Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 257
Filtrar
1.
Immunohematology ; 40(1): 1-9, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38739025

RESUMO

KLF transcription factor 1 (KLF1) and GATA binding protein 1 (GATA1) are transcription factors (TFs) that initiate and regulate transcription of the genes involved in erythropoiesis. These TFs possess DNA-binding domains that recognize specific nucleotide sequences in genes, to which they bind and regulate transcription. Variants in the genes that encode either KLF1 or GATA1 can result in a range of hematologic phenotypes-from benign to severe forms of thrombocytopenia and anemia; they can also weaken the expression of blood group antigens. The Lutheran (LU) blood group system is susceptible to TF gene variations, particularly KLF1 variants. Individuals heterozygous for KLF1 gene variants show reduced Lutheran antigens on red blood cells that are not usually detected by routine hemagglutination methods. This reduced antigen expression is referred to as the In(Lu) phenotype. For accurate blood typing, it is important to distinguish between the In(Lu) phenotype, which has very weak antigen expression, and the true Lunull phenotype, which has no antigen expression. The International Society of Blood Transfusion blood group allele database registers KLF1 and GATA1 variants associated with modified Lutheran expression. Here, we review KLF1 and recent novel gene variants defined through investigating blood group phenotype and genotype discrepancies or, for one report, investigating cases with unexplained chronic anemia. In addition, we include a review of the GATA1 TF, including a case report describing the second GATA1 variant associated with a serologic Lu(a-b-) phenotype. Finally, we review both past and recent reports on variations in the DNA sequence motifs on the blood group genes that disrupt the binding of the GATA1 TF and either remove or reduce erythroid antigen expression. This review highlights the diversity and complexity of the transcription process itself and the need to consider these factors as an added component for accurate blood group phenotyping.


Assuntos
Antígenos de Grupos Sanguíneos , Eritrócitos , Fator de Transcrição GATA1 , Fatores de Transcrição Kruppel-Like , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fator de Transcrição GATA1/genética , Eritrócitos/metabolismo , Eritrócitos/imunologia , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/imunologia , Sistema do Grupo Sanguíneo Lutheran/genética , Regulação da Expressão Gênica , Eritropoese/genética
2.
BMC Genomics ; 25(1): 230, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38429690

RESUMO

BACKGROUND: Krüppel-like factor 1 (KLF1), a crucial erythroid transcription factor, plays a significant role in various erythroid changes and haemolytic diseases. The rare erythrocyte Lutheran inhibitor (In(Lu)) blood group phenotype serves as an effective model for identifying KLF1 hypomorphic and loss-of-function variants. In this study, we aimed to analyse the genetic background of the In(Lu) phenotype in a population-based sample group by high-throughput technologies to find potentially clinically significant KLF1 variants. RESULTS: We included 62 samples with In(Lu) phenotype, screened from over 300,000 Chinese blood donors. Among them, 36 samples were sequenced using targeted Next Generation Sequencing (NGS), whereas 19 samples were sequenced using High Fidelity (HiFi) technology. In addition, seven samples were simply sequenced using Sanger sequencing. A total of 29 hypomorphic or loss-of-function variants of KLF1 were identified, 21 of which were newly discovered. All new variants discovered by targeted NGS or HiFi sequencing were validated through Sanger sequencing, and the obtained results were found to be consistent. The KLF1 haplotypes of all new variants were further confirmed using clone sequencing or HiFi sequencing. The lack of functional KLF1 variants detected in the four samples indicates the presence of additional regulatory mechanisms. In addition, some samples exhibited BCAM polymorphisms, which encodes antigens of the Lutheran (LU) blood group system. However, no BCAM mutations which leads to the absence of LU proteins were detected. CONCLUSIONS: High-throughput sequencing methods, particularly HiFi sequencing, were introduced for the first time into genetic analysis of the In(Lu) phenotype. Targeted NGS and HiFi sequencing demonstrated the accuracy of the results, providing additional advantages such as simultaneous analysis of other blood group genes and clarification of haplotypes. Using the In(Lu) phenotype, a powerful model for identifying hypomorphic or loss-of-function KLF1 variants, numerous novel variants have been detected, which have contributed to the comprehensive understanding of KLF1. These clinically significant KLF1 mutations can serve as a valuable reference for the diagnosis of related blood cell diseases.


Assuntos
Antígenos de Grupos Sanguíneos , Fatores de Transcrição Kruppel-Like , Antígenos de Grupos Sanguíneos/genética , Sequenciamento de Nucleotídeos em Larga Escala , Sistema do Grupo Sanguíneo Lutheran/genética , Mutação , Humanos
3.
STAR Protoc ; 4(3): 102503, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37669162

RESUMO

BCAM-positive basal limbal epithelial cells are an early transit-amplifying cell population (TAC) capable of holoclone formation and corneal epithelial differentiation. Here, we present a protocol for isolating BCAM-positive cells from human donor corneas by flow cytometry and cell sorting. We describe steps for cell dissection and dissociation, antibody staining, and flow cytometry. We then detail procedures for culturing the purified BCAM-positive and BCAM-negative cells for holoclone and cell sheet formation assays to study the factors that regulate corneal regeneration. For complete details on the use and execution of this protocol, please refer to Sasamoto et al.1.


Assuntos
Epitélio Corneano , Limbo da Córnea , Humanos , Citometria de Fluxo , Córnea , Células-Tronco , Sistema do Grupo Sanguíneo Lutheran , Moléculas de Adesão Celular
4.
Transfusion ; 63(4): 798-807, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36738255

RESUMO

BACKGROUND: The basal cell adhesion molecule (BCAM) carries the antigens of the Lutheran (LU, ISBT005) system. We report a novel Lutheran antigen and propose an updated, full-length 3D model of BCAM. STUDY DESIGN AND METHODS: Red blood cell testing, antibody identification, and BCAM genomic DNA sequencing were done by standard methods. Multi-template homology modeling of BCAM used structural templates selected for coverage, highest sequence identity, and protein domain family. All variants causing the loss or gain of a Lutheran antigen were analyzed for residue accessibility and intraprotein interactions. RESULTS: An antibody to a high-prevalence antigen in the plasma of a pregnant woman was determined to be directed at a novel Lutheran antigen. Sequencing of BCAM found three homozygous changes: c.212G > A (p.Arg71His) and two silent, c.711C > T and c.714C > T. The model was built from the first two immunoglobulin crystallized domains of BCAM (D1, D2), three other templates (for D3, D4 and D5 with a higher sequence identity with the target than those used for the model proposed by Burton and Brady in 2008, and for the transmembrane region) and RaptorX (for the intracellular domain). All residues associated with a Lutheran antigen were found to be exposed in wild-type or variant proteins, except p.447 associated with loss of Lu13 expression. CONCLUSION: The c.212G > A change results in the loss of LUGA (LU24) antigen. Whole genome sequencing continues to reveal polymorphisms with uncertain immunogenicity. This model and demonstration that nearly all residues associated with the expression of a Lutheran antigen are exposed will help evaluate the significance of new polymorphisms.


Assuntos
Moléculas de Adesão Celular , Protestantismo , Humanos , Moléculas de Adesão Celular/genética , Prevalência , Eritrócitos/metabolismo , Sistema do Grupo Sanguíneo Lutheran/genética
5.
Hypertension ; 79(12): 2830-2842, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36252138

RESUMO

BACKGROUND: Preeclampsia is a pregnancy syndrome that may utilize multiple pathogenic mechanisms. Insufficient trophoblast invasion and impaired uterine spiral artery remodeling are believed to be the pathological basis; yet the underlying mechanisms remain largely unclear. METHODS: The placental BCAM (basal cell adhesion molecule) expression and important clinical indicators were detected and correlation analysis was performed. MiRNAs directly targeting BCAM were predicted and further verified by dual-luciferase reporter gene, and the downstream molecular mechanisms of BCAM were investigated in both HTR-8/SVneo and JAR cells. In addition, pregnant/nonpregnant rats were treated with adenoviruses containing BCAM shRNA genes (Ad-shBCAM) on gestational 9.5 days to detect the preeclamptic features. RESULTS: The BCAM is highly expressed on the trophoblast membrane and decreased in the preeclamptic placentae. In HTR-8/SVneo and JAR cells, BCAM knockdown inhibited trophoblast proliferation, migration, and invasion, and suppressed phosphorylation on Y705 of STAT3 dependent on the downregulation of PIK3R6. Moreover, miR-199a-5p mediated the degradation of BCAM and also inhibited trophoblast proliferation, migration, and invasion. In vivo, BCAM deficiency induced a preeclampsia-like phenotype included elevated systolic blood pressure, proteinuria, impaired morphology and function of multiple organs (placenta, liver, and kidney), and fetal growth restriction. The expression of placenta BCAM/PIK3R6/p-STAT3 signaling was also downregulated in this preeclampsia rat model. CONCLUSIONS: MiR-199a-5p mediated-BCAM deficiency contributes to the suppression of trophoblast proliferation, migration, and invasion by inhibiting PIK3R6/p-STAT3 signaling, which may lead to poor placentation and result in preeclampsia-like phenotypes. Our study provides a new academic perspective on the pathogenesis of preeclampsia.


Assuntos
MicroRNAs , Pré-Eclâmpsia , Animais , Feminino , Gravidez , Ratos , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Movimento Celular , Sistema do Grupo Sanguíneo Lutheran/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Placenta/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Trofoblastos/metabolismo
6.
Clin Epigenetics ; 14(1): 99, 2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35941663

RESUMO

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is the most common and highly heterogeneous subtype of renal cell carcinoma. Dysregulated basal cell adhesion molecule (BCAM) gene is associated with poor prognosis in various cancers. However, the dysregulated functions and related multi-omics features of BCAM in ccRCC stay unclear. RESULTS: BCAM expression was aberrantly downregulated in ccRCC and correlated with adverse pathological parameters and poor prognosis. Low mRNA expression of BCAM was remarkably associated with its CpG methylation levels and BAP1 mutation status. Patients with lower-expressed BCAM concomitant with BAP1 mutation had a worse prognosis. Using RNA-seq data from The cancer genome atlas, we found that compared to the BCAM-high expression subgroup, ccRCC patients in the BCAM-low expression subgroup had significantly higher levels of immune infiltration, higher immune checkpoint expression levels and lower TIDE (tumor immune dysfunction and exclusion) score, indicating potential better response to immunotherapy. Data from the Clinical Proteomic Tumor Analysis Consortium further validated the association between low BCAM expression and CD8 + inflamed phenotype at protein level. Meanwhile, our results suggested that the angiogenesis-related pathways were enriched in the BCAM-high expression subgroup. More importantly, according to the data from the GDSC database, we revealed that the BCAM-high expression subgroup should be more sensitive to anti-angiogenetic therapies, including sorafenib, pazopanib and axitinib. CONCLUSIONS: These results suggest that BCAM could serve as a biomarker distinguishing different tumor microenvironment phenotypes, predicting prognosis and helping therapeutic decision-making for patients with ccRCC.


Assuntos
Carcinoma de Células Renais , Carcinoma , Neoplasias Renais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma/genética , Carcinoma de Células Renais/metabolismo , Moléculas de Adesão Celular/genética , Metilação de DNA , Epigênese Genética , Humanos , Neoplasias Renais/patologia , Sistema do Grupo Sanguíneo Lutheran/metabolismo , Prognóstico , Proteômica , Microambiente Tumoral/genética
7.
Nutr Metab Cardiovasc Dis ; 32(7): 1753-1765, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35599089

RESUMO

BACKGROUND AND AIMS: Observational studies reveal that different body fat measures are associated with cardiometabolic disease with different effects. However, causality is not reflected by such observations. To explore and compare the causal relationships of general obesity (measured by body mass index (BMI)), adipose obesity (measured by fat mass percentage (FMP)) and central obesity (measured by waist-to-height ratio (WHtR)) with cardiometabolic traits among children. METHODS AND RESULTS: We conducted one sample Mendelian randomization (MR) analysis in 3266 children from Beijing Children and Adolescents Metabolic Syndrome Study. Genetic instruments based on 28 SNPs were performed to explore and compare the causal associations of genetically BMI, FMP and WHtR with cardiometabolic traits. The genetic instruments were robustly correlated with observed BMI, FMP and WHtR. Each genetically 1-SD increment in BMI, FMP and WHtR were causally associated with increment in systolic blood pressure (SBP), diastolic blood pressure (DBP), log-transformed fasting plasma glucose (FPG), log-transformed HOMA-ß, and decrease in log-transformed high-density lipoprotein cholesterol (HDL), respectively (all P < 0.05 after Bonferroni correction). The receiver operating characteristic curve indicated that BMI and FMP showed stronger effects on SBP, DBP, HOMA-ß and HDL than WHtR (all P < 0.05). We also observed causal associations of BMI and FMP with log-transformed fasting insulin and HOMA-IR. CONCLUSIONS: The MR analysis based on population-based cohort indicated a causal relationship of adiposity and body fat distribution with cardiometabolic traits. When compared with central obesity, general obesity and adipose obesity might own stronger effects on blood pressure and blood lipids among children.


Assuntos
Doenças Cardiovasculares , Obesidade Abdominal , Adiposidade/genética , Adolescente , Índice de Massa Corporal , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/genética , Moléculas de Adesão Celular , Criança , Humanos , Sistema do Grupo Sanguíneo Lutheran , Análise da Randomização Mendeliana , Obesidade/diagnóstico , Obesidade/epidemiologia , Obesidade/genética , Obesidade Abdominal/complicações , Fatores de Risco , Circunferência da Cintura
8.
Blood Adv ; 6(21): 5798-5810, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-35349634

RESUMO

Severe malarial anemia (SMA) is the main cause of malaria-associated infant mortality in malaria endemic countries. One major factor that contributes to SMA is the accumulation of uninfected red blood cells (uRBCs) in the spleen. We report the activation of adhesion molecules Lutheran/basal cell adhesion molecule (Lu/BCAM) and CD44 on uRBCs from Plasmodium falciparum in vitro cultures and patients with malaria that mediates adherence to the splenic extracellular matrix (ECM) components laminin-α5 and hyaluronic acid (HA), respectively. This tight ECM-adhesion molecule interaction was associated with elevated intracellular Ca2+ levels, increased shedding of microvesicles, and Lu/BCAM clustering on altered uRBCs. Moreover, we observed that a soluble parasite-derived factor promoted the adhesive phenotype of uRBCs, as the incubation of RBCs with filtered malaria-conditioned medium reproduced the same adhesive effect in malaria culture-derived uRBCs. Eventually, Lu/BCAM and CD44 activation facilitate the adherence to ECM components of the red pulp, resulting in the enhanced splenic retention of uRBCs. Our results suggest a novel adhesion molecule-dependent mechanism that augments malaria-induced anemia.


Assuntos
Anemia Falciforme , Malária , Humanos , Sistema do Grupo Sanguíneo Lutheran/metabolismo , Moléculas de Adesão Celular/genética , Eritrócitos/metabolismo
9.
Development ; 149(1)2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-35020896

RESUMO

In early placental development, progenitor cytotrophoblasts (CTB) differentiate along one of two cellular trajectories: the villous or extravillous pathways. CTB committed to the villous pathway fuse with neighboring CTB to form the outer multinucleated syncytiotrophoblast (SCT), whereas CTB committed to the extravillous pathway differentiate into invasive extravillous trophoblasts (EVT). Unfortunately, little is known about the processes controlling human CTB progenitor maintenance and differentiation. To address this, we established a single cell RNA sequencing (scRNA-seq) dataset from first trimester placentas to identify cell states important in trophoblast progenitor establishment, renewal and differentiation. Multiple distinct trophoblast states were identified, representing progenitor CTB, column CTB, SCT precursors and EVT. Lineage trajectory analysis identified a progenitor origin that was reproduced in human trophoblast stem cell organoids. Heightened expression of basal cell adhesion molecule (BCAM) defined this primitive state, where BCAM enrichment or gene silencing resulted in enhanced or diminished organoid growth, respectively. Together, this work describes at high-resolution trophoblast heterogeneity within the first trimester, resolves gene networks within human CTB progenitors and identifies BCAM as a primitive progenitor marker and possible regulator.


Assuntos
Moléculas de Adesão Celular/metabolismo , Linhagem da Célula , Sistema do Grupo Sanguíneo Lutheran/metabolismo , Trofoblastos/metabolismo , Adulto , Moléculas de Adesão Celular/genética , Diferenciação Celular , Células Cultivadas , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Sistema do Grupo Sanguíneo Lutheran/genética , Organoides/citologia , Organoides/metabolismo , Trofoblastos/citologia
10.
Sci Rep ; 11(1): 18557, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34535703

RESUMO

Beta-hemoglobinopathies become prominent after birth due to a switch from γ-globin to the mutated ß-globin. Haploinsufficiency for the erythroid specific indispensable transcription factor Krueppel-like factor 1 (KLF1) is associated with high persistence of fetal hemoglobin (HPFH). The In(Lu) phenotype, characterized by low to undetectable Lutheran blood group expression is caused by mutations within KLF1 gene. Here we screened a blood donor cohort of 55 Lutheran weak or negative donors for KLF1 variants and evaluated their effect on KLF1 target gene expression. To discriminate between weak and negative Lutheran expression, a flow cytometry (FCM) assay was developed to detect Lu antigen expression. The Lu(a-b-) (negative) donor group, showing a significant decreased CD44 (Indian blood group) expression, also showed increased HbF and HbA2 levels, with one individual expressing HbF as high as 5%. KLF1 exons and promoter sequencing revealed variants in 80% of the Lutheran negative donors. Thirteen different variants plus one high frequency SNP (c.304 T > C) were identified of which 6 were novel. In primary erythroblasts, knockdown of endogenous KLF1 resulted in decreased CD44, Lu and increased HbF expression, while KLF1 over-expressing cells were comparable to wild type (WT). In line with the pleiotropic effects of KLF1 during erythropoiesis, distinct KLF1 mutants expressed in erythroblasts display different abilities to rescue CD44 and Lu expression and/or to affect fetal (HbF) or adult (HbA) hemoglobin expression. With this study we identified novel KLF1 variants to be include into blood group typing analysis. In addition, we provide further insights into the regulation of genes by KLF1.


Assuntos
Moléculas de Adesão Celular/genética , Hemoglobina Fetal/análise , Fatores de Transcrição Kruppel-Like/genética , Sistema do Grupo Sanguíneo Lutheran/genética , gama-Globinas/análise , Células Cultivadas , Células Eritroides/citologia , Células Eritroides/metabolismo , Eritropoese , Hemoglobina Fetal/genética , Humanos , Mutação , Polimorfismo de Nucleotídeo Único , gama-Globinas/genética
14.
J Neurogenet ; 35(2): 59-66, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33970751

RESUMO

Variants in APOE are associated with risk of late onset Alzheimer's disease (LOAD) but the magnitude of the effect has been reported to vary across ancestries. Also, other variants in the region have been reported to show association though it has been unclear whether this was secondary to their linkage disequilibrium with the APOE variants rs429358 and rs7412. Previous analyses of exome-sequenced samples have identified other genes in which rare variants impact risk of disease. In this study 2000 whole genome sequenced cases and controls with different ancestries were subjected to gene-based weighted burden analysis to identify risk genes. Additionally, individual variants in the APOE region were tested for association with LOAD. When using the APOE variants as covariates no individual genes showed statistically significant evidence for association after Bonferroni correction for multiple testing, which may well be a consequence of the modest sample size. Likewise, for those variants initially showing evidence of association with LOAD incorporating the APOE variants as covariates dramatically reduced the strength of association. These results demonstrate that the differential association of APOE across ancestries does not appear to be driven by another variant in the region. It seems likely that no other genes in the region have a direct effect on LOAD risk.


Assuntos
Doença de Alzheimer/genética , Apolipoproteínas E/genética , Desequilíbrio de Ligação/genética , Idoso , Idoso de 80 Anos ou mais , Alelos , Apolipoproteína C-I/genética , Moléculas de Adesão Celular/genética , Feminino , Humanos , Sistema do Grupo Sanguíneo Lutheran/genética , Masculino , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial/genética , Nectinas/genética
15.
Haematologica ; 106(9): 2478-2488, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32855277

RESUMO

Vaso-occlusive crises are the hallmark of sickle cell disease (SCD). They are believed to occur in two steps, starting with adhesion of deformable low-dense red blood cells (RBCs), or other blood cells such as neutrophils, to the wall of post-capillary venules, followed by trapping of the denser RBCs or leukocytes in the areas of adhesion because of reduced effective lumen-diameter. In SCD, RBCs are heterogeneous in terms of density, shape, deformability and surface proteins, which accounts for the differences observed in their adhesion and resistance to shear stress. Sickle RBCs exhibit abnormal adhesion to laminin mediated by Lu/BCAM protein at their surface. This adhesion is triggered by Lu/BCAM phosphorylation in reticulocytes but such phosphorylation does not occur in mature dense RBCs despite firm adhesion to laminin. In this study, we investigated the adhesive properties of sickle RBC subpopulations and addressed the molecular mechanism responsible for the increased adhesion of dense RBCs to laminin in the absence of Lu/BCAM phosphorylation. We provide evidence for the implication of oxidative stress in post-translational modifications of Lu/BCAM that impact its distribution and cis-interaction with glycophorin C at the cell surface activating its adhesive function in sickle dense RBCs.


Assuntos
Anemia Falciforme , Laminina , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Eritrócitos/metabolismo , Humanos , Laminina/metabolismo , Sistema do Grupo Sanguíneo Lutheran/metabolismo , Estresse Oxidativo
16.
Blood Adv ; 4(24): 6218-6229, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33351118

RESUMO

Senescence of erythrocytes is characterized by a series of changes that precede their removal from the circulation, including loss of red cell hydration, membrane shedding, loss of deformability, phosphatidyl serine exposure, reduced membrane sialic acid content, and adhesion molecule activation. Little is known about the mechanisms that initiate these changes nor is it known whether they are interrelated. In this study, we show that Ca2+-dependent K+ efflux (the Gardos effect) drives erythrocyte senescence. We found that increased intracellular Ca2+ activates the Gardos channel, leading to shedding of glycophorin-C (GPC)-containing vesicles. This results in a loss of erythrocyte deformability but also in a marked loss of membrane sialic acid content. We found that GPC-derived sialic acid residues suppress activity of both Lutheran/basal cell adhesion molecule (Lu/BCAM) and CD44 by the formation of a complex on the erythrocyte membrane, and Gardos channel-mediated shedding of GPC results in Lu/BCAM and CD44 activation. This phenomenon was observed as erythrocytes aged and on erythrocytes that were otherwise prone to clearance from the circulation, such as sickle erythrocytes, erythrocytes stored for transfusion, or artificially dehydrated erythrocytes. These novel findings provide a unifying concept on erythrocyte senescence in health and disease through initiation of the Gardos effect.


Assuntos
Sistema do Grupo Sanguíneo Lutheran , Protestantismo , Adesão Celular , Moléculas de Adesão Celular , Eritrócitos
17.
Mol Oncol ; 14(4): 829-845, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31951095

RESUMO

Patients with metastatic gastric cancer (GC) have a poor prognosis; however, the molecular mechanism of GC metastasis remains unclear. Here, we employed bioinformatics to systematically screen the metastasis-associated genes and found that the levels of basal cell adhesion molecule (BCAM) were significantly increased in GC tissues from patients with metastasis, as compared to those without metastasis. The upregulation of BCAM was also significantly associated with a shorter survival time. Depletion of BCAM inhibited GC cell migration and invasion. Knockout (KO) of BCAM by the CRISPR/Cas9 system reduced the invasion and metastasis of GC cells. To explore the mechanism of BCAM upregulation, we identified a previously uncharacterized BCAM sense lncRNA that spanned from exon 6 to intron 6 of BCAM, and named it as BCAM-associated long noncoding RNA (BAN). Knockdown of BAN inhibited BCAM expression at both mRNA and protein levels. Knockdown of BAN suppressed GC cell migration and invasion, which was effectively rescued by ectopic expression of BCAM. Further clinical data showed that BAN upregulation was associated with GC metastasis and poor prognosis. Importantly, BAN expression was also significantly associated with that of BCAM in GC tissues. Taken together, these results indicate that increased expression of BCAM and its sense lncRNA BAN promote GC cell invasion and metastasis, and are associated with poor prognosis of GC patients.


Assuntos
Moléculas de Adesão Celular/genética , Sistema do Grupo Sanguíneo Lutheran/genética , Invasividade Neoplásica/genética , RNA Longo não Codificante/genética , Neoplasias Gástricas/genética , Regulação para Cima , Linhagem Celular Tumoral , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica/diagnóstico , Prognóstico , Neoplasias Gástricas/diagnóstico
18.
J Biol Chem ; 294(41): 14911-14921, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31413112

RESUMO

Tumor cell migration depends on the interactions of adhesion proteins with the extracellular matrix. Lutheran/basal cell adhesion molecule (Lu/BCAM) promotes tumor cell migration by binding to laminin α5 chain, a subunit of laminins 511 and 521. Lu/BCAM is a type I transmembrane protein with a cytoplasmic domain of 59 (Lu) or 19 (Lu(v13)) amino acids. Here, using an array of techniques, including site-directed mutagenesis, immunoblotting, FRET, and proximity-ligation assays, we show that both Lu and Lu(v13) form homodimers at the cell surface of epithelial cancer cells. We mapped two small-XXX-small motifs in the transmembrane domain as potential sites for monomers docking and identified three cysteines in the cytoplasmic domain as being critical for covalently stabilizing dimers. We further found that Lu dimerization and phosphorylation of its cytoplasmic domain were concomitantly needed to promote cell migration. We conclude that Lu is the critical isoform supporting tumor cell migration on laminin 521 and that the Lu:Lu(v13) ratio at the cell surface may control the balance between cellular firm adhesion and migration.


Assuntos
Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Laminina/farmacologia , Sistema do Grupo Sanguíneo Lutheran/química , Sistema do Grupo Sanguíneo Lutheran/metabolismo , Multimerização Proteica/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Células CACO-2 , Cães , Humanos , Células Madin Darby de Rim Canino , Modelos Moleculares , Fosforilação/efeitos dos fármacos , Domínios Proteicos , Estrutura Quaternária de Proteína
19.
Transfus Med Rev ; 33(2): 111-117, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31023581

RESUMO

Erythroid-specific Krüppel-like factor 1, or KLF1, is an integral transcriptional activator for erythropoiesis. Genetic variants within KLF1 can result in a range of erythropoietic clinical phenotypes from benign to significant. The In(Lu) phenotype refers to changes in the quantitative expression of blood group-associated red cell surface molecules due to KLF1 variants which are otherwise clinically benign. These clinically benign KLF1 variants are associated with a reduced expression of 1 or more red cell membrane proteins/carbohydrates that carry blood group antigens for the LU (Lutheran), IN (Indian), P1PK, LW (Landsteiner-Wiener), KN (Knops), OK, RAPH, and I blood group systems. This is of significance during routine serologic blood typing when expression falls below the test sensitivity and therefore impacts on the ability to accurately detect the presence of affected blood group antigens. This is of clinical importance because the transfusion requirements for individuals with the In(Lu) phenotype differ from those of individuals that have a true Lunull phenotype. With this review, we summarize the current body of knowledge with regard to the In(Lu) phenotype and associated KLF1 variants. Our review also highlights discordant reports and provides insights for future research and management strategies. Serological heterogeneity in blood group expression of In(Lu) individuals has been shown, but studies are limited by the low prevalence of the phenotype and therefore the small numbers of samples. They are further limited by availability and inconsistent application of serological reagents and varying test algorithms. With the advent of genome sequence-based testing, an increasing list of In(Lu)-associated KLF1 variants is being revealed. The spectrum of effects on blood group expression due to these variants warrants further attention, and a consistent methodological approach of studies in larger cohorts is required. We propose that a recently reported testing framework of standardized serological studies, flow cytometry, and variant analysis be adopted; and that the international databases be curated to document KLF1 variability and the resultant In(Lu) red cell blood group expression. This will provide better classification of KLF1 variants affecting blood group expression and allow for phenotype prediction from genotype, accurate typing of In(Lu) individuals, and better transfusion management of related challenging transfusion scenarios.


Assuntos
Antígenos de Grupos Sanguíneos/genética , Fatores de Transcrição Kruppel-Like/genética , Sistema do Grupo Sanguíneo Lutheran/genética , Polimorfismo Genético , Transfusão de Sangue , Genótipo , Humanos , Mutação , Fenótipo , Análise de Sequência de DNA
20.
Hematol., Transfus. Cell Ther. (Impr.) ; 41(1): 25-30, Jan.-Mar. 2019. tab
Artigo em Inglês | LILACS | ID: biblio-1002045

RESUMO

Abstract Background Lutheran and Dombrock are two blood group systems with low immunogenic antigens; they can cause mild-to-moderate transfusion reactions. For both, immunophenotyping is not performed in the pretransfusion routine in Brazil. In addition, the distribution of their antigenic frequencies is an important marker of ethnicity. Thus, the goal of this study was to carry out the genotyping of the LU*01, LU*02, DO*01 and DO*02 alleles of the Lutheran and Dombrock blood group systems in blood donors from the southwestern region of the state of Paraná, Southern Brazil. Method Genotyping was performed for 251 blood donors by specific allele-polymerase chain reaction. The genotype and allele frequencies were obtained through direct counting and compared with other Brazilian populations using the chi-square test with Yates correction. Results The distribution of genotype frequencies for LU were 0.4% for LU*01/LU*01, 6.8% for LU*01/LU*02 and 92.8% for LU*02/LU*02 and for DO, they were 19.9% for DO*01/DO*01, 44.6% for DO*01/DO*02 and 35.5% for DO*02/DO*02. The allele and genotype frequencies of LU and DO were similar to those expected for Caucasians, but the DO*01/DO*01 genotype frequency was different to other Brazilian populations. The rare LU*01/LU*01 genotype was found in a loyal blood donor. Conclusion The genotyping techniques allowed the evaluation of the LU*01, LU*02, DO*01 and DO*02 alleles in blood donors registered in the Hemotherapy Center of the southwestern region of Paraná, Southern Brazil, and contributed to a genotyped blood donor database.


Assuntos
Humanos , Antígenos de Grupos Sanguíneos , Técnicas de Genotipagem , Sistema do Grupo Sanguíneo Lutheran
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...