Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cells ; 11(16)2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-36010549

RESUMEN

Cultured red blood cells from human induced pluripotent stem cells (cRBC_iPSCs) are a promising source for future concepts in transfusion medicine. Before cRBC_iPSCs will have entrance into clinical or laboratory use, their functional properties and safety have to be carefully validated. Due to the limitations of established culture systems, such studies are still missing. Improved erythropoiesis in a recently established culture system, closer simulating the physiological niche, enabled us to conduct functional characterization of enucleated cRBC_iPSCs with a focus on membrane properties. Morphology and maturation stage of cRBC_iPSCs were closer to native reticulocytes (nRETs) than to native red blood cells (nRBCs). Whereas osmotic resistance of cRBC_iPSCs was similar to nRETs, their deformability was slightly impaired. Since no obvious alterations in membrane morphology, lipid composition, and major membrane associated protein patterns were observed, reduced deformability might be caused by a more primitive nature of cRBC_iPSCs comparable to human embryonic- or fetal liver erythropoiesis. Blood group phenotyping of cRBC_iPSCs further confirmed the potency of cRBC_iPSCs as a prospective device in pre-transfusional routine diagnostics. Therefore, RBC membrane analyses obtained in this study underscore the overall prospects of cRBC_iPSCs for their future application in the field of transfusion medicine.


Asunto(s)
Células Madre Pluripotentes Inducidas , Diferenciación Celular/fisiología , Eritrocitos/metabolismo , Eritropoyesis , Humanos , Estudios Prospectivos
2.
Hum Mutat ; 43(1): 42-55, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34816548

RESUMEN

γ-Glutamyl carboxylase (GGCX) catalyzes the γ-carboxylation of 15 different vitamin K dependent (VKD) proteins. Pathogenic variants in GGCX cause a rare hereditary bleeding disorder called Vitamin K dependent coagulation factor deficiency type 1 (VKCFD1). In addition to bleedings, some VKCFD1 patients develop skin laxity and skeletal dysmorphologies. However, the pathophysiological mechanisms underlying these non-hemorrhagic phenotypes remain elusive. Therefore, we have analyzed 20 pathogenic GGCX variants on their ability to γ-carboxylate six non-hemostatic VKD proteins in an in vitro assay, where GGCX variants were expressed in GGCX-/- cells and levels of γ-carboxylated co-expressed VKD proteins were detected by a functional ELISA. We observed that GGCX variants causing markedly reduced γ-carboxylation of Gla rich protein (GRP) in vitro were reported in patients with skin laxity. Reduced levels of γ-carboxylated Matrix gla protein (MGP) are not exclusive for causing skeletal dysmorphologies in VKCFD1 patients. In silico docking of vitamin K hydroquinone on a GGCX model revealed a binding site, which was validated by in vitro assays. GGCX variants affecting this site result in disability to γ-carboxylate VKD proteins and hence are involved in the most severe phenotypes. This genotype-phenotype analysis will help to understand the development of non-hemorrhagic phenotypes and hence improve treatment in VKCFD1 patients.


Asunto(s)
Trastornos de la Coagulación Sanguínea Heredados , Ligasas de Carbono-Carbono , Trastornos de la Coagulación Sanguínea Heredados/genética , Ligasas de Carbono-Carbono/química , Ligasas de Carbono-Carbono/genética , Ligasas de Carbono-Carbono/metabolismo , Carboxiliasas , Humanos , Mutación
3.
J Thromb Haemost ; 19(6): 1412-1424, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33590680

RESUMEN

BACKGROUND: Vitamin K dependent coagulation factor deficiency type 1 (VKCFD1) is a rare hereditary bleeding disorder caused by mutations in γ-glutamyl carboxylase (GGCX). VKCFD1 patients are treated life-long with high doses of vitamin K in order to correct the bleeding phenotype. However, normalization of clotting factor activities cannot be achieved for all VKCFD1 patients. OBJECTIVE: The current study aims to investigate the responsiveness to vitamin K for all reported GGCX mutations with respect to clotting factors in order to optimize treatment. METHODS: This study developed an assay using genetically engineered GGCX-/- cells, in which GGCX mutations were analyzed with respect to their ability to γ-carboxylate vitamin K dependent pro-coagulatory and anti-coagulatory clotting factors by ELISA. Additionally, factor VII activity was measured in order to proof protein functionality. For specific GGCX mutations immunofluorescent staining was performed to assess the intracellular localization of clotting factors with respect to GGCX wild-type and mutations. RESULTS: All GGCX mutations were categorized into responder and low responder mutations, thereby determining the efficiency of vitamin K supplementation. Most VKCFD1 patients have at least one vitamin K responsive GGCX allele that is able to γ-carboxylate clotting factors. In few patients, the hemorrhagic phenotype cannot be reversed by vitamin K administration because GGCX mutations on both alleles affect either structural or catalytically important sites thereby resulting in residual ability to γ-carboxylate clotting factors. CONCLUSION: With these new functional data we can predict the hemorrhagic outcome of each VKCFD1 genotype, thus recommending treatments with either vitamin K or prothrombin complex concentrate.


Asunto(s)
Ligasas de Carbono-Carbono , Vitamina K , Ligasas de Carbono-Carbono/genética , Humanos , Mutación , Fenotipo , Vitamina K 1 , Vitamina K Epóxido Reductasas/genética
4.
Int J Mol Sci ; 21(20)2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096636

RESUMEN

In humans, Factor VIII (F8) deficiency leads to hemophilia A and F8 is largely synthesized and secreted by the liver sinusoidal endothelial cells (LSECs). However, the specificity and characteristics of these cells in comparison to other endothelial cells is not well known. In this study, we performed genome wide expression and CpG methylation profiling of fetal and adult human primary LSECs together with other fetal primary endothelial cells from lung (micro-vascular and arterial), and heart (micro-vascular). Our results reveal expression and methylation markers distinguishing LSECs at both fetal and adult stages. Differential gene expression of fetal LSECs in comparison to other fetal endothelial cells pointed to several differentially regulated pathways and biofunctions in fetal LSECs. We used targeted bisulfite resequencing to confirm selected top differentially methylated regions. We further designed an assay where we used the selected methylation markers to test the degree of similarity of in-house iPS generated vascular endothelial cells to primary LSECs; a higher similarity was found to fetal than to adult LSECs. In this study, we provide a detailed molecular profile of LSECs and a guide to testing the effectiveness of production of in vitro differentiated LSECs.


Asunto(s)
Células Endoteliales/fisiología , Hígado/citología , Hígado/embriología , Islas de CpG , Metilación de ADN , Células Endoteliales/citología , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/fisiología , Epigénesis Genética , Factor VIII/genética , Perfilación de la Expresión Génica , Marcadores Genéticos , Humanos , Pulmón/citología , Pulmón/embriología , Masculino , Persona de Mediana Edad , Análisis de la Célula Individual , Sulfitos , Secuenciación Completa del Genoma
5.
Front Cell Dev Biol ; 8: 618760, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33585456

RESUMEN

Development of clinical stem cell interventions are hampered by immature cell progeny under current protocols. Human mesenchymal stem cells (hMSCs) are characterized by their ability to self-renew and differentiate into multiple lineages. Generating hMSCs from pluripotent stem cells (iPSCs) is an attractive avenue for cost-efficient and scalable production of cellular material. In this study we generate mature osteoblasts from iPSCs using a stable expandable MSC intermediate, refining established protocols. We investigated the timeframe and phenotype of cells under osteogenic conditions as well as the effect of menaquinone-7 (MK-7) on differentiation. From day 2 we noted a significant increase in RUNX2 expression under osteogenic conditions with MK-7, as well as decreases in ROS species production, increased cellular migration and changes to dynamics of collagen deposition when compared to differentiated cells that were not treated with MK-7. At day 21 OsteoMK-7 increased alkaline phosphatase activity and collagen deposition, as well as downregulated RUNX2 expression, suggesting to a mature cellular phenotype. Throughout we note no changes to expression of osteocalcin suggesting a non-canonical function of MK-7 in osteoblast differentiation. Together our data provide further mechanistic insight between basic and clinical studies on extrahepatic activity of MK-7. Our findings show that MK-7 promotes osteoblast maturation thereby increasing osteogenic differentiation.

6.
PLoS One ; 10(6): e0130416, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26121690

RESUMEN

Mutations in the maternal effect gene NLRP7 cause biparental hydatidiform mole (HYDM1). HYDM1 is characterized by abnormal growth of placenta and lack of proper embryonic development. The molar tissues are characterized by abnormal methylation patterns at differentially methylated regions (DMRs) of imprinted genes. It is not known whether this occurs before or after fertilization, but the high specificity of this defect to the maternal allele indicates a possible maternal germ line-specific effect. To better understand the unknown molecular mechanism leading to HYDM1, we performed a yeast two-hybrid screen against an ovarian library using NLRP7 as bait. We identified the transcriptional repressor ZBTB16 as an interacting protein of NLRP7 and verified this interaction in mammalian cells by immunoprecipitation and confocal microscopy. Native protein analysis detected NLRP7 and ZBTB16 in a 480kD protein complex and both proteins co-localize in the cytoplasm in juxtanuclear aggregates. HYDM1-causing mutations in NLRP7 did not show altered patterns of interaction with ZBTB16. Hence, the biological significance of the NLRP7-ZBTB16 interaction remains to be revealed. However, a clear effect of harvesting ZBTB16 to the cytoplasm when the NLRP7 protein is overexpressed may be linked to the pathology of the molar pregnancy disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Mola Hidatiforme/genética , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/fisiología , Alelos , Citoplasma/metabolismo , Metilación de ADN , Femenino , Regulación de la Expresión Génica , Biblioteca de Genes , Células HEK293 , Humanos , Factores de Transcripción de Tipo Kruppel/fisiología , Masculino , Microscopía Confocal , Mutación , Ovario/metabolismo , Embarazo , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Técnicas del Sistema de Dos Híbridos
7.
Clin Epigenetics ; 7: 17, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25798207

RESUMEN

BACKGROUND: Hypomethylation of long interspersed element (LINE)-1 has been observed in tumorigenesis when using degenerate assays, which provide an average across all repeats. However, it is unknown whether individual LINE-1 loci or different CpGs within one specific LINE-1 promoter are equally affected by methylation changes. Conceivably, studying methylation changes at specific LINE-1 may be more informative than global assays for cancer diagnostics. Therefore, with the aim of mapping methylation at individual LINE-1 loci at single-CpG resolution and exploring the diagnostic potential of individual LINE-1 locus methylation, we analyzed methylation at 11 loci by pyrosequencing, next-generation bisulfite sequencing as well as global LINE-1 methylation in bladder, colon, pancreas, prostate, and stomach cancers compared to paired normal tissues and in blood samples from some of the patients compared to healthy donors. RESULTS: Most (72/80) tumor samples harbored significant methylation changes at at least one locus. Notably, our data revealed not only the expected hypomethylation but also hypermethylation at some loci. Specific CpGs within the LINE-1 consensus sequence appeared preferentially hypomethylated suggesting that these could act as seeds for hypomethylation. In silico analysis revealed that these CpG sites more likely faced the histones in the nucleosome. Multivariate logistic regression analysis did not reveal a significant clinical advantage of locus-specific methylation markers over global methylation markers in distinguishing tumors from normal tissues. CONCLUSIONS: Methylation changes at individual LINE-1 loci are heterogeneous, whereas specific CpGs within the consensus sequence appear to be more prone to hypomethylation. With a broader selection of loci, locus-specific LINE-1 methylation could become a tool for tumor detection.

8.
Mol Hum Reprod ; 20(10): 990-1001, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25082979

RESUMEN

Mutations in NLRP7 (NOD-like-receptor family, pyrin domain containing 7) are responsible for a type of recurrent pregnancy loss known as recurrent hydatidiform mole (HYDM1). This condition is characterized by abnormal growth of the placenta, a lack of proper embryonic development and abnormal methylation patterns at multiple imprinted loci in diploid biparental molar tissues. The role of NLRP7 protein in the disease manifestation is currently not clear. In order to better understand how the effects of HYDM1 are associated with mutations on the structure of NLRP7, we performed an inter-domain interaction screen using a yeast two-hybrid system. Additionally, we generated in silico structural models of NLRP7 in its non-activated and activated forms. Our observations from the yeast two-hybrid screen and modeling suggest that the NACHT-associated domain (NAD) of the NLRP7 protein is central to its oligomeric assembly. Upon activation, the NAD and a small part of the leucine rich repeat (LRR) of one molecule emerged out of the protective LRR domain and interact with the NACHT domain of the second molecule to form an oligomer. Furthermore, we investigated the molecular basis for the pathophysiological effect of four missense mutations, three HYDM1-causing and one rare non-synonymous variant, on the protein using confocal microscopy of transiently transfected NLRP7 in HEK293T cells and in silico structural analysis. We found that with the two clinically severe missense mutations, L398R and R693W, the normal molecule to molecule interaction was apparently affected thus decreasing their oligomerization potential while aggresome formation was increased; these changes could disturb the normal downstream functions of NLRP7 and therefore be a possible molecular effect underlying their pathophysiological impact.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Mola Hidatiforme/genética , Pliegue de Proteína , Deficiencias en la Proteostasis/genética , Línea Celular , Femenino , Células HEK293 , Humanos , Modelos Moleculares , Mutación Missense , NAD/genética , Placentación , Embarazo , Agregación Patológica de Proteínas/genética , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína , Técnicas del Sistema de Dos Híbridos
9.
Hum Mol Genet ; 21(1): 219-35, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21972244

RESUMEN

LINE-1 repeats account for ~17% of the human genome. Little is known about their individual methylation patterns, because their repetitive, almost identical sequences make them difficult to be individually targeted. Here, we used bisulfite conversion to study methylation at individual LINE-1 repeats. The loci studied included 39 X-linked loci and 5 autosomal loci. On the X chromosome in women, we found statistically significant less methylation at almost all L1Hs compared with men. Methylation at L1P and L1M did not correlate with the inactivation status of the host DNA, while the majority of L1Hs that were possible to be studied lie in inactivated regions. To investigate whether the male-female differences at L1Hs on the X are linked to the inactivation process itself rather than to a mere influence of gender, we analyzed six of the L1Hs loci on the X chromosome in Turners and Klinefelters which have female and male phenotype, respectively, but with reversed number of X chromosomes. We could confirm that all samples with two X chromosomes are hypomethylated at the L1Hs loci. Therefore, the inactive X is hypomethylated at L1Hs; the latter could play an exclusive role in the X chromosome inactivation process. At autosomal L1Hs, methylation levels showed a correlation tendency between methylation level and genome size, with higher methylation observed at most loci in individuals with one X chromosome and the lowest in XXY individuals. In summary, loci-specific LINE-1 methylation levels show considerable plasticity and depend on genomic position and constitution.


Asunto(s)
Cromosomas Humanos X/genética , Tamaño del Genoma , Síndrome de Klinefelter/genética , Elementos de Nucleótido Esparcido Largo , Mutagénesis Insercional , Regiones Promotoras Genéticas , Síndrome de Turner/genética , Inactivación del Cromosoma X , Adulto , Metilación de ADN , Femenino , Humanos , Masculino , Adulto Joven
10.
Methods Mol Biol ; 791: 89-100, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21913073

RESUMEN

Genome-wide sequence-specific methylation analysis has become a readily available and affordable procedure in epigenetic laboratories. Most of these procedures are based on immunoprecipitation, microarrays, or next generation deep bisulfite sequencing. However, most of these protocols are far from being quantitative. Moreover, abnormal or specific methylation patterns must always be further validated by quantitative sequence-specific methylation analysis. In this chapter, we describe a detailed and simplified protocol (using one universal HPLC gradient for all separations as well as one SNuPE annealing temperature for all primers) for the previously published SIRPH analysis, which is based on the single nucleotide primer extension combined with high-performance liquid chromatography. This method is highly accurate, reproducible, quantitative, and suitable for analysis of as little as 50 ng of PCR product derived from limited starting materials.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Cromatografía de Fase Inversa/métodos , Islas de CpG/genética , Metilación de ADN/genética , Cartilla de ADN/genética
11.
PLoS One ; 6(1): e16252, 2011 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-21311577

RESUMEN

Previously, we reported on inter-individual and gender specific variations of LINE-1 methylation in healthy individuals. In this study, we investigated whether this variability could be influenced by age or sex hormones in humans. To this end, we studied LINE-1 methylation in vivo in blood-derived DNA from individuals aged 18 to 64 years and from young healthy females at various hormone levels during the menstrual cycle. Our results show that no significant association with age was observed. However, the previously reported increase of LINE-1 methylation in males was reconfirmed. In females, although no correlation between LINE-1 or Alu methylation and hormone levels was observed, a significant stable individual specific level of methylation was noted. In vitro results largely confirmed these findings, as neither estrogen nor dihydrotestosterone affected LINE-1 or Alu methylation in Hek293T, HUVEC, or MDA-kb2 cell lines. In contrast, a decrease in methylation was observed in estrogen-treated T47-Kbluc cell lines strongly expressing estrogen receptor. The very low expression of estrogen receptor in blood cells could explain the observed insensitivity of methylation at LINE-1 to natural hormonal variations in females. In conclusion, neither natural cycle of hormones nor age has a detectable effect on the LINE-1 methylation in peripheral blood cells, while gender remains an important factor.


Asunto(s)
Metilación de ADN , ADN/sangre , Elementos de Nucleótido Esparcido Largo/genética , Adolescente , Adulto , Factores de Edad , Sangre , Línea Celular , Femenino , Hormonas Esteroides Gonadales/análisis , Hormonas Esteroides Gonadales/fisiología , Humanos , Masculino , Persona de Mediana Edad , Receptores de Estrógenos/análisis , Receptores de Estrógenos/fisiología , Factores Sexuales , Adulto Joven
12.
Blood ; 107(7): 2759-65, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16339403

RESUMEN

Hemophilia A (HA) is caused by partial or total deficiency of F8 protein activity. In a small group, about 1.8% of patients with HA, no mutation is found in the F8 gene. Among this group, we report here on one patient with severe HA in whom no mRNA of the F8 gene was detected. Using 2 common polymorphisms in F8 exon 14, we were able to show that the same allele shared by the patient, his mother, and his sister was not detected by reverse transcription-polymerase chain reaction (RT-PCR) from total blood mRNA. Skewed X-chromosome inactivation in both the mother and the sister was excluded by studying the methylation profile of the androgen receptor gene (HUMARA locus). These findings strongly suggest that the cause of HA in this patient is either absence or rapid degradation of the F8 mRNA, which points to a novel mechanism leading to HA.


Asunto(s)
Factor VIII/genética , Hemofilia A/genética , Mutación , Adolescente , Metilación de ADN , Familia , Femenino , Humanos , Masculino , Linaje , ARN Mensajero/genética , Receptores Androgénicos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA