(16)2020 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-32823905

RESUMEN

Preeclampsia (PE) is a human specific syndrome with unknown etiology causing maternal and fetal morbidities and mortalities. In PE, maternal inflammatory responses are more exaggerated if the fetus is male than female. Other pregnancy complications such as spontaneous abortions are also more common if the fetus is male. Recent transcriptome findings showed an increased expression of CD99 in erythroid cells from male cord blood in PE. The single nucleotide polymorphism (SNP) rs311103, located in a GATA-binding site in a regulatory region on the X/Y chromosomes, governs a coordinated expression of the Xg blood group members CD99 and Xga in hematopoietic cells in a sex-dependent fashion. The rs311103C disrupts the GATA-binding site, resulting in decreased CD99 expression. We aimed to investigate the association between PE and the allele frequency of rs311103 in pregnancies in a fetal sex-dependent fashion. In a case-controlled study, we included 241 pregnant women, i.e., 105 PE cases and 136 normotensive controls. A SNP allelic discrimination analysis was performed on DNA from maternal venous blood and fetal cord blood by qPCR. A statistically significant association was observed between rs311103 allele frequency and PE in mothers carrying male fetuses. Therefore, the rs311103 genotype may play a role in the pathogenesis of PE in a fetal sex-specific manner.


Asunto(s)
Antígeno 12E7/genética , Feto/patología , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple/genética , Preeclampsia/genética , Adulto , Etiopía , Femenino , Frecuencia de los Genes/genética , Humanos , Masculino , Embarazo , Resultado del Embarazo
19.
Nat Commun ; 11(1): 3569, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32678083

RESUMEN

The clinically important MAM blood group antigen is present on haematopoietic cells of all humans except rare MAM-negative individuals. Its molecular basis is unknown. By whole-exome sequencing we identify EMP3, encoding epithelial membrane protein 3 (EMP3), as a candidate gene, then demonstrate inactivating mutations in ten known MAM-negative individuals. We show that EMP3, a purported tumour suppressor in various solid tumours, is expressed in erythroid cells. Disruption of EMP3 by CRISPR/Cas9 gene editing in an immortalised human erythroid cell line (BEL-A2) abolishes MAM expression. We find EMP3 to associate with, and stabilise, CD44 in the plasma membrane. Furthermore, cultured erythroid progenitor cells from MAM-negative individuals show markedly increased proliferation and higher reticulocyte yields, suggesting an important regulatory role for EMP3 in erythropoiesis and control of cell production. Our data establish MAM as a new blood group system and demonstrate an interaction of EMP3 with the cell surface signalling molecule CD44.


Asunto(s)
Antígenos de Grupos Sanguíneos/genética , Proliferación Celular , Células Eritroides/citología , Glicoproteínas de Membrana/genética , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/metabolismo , Plaquetas/metabolismo , Células Cultivadas , Membrana Eritrocítica/metabolismo , Células Eritroides/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Mutación , Fenotipo , Unión Proteica , Secuenciación del Exoma
20.
Immunohematology ; 36(1): 4-6, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32324038

RESUMEN

CONCLUSIONS: This update of the Xg blood group system (Johnson NC. XG: The forgotten blood group system. Immunohematology 2011;27:68-71) notes the identification of a cis-regulatory element of both XG and CD99 expression, remarkably by two independent groups during 2018, and confirmed by another in 2019. A single nucleotide change at the XG locus (rs311103) abolishes GATA1 binding and suppresses both XG and CD99. The last blood group system to resist elucidation of its genetic basis was thereby resolved. Soon afterwards, it was discovered that the rare anti-Xga response, mainly seen in men, is produced by individuals primarily carrying a large deletion in the X chromosome that truncates XG and leads to the Xgnull phenotype.


Asunto(s)
Antígenos de Grupos Sanguíneos/inmunología , Humanos , Masculino , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA