Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Transfusion ; 63(8): 1441-1446, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37165957

RESUMO

BACKGROUND: The Kidd blood group gene SLC14A1 (JK) accounts for approximately 20 Kb from initiation codon to stop codon in the genome. In genomic DNA analysis using Sanger sequencing or short-read-based next generation sequencing, it is difficult to determine the cis or trans positions of single nucleotide variations (SNVs), which are occasionally more than 1 Kb away from each other. We aimed to determine the complete nucleotide sequence of a 20-Kb genomic DNA amplicon to characterize the JK allelic variants associated with Kidd antigen silencing in a blood donor. STUDY DESIGN AND METHODS: The Jk(a-b-) phenotype was identified in this donor by standard serological typing. A DNA sample obtained from whole blood was amplified by long-range PCR to obtain a 20-Kb fragment of the SLC14A1 gene, including the initiation and stop codons. The fragment was then analyzed by Sanger sequencing and single-molecule sequencing. Transfection and expression studies were performed in CHO cells using the expression vector construct of JK alleles. RESULTS: Sanger sequencing and single-molecule sequencing revealed that the donor was heterozygous with JK*01 having c.276G>A (rs763262711, p.Trp92Ter) and JK*02 having c.499A>G (rs2298719, p.Met167Val), c.588A>G (rs2298718, p.Pro196Pro), and c.743C>A (p.Ala248Asp). The two JK alleles identified have not been previously described. Transfection and expression studies indicated that the CHO cells transfected with JK*02 having c.743C>A did not express the Jkb and Jk3 antigens. CONCLUSIONS: We identified new JK silencing alleles and their critical SNVs by single-molecule sequencing and the findings were confirmed by transfection and expression studies.


Assuntos
DNA , Sistema do Grupo Sanguíneo Kidd , Animais , Cricetinae , Sistema do Grupo Sanguíneo Kidd/genética , Alelos , Cricetulus , Heterozigoto
2.
Vox Sang ; 118(5): 392-397, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36908196

RESUMO

BACKGROUND AND OBJECTIVES: The RHAG blood group system contains five antigens: Duclos (RHAG001), Ola (RHAG002), DSLK (RHAG003), Kg (RHAG005) and SHER (RHAG006). Individuals who are DSLK-negative and Kg-positive have the same allele RHAG*01.-3, with a single-nucleotide variation (rs144305805), c.490A>C (p.Lys164Gln), in exon 3 of the RHAG gene. We aimed to confirm whether DSLK and Kg are antithetical antigens. MATERIALS AND METHODS: Blood samples of the original DSLK-negative proband with anti-DSLK, her son and another DSLK-negative individual were examined. The RHAG gene was analysed by polymerase chain reaction and Sanger sequencing. Immunocomplex capture fluorescence assays (ICFAs) and monocyte phagocytosis assays were performed to characterize the anti-DSLK antibody. Cross-testing of alloanti-DSLK and monoclonal anti-Kg (OSK46) was performed using transduced HEK293 cells by inducing the construct of expression vectors encoding wild-type RHAG*01 or the variant RHAG*01.-3. RESULTS: ICFA using monoclonal anti-RHAG (LA18.18) revealed that the anti-DSLK and anti-Kg antibodies reacted with the wild-type and variant RhAG (Rh-associated glycoprotein), respectively. The proband and a DSLK-negative individual appeared to be homozygous for variant RHAG*01.-3, and the proband's son was typed as RHAG*01/RHAG*01.-3 heterozygote. HEK293 cells with wild-type RhAG reacted with the anti-DSLK but not anti-Kg antibody, whereas HEK293 cells expressing the variant RhAG reacted with the anti-Kg but not anti-DSLK antibody. Monocyte phagocytosis assays indicated that 64% of red cells sensitized with anti-DSLK were phagocytosed by monocytes. CONCLUSION: Our results demonstrate that DSLK and Kg are antithetical antigens in the RHAG blood group system. Anti-DSLK may be a clinically significant antibody.


Assuntos
Antígenos de Grupos Sanguíneos , Sistema do Grupo Sanguíneo Rh-Hr , Humanos , Feminino , Sistema do Grupo Sanguíneo Rh-Hr/genética , Células HEK293 , Glicoproteínas de Membrana/genética , Antígenos de Grupos Sanguíneos/genética , Eritrócitos/metabolismo , Proteínas Sanguíneas
3.
Vox Sang ; 117(10): 1235-1239, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36102098

RESUMO

BACKGROUND AND OBJECTIVES: The Xg blood group is composed of two antigens, Xga (XG1) and CD99 (XG2 and MIC2). The XG and CD99 are homologous genes located on pseudoautosomal region 1 of the X and Y chromosomes. The expressions of Xga and CD99 are co-regulated by a single nucleotide polymorphism (rs311103) in the GATA-1 binding region. Another mechanism of the Xg(a-) phenotype is the genomic deletion of approximately 114 kb, including the XG gene. Anti-Xga seems to be naturally occurring by detection in males who have never been transfused. MATERIALS AND METHODS: In this study, we identified 23 anti-Xga producers among 580,115 donors (0.004%). Additional 12 anti-Xga producers were also identified from a separate cohort. RESULTS: All 35 anti-Xga producers were male. Genomic DNA was obtained from 34 of 35 producers, and all 34 producers were confirmed to carry the XG-gene-deficient allele (XGdel). The breakpoints of all 34 producers were identical. The XGdel was also identified in 12 non-producers of anti-Xga among 860 donors who have no antibodies against RBCs, and the breakpoints were also identical with the anti-Xga producers. CONCLUSION: Our results will serve as the basis for a more complete understanding of Xg blood group polymorphisms.


Assuntos
Antígenos de Grupos Sanguíneos , Doadores de Sangue , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Feminino , Patrimônio Genético , Humanos , Japão , Masculino , Fenótipo
4.
Vox Sang ; 117(1): 94-98, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34227688

RESUMO

BACKGROUND AND OBJECTIVES: Antigens of the MNS blood group system are expressed on the red blood cell (RBC) membrane on glycophorin A (GPA) and glycophorin B (GPB) or on hybrid molecules of GPA and GPB. This study investigated the distribution of glycophorin variants and alloantibodies against Hil and MINY among Japanese individuals. METHODS: Mi(a+) or Hil+ RBCs were screened using an automated blood grouping machine (PK7300) with monoclonal anti-Mia or polyclonal anti-Hil. Glycophorin variants were defined by serology with monoclonal antibodies against Mia , Vw, MUT and Mur, and polyclonal antibodies against Hil, MINY and Hop + Nob (KIPP). The glycophorin variants were further confirmed by immunoblotting and Sanger sequencing. Alloanti-Hil and alloanti-MINY in the plasma were screened using GP.Hil RBCs in an antiglobulin test. The specificity of anti-Hil or anti-MINY was assessed using GP.Hil (Hil+MINY+) and GP.JL (Hil-MINY+) RBCs. RESULTS: The GP.HF, GP.Mur, GP.Hut, GP.Vw, GP.Kip and GP.Bun frequencies in 1 005 594 individuals were 0·0357%, 0·0256%, 0·0181%, 0·0017%, 0·0009% and 0·0007%, respectively. GP.Hil was found in as four of the 13 546 individuals (0·0295%). Of 137 370 donors, 10 had anti-Hil (0·0073%) and three had anti-MINY (0·0022%). CONCLUSIONS: Glycophorin variants were relatively rare in Japanese individuals, with the major variants being GP.HF (0·0357%), GP.Hil (0·0295%) and GP.Mur (0·0256%). Only one example of anti-MINY was previously reported, but we found three more in this study.


Assuntos
Glicoforinas , Isoanticorpos , Tipagem e Reações Cruzadas Sanguíneas , Humanos , Japão , Sistema do Grupo Sanguíneo MNSs
5.
Transfusion ; 61(10): 2825-2829, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34369596

RESUMO

BACKGROUND: In this study, we identified a novel glycophorin variant (GP.MOT) in a Mia -positive Japanese blood donor. The proband with this glycophorin variant was discovered by antigen screening of samples from 475,493 Japanese blood donors using monoclonal anti-Mia . STUDY DESIGN AND METHODS: Standard serological techniques and flow cytometry were performed. GP.MOT RBCs were examined by immunoblotting using anti-GPA, anti-MUT or anti-Mur. Genome DNA was extracted from whole blood, and the GYPA/GYPB was analyzed by polymerase chain reactions and Sanger sequencing. RESULTS: The MNS blood group of the proband was M + N + w S-s + with the presence of other low-frequency antigens including Mia , Mur, MUT, and KIPP. A 43-kDa molecule, which is almost equivalent in size to glycophorin A (GPA), was identified by immunoblotting using monoclonal anti-MUT and anti-Mur. Sanger sequencing clearly indicated that the proband had two different GYPA*M alleles at SNP rs62334651 (GYPA*M232 + 55A and GYPA*M232 + 55G), as well as a GYP(B-A) hybrid allele (GYP*MOT) with breakpoints located on pseudoexon 3 of GYPB from c.210 to c.219. DISCUSSION: We identified a hybrid glycophorin GP.MOT with the deduced unique amino acid sequence GPB (20-45)-GPΨB (46-70)-GPA (71-149), which has not been previously reported.


Assuntos
Glicoforinas/genética , Alelos , Sequência de Aminoácidos , Sequência de Bases , Doadores de Sangue , Variação Genética , Humanos , Japão , Sistema do Grupo Sanguíneo MNSs/genética , Análise de Sequência de DNA
6.
EJHaem ; 2(4): 750-756, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35757680

RESUMO

Serologic RhD-negative red cells can cause anti-D alloimmunization if they carry the Asian-type DEL or other DEL variants. RHD genotyping is a viable countermeasure if available, but inexpensive alternatives are worthy of consideration. RhD-negative blood donors in Japan were studied by anti-D adsorption-elution and RHD genotyping. We collated published case reports of RhD-negative red cell transfusions associated with inexplicable anti-D immunization. Of 2754 serologic RhD-negative donors, 378 were genotyped D/d. Anti-D adsorption-elution revealed 63.5% (240 of 378) to be DEL, of whom 96.7% (232 of 240) had the 1227G > A variant, diagnostic for the Asian-type DEL. All 240 donors also carried at least one C antigen; none had a cc phenotype. The chance of transfusing DEL red cells to genuinely RhD-negative Asian patients (based on a three-unit transfusion) ranges from 16.7% in Korea to 69.4% in Taiwan, versus 0.6% in Germany. Among 22 RhD-negative recipients of serologic RhD-negative red cells, who produced new or increased anti-D antibody titers, all 17 from East Asia were transfused with red cells with a C-positive phenotype or known to be Asian-type DEL or both. Serologic RhD-negative East Asians with a cc phenotype can be red cell donors for RhD-negative recipients, especially those of childbearing potential.

7.
Br J Haematol ; 191(5): 920-926, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32705675

RESUMO

The Kg-antigen was first discovered in an investigation of a mother whose infant had haemolytic disease of the newborn (HDN). The antibody against the Kg-antigen is believed to be responsible for HDN. The Kg-antigen is provisionally registered under the number 700045, according to the Red Cell Immunogenetics and Blood Group Terminology. However, the molecular nature of the Kg-antigen has remained a mystery for over 30 years. In this study, a monoclonal antibody against the Kg-antigen and the recombinant protein were developed that allowed for the immunoprecipitation analysis. Immunoprecipitants from the propositus' red blood cell ghosts were subjected to mass spectrometry analysis, and DNA sequence analysis of the genes was also performed. A candidate for the Kg-antigen was molecularly isolated and confirmed to be a determinant of the Kg-antigen by cell transfection and flow cytometry analyses. The Kg-antigen and the genetic mutation were then screened for in a Japanese population. The molecular nature of the Kg-antigen was shown to be RhAG with a Lys164Gln mutation. Kg phenotyping further clarified that 0.22% of the Japanese population studied was positive for the Kg-antigen. These findings provide important information on the Kg-antigen, which has been clinically presumed to give rise to HDN.


Assuntos
Eritroblastose Fetal/genética , Membrana Eritrocítica/genética , Isoantígenos/genética , Mutação de Sentido Incorreto , Sistema do Grupo Sanguíneo Rh-Hr/genética , Substituição de Aminoácidos , Eritroblastose Fetal/metabolismo , Membrana Eritrocítica/metabolismo , Feminino , Humanos , Recém-Nascido , Masculino , Sistema do Grupo Sanguíneo Rh-Hr/metabolismo
8.
Transfusion ; 60(6): 1287-1293, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32358867

RESUMO

BACKGROUND: MNS is one of the highly polymorphic blood groups comprising many antigens generated by genomic recombination among the GYPA, GYPB, and GYPE genes as well as by single-nucleotide changes. We report a patient with red blood cell (RBC) antibody against an unknown low-frequency antigen, tentatively named SUMI, and investigated its carrier molecule and causal gene. STUDY DESIGN AND METHODS: Standard serologic tests, including enzyme tests, were performed. Monoclonal anti-SUMI-producing cells (HIRO-305) were established by transformation and hybridization methods using lymphocytes from a donor having anti-SUMI. SUMI+ RBCs were examined by immunocomplex capture fluorescence analysis (ICFA) using HIRO-305 and murine monoclonal antibodies against RBC membrane proteins carrying blood group antigens. Genomic DNA was extracted from whole blood, and the GYPA gene was analyzed by polymerase chain reactions and Sanger sequencing. RESULTS: Serologic screening revealed that 23 of the 541,522 individuals (0.0042%) were SUMI+, whereas 1351 of the 10,392 individuals (13.0%) had alloanti-SUMI. SUMI antigen was sensitive to ficin, trypsin, pronase, and neuraminidase, but resistant to α-chymotrypsin and sulfydryl-reducing agents. ICFA revealed that the SUMI antigen was carried on glycophorin A (GPA). According to Sanger sequencing and cloning, the SUMI+ individuals had a GYPA*M allele with c.91A>C (p.Thr31Pro), which may abolish the O-glycan attachment site. CONCLUSIONS: The new low-frequency antigen SUMI is carried on GPA encoded by the GYPA*M allele with c.91A>C (p.Thr31Pro). Neuraminidase sensitivity suggests that glycophorin around Pro31 are involved in the SUMI determinant.


Assuntos
Eritrócitos/imunologia , Glicoforinas/genética , Sistema do Grupo Sanguíneo MNSs/genética , Mutação de Sentido Incorreto , Substituição de Aminoácidos , Feminino , Glicoforinas/imunologia , Humanos , Sistema do Grupo Sanguíneo MNSs/imunologia , Masculino
9.
Vox Sang ; 115(8): 756-766, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32394466

RESUMO

BACKGROUND: The low-incidence antigen Sta of the MNS system is usually associated with the GP(B-A) hybrid molecule, which carries the 'N' antigen at the N terminus. The GP(A-A) molecule with trypsin-resistant M antigen has been found in a few St(a+) individuals. MATERIALS AND METHODS: Among Japanese blood donors, we screened 24 292 individuals for the presence of St(a+) with trypsin-resistant 'N' antigen and 193 009 individuals for the presence of St(a+) with trypsin-resistant M antigen. The breakpoints responsible for the Sta antigen were analysed by sequencing the genomic DNAs. RESULTS: A total of 1001 (4·1%) individuals were identified as St(a+) with trypsin-resistant 'N' antigen. Out of 1001 individuals, 115 were selected randomly for sequencing. Two novel GYP*Sch (GYP*401) variants with new intron 3 breakpoints of GYPA were detected in three cases. Twenty-five (0·013%) individuals were identified as St(a+) with trypsin-resistant M antigen. Five individuals had the GYP(A-ψB-A) hybrid allele; two of these five individuals were GYP*Zan (GYP*101.01), and the remaining three had a novel GYP(A-ψB-A) allele with the first breakpoint in GYPA exon A3 between c.178 and c.203. Nine individuals had a novel GYP(A-E-A) allele with GYPE exon E2 and pseudoexon E3 instead of GYPA exon A2 and A3. The 11 remaining individuals had a novel GYP(A-A) allele with a 9-bp deletion that included the donor splice site of intron 3 of GYPA. CONCLUSION: Our finding on diversity of glycophorin genes responsible for Sta antigen provides evidence for further complexity in the MNS system.


Assuntos
Doadores de Sangue , Glicoforinas/genética , Mutação , Sítios de Splice de RNA , Alelos , Povo Asiático/genética , Éxons , Humanos , Japão , Sistema do Grupo Sanguíneo MNSs/genética
10.
Vox Sang ; 115(7): 579-585, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32314425

RESUMO

BACKGROUND AND OBJECTIVES: MNS is a highly polymorphic blood group comprising 49 antigens recognized by International Society of Blood Transfusion, some of which may have been generated by genomic recombination among the closely linked genes GYPA, GYPB and GYPE. The GYPE gene has an almost identical sequence to GYPA*01 allele in exon 2 (99% homology), which accounts for M antigen. We investigated an unusual glycophorin molecule with protease-resistant M antigen. METHODS: Blood samples were screened by an automated blood typing system (PK7300) using bromelain-treated red blood cells (RBCs) and murine monoclonal anti-M. The M-positive RBC samples were analysed by immunoblotting using anti-M as the primary antibody. GYPA, GYPB and GYPE genes were analysed by polymerase chain reaction (PCR), cloning and sequencing using reticulocyte mRNA and genomic DNA. RESULTS: Serological tests and immunoblotting revealed that 103 of the 193 009 individuals (0·0534%) expressed protease-resistant M-active glycophorin having a molecular weight of 20 kDa. All the 103 individuals were S+ s- or S- s+. When reticulocyte mRNA from the individuals with M-active glycophorin (20 kDa) was examined by PCR and cloning followed by sequencing, a novel GYPE-B hybrid transcript was identified. Long-range PCR and sequencing using genomic DNA revealed that the individuals had a GYPB-E(2-4)-B hybrid gene. This hybrid gene was predicted to encode a 59-amino-acid mature glycoprotein that expresses no S or s antigens CONCLUSIONS: The prevalence of the M-active glycophorin (20 kDa) in the Japanese population is 0·0534%. This glycophorin is predicted to be a 59 amino acids polypeptide encoded by the novel GYPB-E(2-4)-B hybrid gene.


Assuntos
Alelos , Glicoforinas/genética , Células Cultivadas , Glicoforinas/química , Glicoforinas/metabolismo , Humanos , Peptídeo Hidrolases/metabolismo , Polimorfismo Genético , Domínios Proteicos , Proteólise , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
12.
Transfusion ; 60(1): 184-196, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31840280

RESUMO

BACKGROUND: Loss of blood group ABO antigens on red blood cells (RBCs) is well known in patients with leukemias, and such decreased ABO expression has been reported to be strongly associated with hypermethylation of the ABO promoter. We investigated the underlying mechanism responsible for A-antigen reduction on RBCs in a patient with myelodysplastic syndrome. STUDY DESIGN AND METHODS: Genetic analysis of ABO was performed by PCR and sequencing using peripheral blood. RT-PCR were carried out using cDNA prepared from total bone marrow (BM) cells. Bisulfite genomic sequencing was performed using genomic DNA from BM cells. Screening of somatic mutations was carried out using a targeted sequencing panel with genomic DNA from BM cells, followed by transient transfection assays. RESULTS: Genetic analysis of ABO did not reveal any mutation in coding regions, splice sites, or regulatory regions. RT-PCR demonstrated reduction of A-transcripts when the patient's RBCs were not agglutinated by anti-A antibody and did not indicate any significant increase of alternative splicing products in the patient relative to the control. DNA methylation of the ABO promoter was not obvious in erythroid cells. Targeted sequencing identified somatic mutations in ASXL1, EZH2, RUNX1, and WT1. Experiments involving transient transfection into K562 cells showed that the expression of ABO was decreased by expression of the mutated RUNX1. CONCLUSION: Because the RUNX1 mutation encoded an abnormally elongated protein without a transactivation domain which could act as dominant negative inhibitor, this frame-shift mutation in RUNX1 may be a genetic candidate contributing to A-antigen loss on RBCs.


Assuntos
Sistema ABO de Grupos Sanguíneos/biossíntese , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Eritrócitos/metabolismo , Regulação da Expressão Gênica , Mutação , Síndromes Mielodisplásicas , Sistema ABO de Grupos Sanguíneos/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/biossíntese , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Feminino , Humanos , Células K562 , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/sangue , Síndromes Mielodisplásicas/genética , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Proteínas WT1/biossíntese , Proteínas WT1/genética
13.
Transfus Med Rev ; 33(3): 183-188, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31227265

RESUMO

The severity of the hemolytic disease of the fetus and newborn (HDFN) due to Jra mismatch ranges from no symptoms to severe anemia that requires intrauterine and exchange transfusions. We encountered a newborn, born to a healthy mother having anti-Jra at 38 weeks of pregnancy, who had moderate anemia, a positive direct antiglobulin test (DAT) result, no increased erythropoiesis, and no jaundice at birth. Flow cytometry revealed that the Jra antigen of red cells in the infant was nearly negative at birth, biphasic at 5 weeks, and lowly expressed at 7 months of life. We searched online for previous case reports on HDFN due to Jra incompatibility. Among 63 reported cases, excluding 25 cases, 38 were included with the present case for analysis. Of 39 newborns, 10 developed clear anemia (hemoglobin <10.0 g/dL), and 1 died, 5 developed hydrops fetalis, 4 needed intrauterine transfusion and/or exchange transfusion, and 3 received red cell transfusion after birth; overlaps were included. Among 29 neonates with no anemia, 8 needed interventions including phototherapy and γ-globulin infusion, and the remaining 21 received conservative supports only. The maternal anti-Jra titer, ranging between 4 and 2048, did not correlate with the severity of anemia, levels of bilirubin, or any interventions required. The DAT of red cells was positive in 29 of 36 fetuses/newborns tested, whereas it was often negative among anemic neonates (4 of 9) (P < .05). Hematopoiesis did not increase effectively, as indicated by reticulocyte ratios between 1.7% and 22.3%, even with the increase in reticulocytes in anemic neonates compared with nonanemic neonates (P < .05). Total bilirubin levels ranged broadly between 0.2 and 14.3 mg/dL but were generally low. The maternal anti-Jra titer and IgG3 subclass did not correlate with the morbidity of the newborns. Being identical/compatible between mothers and their infants may possibly enhance infants' morbidity, as a weak tendency was observed (P = .053). Maternal anti-Jra may suppress erythropoiesis in fetuses via a mechanism different from the established HDFN, such as anti-D, as evidenced by the lower reticulocyte count and small increase in bilirubin in neonates. As the anti-Jra titer, IgG subclass, and DAT were not correlated with the severity, the mechanism of anti-Jra-induced HDFN remains to be elucidated.


Assuntos
Incompatibilidade de Grupos Sanguíneos/diagnóstico , Eritroblastose Fetal/diagnóstico , Adulto , Incompatibilidade de Grupos Sanguíneos/sangue , Incompatibilidade de Grupos Sanguíneos/imunologia , Eritroblastose Fetal/sangue , Eritroblastose Fetal/imunologia , Eritropoese , Feminino , Hemólise , Humanos , Recém-Nascido , Masculino , Índice de Gravidade de Doença
14.
Transfusion ; 59(7): 2429-2435, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31020675

RESUMO

BACKGROUND: Anti-KANNO, a broadly reactive RBC alloantibody, is found among some Japanese pregnant women, but the genetic basis of the corresponding antigen remains unclear. STUDY DESIGN AND METHODS: We integrated a statistical approach to identify the coding gene for KANNO antigen by conducting a genome-wide association study (GWAS) on four KANNO-negative individuals and 415 healthy Japanese. We also applied whole-exome sequencing to them and performed a replication study to confirm the identified genome variation using independent 14 KANNO-negative individuals. A monoclonal antibody-specific immobilization of erythrocyte antigens (MAIEA) assay was used to locate KANNO antigen on RBC-specific membrane protein. In vivo and in vitro binding assays of anti-KANNO were further applied to the cells expressing a candidate protein. RESULTS: The GWAS revealed a genome-wide significant association of chromosome 20p13 locus (p = 2.76E-08; odds ratio > 1000 [95% confidence interval = 48-23,674]). The identified single-nucleotide polymorphism located in an intronic region of the prion protein (PRNP) gene. Whole-exome sequencing revealed a missense variant in the PRNP gene (rs1800014, E219K), which is in linkage disequilibrium with the single-nucleotide polymorphism identified in the GWAS. All 18 KANNO-negative individuals possessed the homozygous genotype of the missense variant. The MAIEA assay using anti-KANNO and mouse antihuman prion protein showed a clear difference between KANNO-positive and KANNO-negative RBCs. Anti-KANNO showed direct binding to CHO-K1 cells expressing wild-type PRNP but not to those expressing E219K PRNP. CONCLUSION: We first identified the coding gene of the high-frequency antigen KANNO located in PRNP and the missense variation (E219K) that affects the seropositivity of the KANNO antigen, which were confirmed by PRNP overexpressed cells.


Assuntos
Antígenos de Grupos Sanguíneos/genética , Cromossomos Humanos Par 20/genética , Frequência do Gene , Genoma Humano , Glicoproteínas/genética , Polimorfismo de Nucleotídeo Único , Proteínas Priônicas/genética , Estudo de Associação Genômica Ampla , Humanos
15.
Transfusion ; 59(8): 2519-2522, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30990901

RESUMO

BACKGROUND: The Rh complex contributes to cell membrane structural integrity of erythrocytes. Rhnull syndrome is characterized by the absence of the Rh antigen on the erythrocyte membrane, resulting in chronic hemolytic anemia. We recently came across 3 Rhnull phenotype probands within two families with the same novel RHAG mutation in the Japanese population. MATERIALS AND METHODS: Detailed Rh phenotyping by hemagglutination was performed using monoclonal and polyclonal anti-D, -C, -c, -E, and -e; monoclonal and polyclonal anti-Rh17 antibodies; and polyclonal anti-Rh29 antibodies. RHAG mRNA transcripts were analyzed by reverse transcription-polymerase chain reaction, and the mutation was verified by genomic sequencing. RESULTS: The genomic region spanning exon 6 contained a G > A transition in the invariant GT motif of the 5' donor splice-site of Intron 6 (c.945+1G>A). The Rhnull phenotype was caused by an autosomal recessive mutation in Probands 1 and 2, determined by family history. Regarding clinical features, the degree of hemolysis varied slightly between these individuals, with Proband 3 displaying acute hemolytic anemia with an infection. While no standard therapy has been established, the condition of the patient in this study improved with conservative treatment, including hydration and antibiotics. CONCLUSION: The mechanisms of hemolysis due to the Rhnull phenotype can vary, but our findings indicate that acute hemolytic crisis caused by the Rhnull syndrome could be associated with infection.


Assuntos
Proteínas Sanguíneas/genética , Glicoproteínas de Membrana/genética , Mutação , Povo Asiático , Tipagem e Reações Cruzadas Sanguíneas , Análise Mutacional de DNA , Hemólise/genética , Humanos , Japão , Masculino , Glicoproteínas de Membrana/sangue , Pessoa de Meia-Idade
16.
Transfusion ; 59(4): 1190-1195, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30784074

RESUMO

BACKGROUND: Weak D-type RBCs have fewer D epitopes, but it remains unclear whether individuals with certain types of weak D produce alloanti-D directed at D epitopes absent from the RBCs, and whether it is an alloantibody or an autoantibody. We report the first case of a patient with a weak D Type 15 who produced autoantibodies mimicking alloanti-D. CASE REPORT: A 52-year-old Japanese male with weak D developed anti-D 3 months after transfusion of D-negative and -positive RBCs, and the antibody persisted for 24 months with a consistently negative direct antiglobulin test. Eluates from the patient's RBCs demonstrated anti-D specificity. The recipient did not exhibit any signs of delayed hemolytic transfusion reaction. As his anti-D was removed by the different adsorbing cells of weak D Type 15 and autologous as well as D positive, D negative, weak D Type 24, and partial DVa, it was thought to be an autoantibody mimicking anti-D rather than an alloantibody. The patient's RBCs reacted weakly with the 13 anti-D reagents used in the study. Polymerase chain reaction and nucleotide sequencing revealed that the patient had an RHD genotype of RHD*01N.01/RHD*15. CONCLUSION: Anti-D, produced in a patient with weak D Type 15 after transfusion, was found to be mimicking autoanti-D. Alloanti-D was excluded by an adsorption study with different RBC types.


Assuntos
Transfusão de Sangue , Eritrócitos/metabolismo , Isoanticorpos/sangue , Imunoglobulina rho(D)/sangue , Reação Transfusional/sangue , Humanos , Masculino , Pessoa de Meia-Idade
18.
Vox Sang ; 114(2): 171-173, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30523644

RESUMO

We found an individual with weakened S antigen expression on red blood cells (RBCs) during routine blood grouping. The proband was typed S+s+ by polyclonal antibodies, but the RBCs demonstrated different reactivity with three monoclonal anti-S. The proband did not have alloanti-S. Cloning and Sanger sequencing revealed that the proband had a c.166A>T (p.Thr56Ser) mutation in exon 4 of GYPB*S. When antibody screening of 60 455 blood donors was performed using the proband RBCs, no antibodies were detected. GYPB*S with c.166T should encode an unusual S antigen but the creation of a novel antigen remains to be investigated.


Assuntos
Antígenos de Grupos Sanguíneos/genética , Glicoforinas/genética , Mutação , Doadores de Sangue , Antígenos de Grupos Sanguíneos/metabolismo , Eritrócitos/metabolismo , Frequência do Gene , Glicoforinas/metabolismo , Humanos
19.
Transfusion ; 58(11): 2675-2682, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30180269

RESUMO

BACKGROUND: Antibody screening in pretransfusion tests is necessary to avoid critical complications of blood transfusion. Although red blood cells (RBCs) expressing relevant alloantigen(s) have been used for serologic antibody screening, little attention has been given to the use of cell lines, in which blood group antigen gene(s) are transduced, as reagent RBCs for antibody screening. STUDY DESIGN AND METHODS: The use of an erythroid progenitor cell line for serologic tests was studied. The expression of blood group antigens of erythroid progenitor cells was analyzed by genotyping and flow cytometry. Serologic analysis including hemagglutination was performed using erythroid progenitor cells to evaluate their sensitivity for antibody detection. Overexpression of exogenous erythroid antigen by lentiviral transduction was carried out and investigated for antibody detection sensitivity. RESULTS: Erythroid progenitor cells contained a substantial amount of hemoglobin and expressed sufficient levels of blood group antigens to detect corresponding monoclonal antibodies. Furthermore, the cell line could acquire an exogenous RBC antigen after lentiviral transduction and detected corresponding monoclonal and alloantibodies with equal sensitivity to antigen-positive RBCs. CONCLUSION: Application of erythroid progenitor cell lines for screening for unexpected antibodies could be helpful in solving issues such as reagent availability associated with the conventional RBC-based assay. The genetic expandability of erythroid progenitor cell lines by gene modification techniques could lead to the development of more convenient reagent RBCs.


Assuntos
Eritrócitos/imunologia , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/imunologia , Isoanticorpos/imunologia , Proteína 1 de Troca de Ânion do Eritrócito/genética , Linhagem Celular , Citometria de Fluxo , Humanos , Células K562
20.
Transfusion ; 57(4): 1072-1077, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28194794

RESUMO

BACKGROUND: In(Lu) is characterized by a reduced expression of antigens in the Lutheran blood group system as well as other blood group antigens. Mutations of the erythroid transcription factor, KLF1, have been reported to cause the In(Lu) phenotype, and we investigated Japanese In(Lu) to estimate the prevalence of the phenotype and KLF1 polymorphism. STUDY DESIGN AND METHODS: Blood samples were screened by monoclonal anti-CD44 and the In(Lu) phenotype was confirmed by tube tests including adsorption and elution tests using anti-Lua and anti-Lub . KLF1, LU, and A4GALT genes were analyzed by polymerase chain reaction and sequencing. RESULTS: We identified 100 of 481,322 blood donors (0.02%), and the previously characterized 20 donors, who had the In(Lu) phenotype with the LUB/LUB genotype. A total of 100 of the 120 In(Lu) individuals had mutant KLF1 alleles, and we identified 13 known and 21 novel alleles. The mutant KLF1 alleles with c.947G>A (p.Cys316Tyr), c.862A>G (p.Lys288Glu), or c.968C>G (p.Ser323Trp) were major in the In(Lu) individuals. The P1 antigen of 29 In(Lu) (two P1 /P1 , 27 P1 /P2 ) showed significantly weakened expression by hemagglutination. CONCLUSIONS: The prevalence of the In(Lu) phenotype in the Japanese population was 0.02%, and we identified 13 known and 21 novel KLF1 alleles. The KLF1 mutations cause the reduced expression of the P1 antigen.


Assuntos
Moléculas de Adesão Celular/genética , Fatores de Transcrição Kruppel-Like/genética , Sistema do Grupo Sanguíneo Lutheran/genética , Mutação de Sentido Incorreto , Fenótipo , Substituição de Aminoácidos , Povo Asiático , Moléculas de Adesão Celular/sangue , Feminino , Galactosiltransferases/biossíntese , Galactosiltransferases/genética , Globosídeos/biossíntese , Globosídeos/metabolismo , Humanos , Japão , Fatores de Transcrição Kruppel-Like/sangue , Sistema do Grupo Sanguíneo Lutheran/sangue , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA