Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Hum Genome Var ; 8(1): 32, 2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34321463

RESUMO

Chromosomal heteromorphisms (CHMs) are currently largely disregarded in human genetic diagnostics. One exception is der(Y)t(Y;acro)(q12;p1?2), which has at least been mentioned in karyotypes and discussed in reports. This derivative is frequently observed in healthy males with idiopathic infertility, which is not uncommon for CHMs. Here, we present the first systematic fluorescence in situ hybridization (FISH)-based study of 7 carriers of der(Y)t(Y;acro)(q12;p1?2). Specific probes for 15p11.2 (D15Z1) and 22p11.2 (D22Z4) were applied to answer the question of whether either of the short arms may be involved in the formation of the derivative Y-chromosome. In 6 out of 7 cases, specific staining was achieved using the D15Z1 probe, while the derivative acrocentric chromosomal region was not positive for D22Z4 in any of the 7 cases.In conclusion, this study implies that the acrocentric chromosomal region is derived from chromosome 15 in the majority of cases with der(Y)t(Y;acro)(q12;p1?2).

2.
Curr Protoc Stem Cell Biol ; 55(1): e127, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32956561

RESUMO

The reproducibility of stem cell research relies on the constant availability of quality-controlled cells. As the quality of human induced pluripotent stem cells (hiPSCs) can deteriorate in the course of a few passages, cell banking is key to achieve consistent results and low batch-to-batch variation. Here, we provide a cost-efficient route to generate master and working cell banks for basic research projects. In addition, we describe minimal protocols for quality assurance including tests for sterility, viability, pluripotency, and genetic integrity. © 2020 The Authors. Basic Protocol 1: Expansion of hiPSCs Basic Protocol 2: Cell banking of hiPSCs Support Protocol 1: Pluripotency assessment by flow cytometry Support Protocol 2: Thawing control: Viability and sterility Support Protocol 3: Potency, viral clearance, and pluripotency: Spontaneous differentiation and qRT-PCR Support Protocol 4: Identity: Short tandem repeat analysis.


Assuntos
Criopreservação/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes/citologia , Linhagem Celular , Humanos , Controle de Qualidade , Reprodutibilidade dos Testes
3.
Am J Med Genet A ; 182(11): 2793-2796, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32783269

RESUMO

Roberts syndrome (also known as Roberts-SC phocomelia syndrome) is an autosomal recessive developmental disorder, characterized by pre- and postnatal growth retardation, limb malformations including bilateral symmetric tetraphocomelia or mesomelia, and craniofacial dysmorphism. Biallelic loss-of-function variants in ESCO2, which codes for establishment of sister chromatid cohesion N-acetyltransferase 2, cause Roberts syndrome. Phenotypic spectrum among patients is broad, challenging clinical diagnosis in mildly affected individuals. Here we report a 3-year-old boy with a mild phenotype of Roberts syndrome with bilateral elbow contractures, humeroradial synostosis, mild lower limb disparity, and facial dysmorphism. Trio whole-exome sequencing identified the novel biallelic splice variant c.1673+1G>A in ESCO2 in the patient. Aberrant ESCO2 pre-mRNA splicing, reduced relative ESCO2 mRNA amount, and characteristic cytogenetic defects, such as premature centromere separation, heterochromatin repulsion, and chromosome breaks, in patient cells strongly supported pathogenicity of the ESCO2 variant affecting one of the highly conserved guanine-thymine dinucleotide of the donor splice site. Our case highlights the difficulty in establishing a clinical diagnosis in individuals with minor clinical features of Roberts syndrome and normal intellectual and social development. However, next-generation sequencing tools allow for molecular diagnosis in cases presenting with mild developmental defects.


Assuntos
Acetiltransferases/genética , Proteínas Cromossômicas não Histona/genética , Contratura/congênito , Anormalidades Craniofaciais/patologia , Ectromelia/patologia , Cotovelo/patologia , Úmero/anormalidades , Hipertelorismo/patologia , Mutação , Splicing de RNA , Rádio (Anatomia)/anormalidades , Sinostose/patologia , Pré-Escolar , Contratura/complicações , Contratura/genética , Contratura/patologia , Anormalidades Craniofaciais/complicações , Anormalidades Craniofaciais/genética , Ectromelia/complicações , Ectromelia/genética , Homozigoto , Humanos , Úmero/patologia , Hipertelorismo/complicações , Hipertelorismo/genética , Masculino , Fenótipo , Rádio (Anatomia)/patologia , Sinostose/complicações , Sinostose/genética
4.
Stem Cells Dev ; 29(18): 1179-1189, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32723003

RESUMO

Pluripotent stem cells are promising candidates for cell-based regenerative therapies. To avoid rejection of transplanted cells, several approaches are being pursued to reduce immunogenicity of the cells or modulate the recipient's immune response. These include gene editing to reduce the antigenicity of cell products, immunosuppression of the host, or using major histocompatibility complex-matched cells from cell banks. In this context, we have investigated the antigenicity of H-Y antigens, a class of minor histocompatibility antigens encoded by the Y chromosome, to assess whether the gender of the donor affects the cell's antigenicity. In a murine transplant model, we show that the H-Y antigen in undifferentiated embryonic stem cells (ESCs), as well as ESC-derived endothelial cells, provokes T- and B cell responses in female recipients.


Assuntos
Células-Tronco Embrionárias/metabolismo , Rejeição de Enxerto/imunologia , Antígeno H-Y/metabolismo , Animais , Animais Recém-Nascidos , Linfócitos B/imunologia , Feminino , Tolerância Imunológica , Imunidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Células-Tronco , Análise de Sobrevida , Linfócitos T/imunologia
5.
Hum Mutat ; 41(9): 1645-1661, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32623794

RESUMO

The family of Tre2-Bub2-Cdc16 (TBC)-domain containing GTPase activating proteins (RABGAPs) is not only known as key regulatorof RAB GTPase activity but also has GAP-independent functions. Rab GTPases are implicated in membrane trafficking pathways, such as vesicular trafficking. We report biallelic loss-of-function variants in TBC1D2B, encoding a member of the TBC/RABGAP family with yet unknown function, as the underlying cause of cognitive impairment, seizures, and/or gingival overgrowth in three individuals from unrelated families. TBC1D2B messenger RNA amount was drastically reduced, and the protein was absent in fibroblasts of two patients. In immunofluorescence analysis, ectopically expressed TBC1D2B colocalized with vesicles positive for RAB5, a small GTPase orchestrating early endocytic vesicle trafficking. In two independent TBC1D2B CRISPR/Cas9 knockout HeLa cell lines that serve as cellular model of TBC1D2B deficiency, epidermal growth factor internalization was significantly reduced compared with the parental HeLa cell line suggesting a role of TBC1D2B in early endocytosis. Serum deprivation of TBC1D2B-deficient HeLa cell lines caused a decrease in cell viability and an increase in apoptosis. Our data reveal that loss of TBC1D2B causes a neurodevelopmental disorder with gingival overgrowth, possibly by deficits in vesicle trafficking and/or cell survival.


Assuntos
Proteínas Ativadoras de GTPase/genética , Crescimento Excessivo da Gengiva/genética , Transtornos do Neurodesenvolvimento/genética , Convulsões/genética , Adulto , Criança , Endocitose , Feminino , Células HeLa , Humanos , Lactente , Mutação com Perda de Função , Masculino , Linhagem , Sequenciamento do Exoma , Adulto Jovem
7.
J Hum Genet ; 65(9): 783-795, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32398760

RESUMO

Pericentric inversions are among the known polymorphisms detected in the general population at a frequency of 1-2%. Despite their generally benign nature, pericentric inversions affect the reproductive potential of carriers by increasing the risk for unbalanced live-born offspring, miscarriages, or other fertility problems. Here we present a novel large pericentric inversion of chromosome 9, inv(9)(p23q22.3), detected in 30 heterozygote carriers, 24 from seven apparently unrelated families and 6 isolated patients, where the probands were mainly referred for fertility and prenatal problems. The inversion carries a significant risk for recombinant abnormal chromosomes, as in two families one supernumerary rec(9)dup(9p) and one rec(9)dup(9q) were identified, leading to neonatal death and miscarriage, respectively. The inversion carriers were identified by three different laboratories in Greece, Cyprus and Germany respectively, however all carriers have Southeast European origin. The inversion appears to be more frequent in the Greek population, as the majority of the carriers were identified in Greece. We were able to determine that the inversion is identical in all individuals included in the study by applying a combination of several methodologies, such as karyotype, fluorescence in situ hybridization (FISH), chromosomal microarrays (CMA) and haplotype analysis. In addition, haplotype analysis supports that the present inversion is identical by descent (IBD) inherited from a single common ancestor. Our results are, therefore, highly indicative of a founder effect of this inversion, presumably reflecting an event that was present in a small number of individuals that migrated to the current Southeast Europe/Northern Greece from a larger population.


Assuntos
Aborto Espontâneo/genética , Cromossomos Humanos Par 9/genética , Fertilidade/genética , Oligospermia/genética , Morte Perinatal/etiologia , Aborto Espontâneo/epidemiologia , Adulto , Criança , Inversão Cromossômica , Chipre , Feminino , Alemanha , Grécia , Heterozigoto , Humanos , Hibridização in Situ Fluorescente , Recém-Nascido , Cariotipagem , Masculino , Análise em Microsséries , Oligospermia/epidemiologia , Gravidez
8.
Hum Genet ; 139(4): 483-498, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32055997

RESUMO

Biallelic variants in TOR1AIP1, encoding the integral nuclear membrane protein LAP1 (lamina-associated polypeptide 1) with two functional isoforms LAP1B and LAP1C, have initially been linked to muscular dystrophies with variable cardiac and neurological impairment. Furthermore, a recurrent homozygous nonsense alteration, resulting in loss of both LAP1 isoforms, was identified in seven likely related individuals affected by multisystem anomalies with progeroid-like appearance and lethality within the 1st decade of life. Here, we have identified compound heterozygosity in TOR1AIP1 affecting both LAP1 isoforms in two unrelated individuals affected by congenital bilateral hearing loss, ventricular septal defect, bilateral cataracts, mild to moderate developmental delay, microcephaly, mandibular hypoplasia, short stature, progressive muscular atrophy, joint contractures and severe chronic heart failure, with much longer survival. Cellular characterization of primary fibroblasts of one affected individual revealed absence of both LAP1B and LAP1C, constitutively low lamin A/C levels, aberrant nuclear morphology including nuclear cytoplasmic channels, and premature senescence, comparable to findings in other progeroid forms of nuclear envelopathies. We additionally observed an abnormal activation of the extracellular signal-regulated kinase 1/2 (ERK 1/2). Ectopic expression of wild-type TOR1AIP1 mitigated these cellular phenotypes, providing further evidence for the causal role of identified genetic variants. Altogether, we thus further expand the TOR1AIP1-associated phenotype by identifying individuals with biallelic loss-of-function variants who survived beyond the 1st decade of life and reveal novel molecular consequences underlying the TOR1AIP1-associated disorders.


Assuntos
Doenças Genéticas Inatas/genética , Proteínas de Choque Térmico HSC70/genética , Mutação com Perda de Função , Membrana Nuclear/genética , Adulto , Feminino , Humanos , Masculino , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Isoformas de Proteínas
10.
PLoS One ; 13(9): e0204580, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30261016

RESUMO

Following a certain type-specific number of mitotic divisions, terminally differentiated cells undergo proliferative senescence, thwarting efforts to expand different cell populations in vitro for the needs of scientific research or medical therapies. The primary cause of this phenomenon is the progressive shortening of the telomeres and the subsequent activation of cell cycle control pathways leading to a block of cell proliferation. Restoration of telomere length by transgenic expression of telomerase reverse transcriptase (TERT) usually results in bypassing of the replicative senescence and ultimately in cell immortalization. To date, there have not been any reports regarding immortalization of cells from common marmoset (Callithrix jacchus), an important non-human primate model for various human diseases, with the use of exogenous human TERT (hTERT). In this study, marmoset fibroblasts were successfully immortalized with transposon-integrated transgenic hTERT and expanded in vitro for over 500 population doublings. Calculation of population doubling levels (PDL) showed that the derived hTERT-transgenic lines had significantly higher proliferation potential than the wild-type fibroblasts, which reached only a maximum of 46 doublings. However, the immortalized cells exhibited differences in the morphology compared with the control fibroblasts and transcriptome analysis also revealed changes in the gene expression patterns. Finally, the karyotypes of all hTERT-transgenic cell lines showed various aberrations such as presence of extra Chromosome 17, isochromosome 21q, or tetraploidy. By single-cell expansion of the least affected monoclonal immortalized line, one sub-clonal line with normal karyotype was established, suggesting the possibility to derive immortal marmoset cells with normal karyotypes. The results of this study are an important step towards the development and optimization of methods for the production of immortalized cells from common marmoset monkeys.


Assuntos
Callithrix , Fibroblastos/citologia , Fibroblastos/enzimologia , Telomerase/genética , Animais , Linhagem Celular Transformada , Proliferação de Células/genética , Células Cultivadas , Senescência Celular/genética , Elementos de DNA Transponíveis/genética , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Cariotipagem , Proteínas Recombinantes/genética , Homeostase do Telômero/genética
11.
Nat Protoc ; 12(6): 1177-1197, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28492526

RESUMO

Since the advent of the generation of human induced pluripotent stem cells (hiPSCs), numerous protocols have been developed to differentiate hiPSCs into cardiomyocytes and then subsequently assess their ability to recapitulate the properties of adult human cardiomyocytes. However, hiPSC-derived cardiomyocytes (hiPSC-CMs) are often assessed in single-cell assays. A shortcoming of these assays is the limited ability to characterize the physiological parameters of cardiomyocytes, such as contractile force, due to random orientations. This protocol describes the differentiation of cardiomyocytes from hiPSCs, which occurs within 14 d. After casting, cardiomyocytes undergo 3D assembly. This produces fibrin-based engineered heart tissues (EHTs)-in a strip format-that generate force under auxotonic stretch conditions. 10-15 d after casting, the EHTs can be used for contractility measurements. This protocol describes parallel expansion of hiPSCs; standardized generation of defined embryoid bodies, growth factor and small-molecule-based cardiac differentiation; and standardized generation of EHTs. To carry out the protocol, experience in advanced cell culture techniques is required.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos/fisiologia , Engenharia Tecidual/métodos , Humanos
12.
Am J Med Genet A ; 173(5): 1334-1341, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28371302

RESUMO

Pattern of X chromosome inactivation (XCI) is typically random in females. However, chromosomal rearrangements affecting the X chromosome can result in XCI skewing due to cell growth disadvantage. In case of an X;autosome translocation, this usually leads to an XCI pattern of 100:0 with the derivative X being the active one in the majority of females. A de novo balanced X;6 translocation [46,X,t(X;6)(p22.1;q27)] and a completely skewed XCI pattern (100:0) were detected in a female patient with microcephaly, cerebellar vermis hypoplasia, heart defect, and severe developmental delay. We mapped the breakpoint regions using fluorescence in situ hybridization and found the X-linked gene POLA1 to be disrupted. POLA1 codes for the catalytic subunit of the polymerase α-primase complex which is responsible for initiation of the DNA replication process; absence of POLA1 is probably incompatible with life. Consequently, by RBA banding we determined which of the X chromosomes was the active one in the patient. In all examined lymphocytes the wild-type X chromosome was active. We propose that completely skewed XCI favoring the normal X chromosome resulted from death of cells with an active derivative X that was caused by a non-functional POLA1 gene. In summary, we conclude that functional monosomy of 6q27-qter and functional disomy of Xpter-p22.11 are responsible for the clinical phenotype of the patient. This case demonstrates the importance of determining which one of the X chromosomes underwent inactivation to correlate clinical features of a female with an X;autosome translocation with the nature of the genetic alteration.


Assuntos
Cromossomos Humanos X/genética , DNA Polimerase III/genética , Deficiências do Desenvolvimento/genética , Microcefalia/genética , Inativação do Cromossomo X/genética , Adulto , Vermis Cerebelar/fisiopatologia , Cromossomos Humanos Par 6/genética , Deficiências do Desenvolvimento/fisiopatologia , Feminino , Genes Ligados ao Cromossomo X , Genótipo , Humanos , Hibridização in Situ Fluorescente , Microcefalia/fisiopatologia , Monossomia/genética , Fenótipo , Translocação Genética/genética , Dissomia Uniparental/genética
13.
Sci Rep ; 6: 29122, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27385131

RESUMO

Embryonic stem cells (ESCs) are useful for the study of embryonic development. However, since research on naturally conceived human embryos is limited, non-human primate (NHP) embryos and NHP ESCs represent an excellent alternative to the corresponding human entities. Though, ESC lines derived from naturally conceived NHP embryos are still very rare. Here, we report the generation and characterization of four novel ESC lines derived from natural preimplantation embryos of the common marmoset monkey (Callithrix jacchus). For the first time we document derivation of NHP ESCs derived from morula stages. We show that quantitative chromosome-wise transcriptome analyses precisely reflect trisomies present in both morula-derived ESC lines. We also demonstrate that the female ESC lines exhibit different states of X-inactivation which is impressively reflected by the abundance of the lncRNA X inactive-specific transcript (XIST). The novel marmoset ESC lines will promote basic primate embryo and ESC studies as well as preclinical testing of ESC-based regenerative approaches in NHP.


Assuntos
Callithrix/genética , Células-Tronco Embrionárias/metabolismo , Genoma , Transcriptoma/genética , Animais , Biomarcadores/metabolismo , Blastocisto/citologia , Diferenciação Celular/genética , Linhagem Celular , Forma Celular , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Cariotipagem , Masculino , Mórula/citologia , Processos de Determinação Sexual/genética , Teratoma/patologia , Inativação do Cromossomo X/genética
14.
Am J Hum Genet ; 97(3): 493-500, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26340335

RESUMO

CHAMP1 encodes a protein with a function in kinetochore-microtubule attachment and in the regulation of chromosome segregation, both of which are known to be important for neurodevelopment. By trio whole-exome sequencing, we have identified de novo deleterious mutations in CHAMP1 in five unrelated individuals affected by intellectual disability with severe speech impairment, motor developmental delay, muscular hypotonia, and similar dysmorphic features including short philtrum and a tented upper and everted lover lip. In addition to two frameshift and one nonsense mutations, we found an identical nonsense mutation, c.1192C>T (p.Arg398*), in two affected individuals. All mutations, if resulting in a stable protein, are predicted to lead to the loss of the functionally important zinc-finger domains in the C terminus of the protein, which regulate CHAMP1 localization to chromosomes and the mitotic spindle, thereby providing a mechanistic understanding for their pathogenicity. We thus establish deleterious de novo mutations in CHAMP1 as a cause of intellectual disability.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Proteínas Cromossômicas não Histona/genética , Códon sem Sentido/genética , Deficiência Intelectual/genética , Fosfoproteínas/genética , Distúrbios da Fala/genética , Sequência de Bases , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Análise de Sequência de DNA
15.
Orphanet J Rare Dis ; 9: 53, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24735900

RESUMO

BACKGROUND: Segmental Xp22.2 monosomy or a heterozygous HCCS mutation is associated with the microphthalmia with linear skin defects (MLS) or MIDAS (microphthalmia, dermal aplasia, and sclerocornea) syndrome, an X-linked disorder with male lethality. HCCS encodes the holocytochrome c-type synthase involved in mitochondrial oxidative phosphorylation (OXPHOS) and programmed cell death. METHODS: We characterized the X-chromosomal abnormality encompassing HCCS or an intragenic mutation in this gene in six new female patients with an MLS phenotype by cytogenetic analysis, fluorescence in situ hybridization, sequencing, and quantitative real-time PCR. The X chromosome inactivation (XCI) pattern was determined and clinical data of the patients were reviewed. RESULTS: Two terminal Xp deletions of ≥ 11.2 Mb, two submicroscopic copy number losses, one of ~850 kb and one of ≥ 3 Mb, all covering HCCS, 1 nonsense, and one mosaic 2-bp deletion in HCCS are reported. All females had a completely (>98:2) or slightly skewed (82:18) XCI pattern. The most consistent clinical features were microphthalmia/anophthalmia and sclerocornea/corneal opacity in all patients and congenital linear skin defects in 4/6. Additional manifestations included various ocular anomalies, cardiac defects, brain imaging abnormalities, microcephaly, postnatal growth retardation, and facial dysmorphism. However, no obvious clinical sign was observed in three female carriers who were relatives of one patient. CONCLUSION: Our findings showed a wide phenotypic spectrum ranging from asymptomatic females with an HCCS mutation to patients with a neonatal lethal MLS form. Somatic mosaicism and the different ability of embryonic cells to cope with an OXPHOS defect and/or enhanced cell death upon HCCS deficiency likely underlie the great variability in phenotypes.


Assuntos
Doenças Genéticas Ligadas ao Cromossomo X/patologia , Microftalmia/patologia , Anormalidades da Pele/patologia , Pele/patologia , Criança , Pré-Escolar , Cromossomos Humanos X/genética , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/genética , Humanos , Lactente , Microftalmia/genética , Anormalidades da Pele/genética , Inativação do Cromossomo X/genética
16.
Neuropediatrics ; 44(5): 268-71, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23436495

RESUMO

Terminal deletions of chromosome 3p26.3 confined to the CHL1 gene have previously been described in children with intellectual disability and epilepsy. Here, we report for the first time, a 3p26.3 duplication including only the CHL1 gene in an intellectually disabled girl with epilepsy. The penetrance of both deletions and duplications in 3p26.3 is reduced because all chromosomal imbalances were inherited from healthy parents. Further studies are needed to specify the pathogenic mechanism of 3p26.3 imbalances and to estimate recurrence risks in genetic counseling. However, the description of both deletions and duplications of chromosome 3p26.3 in nonsyndromic intellectual disability suggests that CHL1 is a dosage-sensitive gene with an important role for normal cognitive development.


Assuntos
Moléculas de Adesão Celular/genética , Cognição/fisiologia , Epilepsia/genética , Deficiência Intelectual/genética , Adolescente , Duplicação Cromossômica , Epilepsia/complicações , Feminino , Humanos , Deficiência Intelectual/complicações , Fenótipo
17.
J Clin Invest ; 123(3): 1285-98, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23434590

RESUMO

Uniparental parthenotes are considered an unwanted byproduct of in vitro fertilization. In utero parthenote development is severely compromised by defective organogenesis and in particular by defective cardiogenesis. Although developmentally compromised, apparently pluripotent stem cells can be derived from parthenogenetic blastocysts. Here we hypothesized that nonembryonic parthenogenetic stem cells (PSCs) can be directed toward the cardiac lineage and applied to tissue-engineered heart repair. We first confirmed similar fundamental properties in murine PSCs and embryonic stem cells (ESCs), despite notable differences in genetic (allelic variability) and epigenetic (differential imprinting) characteristics. Haploidentity of major histocompatibility complexes (MHCs) in PSCs is particularly attractive for allogeneic cell-based therapies. Accordingly, we confirmed acceptance of PSCs in MHC-matched allotransplantation. Cardiomyocyte derivation from PSCs and ESCs was equally effective. The use of cardiomyocyte-restricted GFP enabled cell sorting and documentation of advanced structural and functional maturation in vitro and in vivo. This included seamless electrical integration of PSC-derived cardiomyocytes into recipient myocardium. Finally, we enriched cardiomyocytes to facilitate engineering of force-generating myocardium and demonstrated the utility of this technique in enhancing regional myocardial function after myocardial infarction. Collectively, our data demonstrate pluripotency, with unrestricted cardiogenicity in PSCs, and introduce this unique cell type as an attractive source for tissue-engineered heart repair.


Assuntos
Células-Tronco Embrionárias/fisiologia , Coração/fisiologia , Engenharia Tecidual , Potenciais de Ação , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Sinalização do Cálcio , Diferenciação Celular , Forma Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Genótipo , Histocompatibilidade/genética , Antígenos de Histocompatibilidade Classe II/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos SCID , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Técnicas de Cultura de Órgãos , Organoides/transplante , Partenogênese , Fenótipo , Transplante de Células-Tronco , Transplante Homólogo
18.
Neurogenetics ; 13(2): 181-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22411505

RESUMO

We performed a replication study in 883 Austrian multiple sclerosis (MS) patients and 972 control individuals for 25 previously risk-associated loci (39 SNPs). Two loci, rs1109670 (DDEF2/MBOAT2, p < 0.02) and rs16914086 (TBC1D2, p < 0.05), are replicated here for the first time. Furthermore, we tested all 39 SNPs for association with age at disease onset and measures of disease severity. We observed a trend for association of rs3135388 (HLA-DRB1*1501, p < 0.01), rs7090530 (IL2RA, p < 0.026) and rs1841770 (ZIC1, p < 0.017) with a younger age at MS onset and of rs12044852 (CD58, p < 0.035) with shorter time to reach EDSS6.


Assuntos
Replicação do DNA , DNA/genética , Predisposição Genética para Doença , Esclerose Múltipla/genética , Adulto , Idade de Início , Idoso , Alelos , Áustria , Estudos de Coortes , DNA/metabolismo , Feminino , Frequência do Gene/genética , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/metabolismo , Polimorfismo de Nucleotídeo Único , Índice de Gravidade de Doença
19.
Cancer Lett ; 256(1): 120-7, 2007 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-17630152

RESUMO

As a culture model to study hepatocarcinogenesis, telomerase-immortalized human fetal hepatocytes were monitored for karyotype changes evolving in long-term culture and development of functional defects in DNA damage response. G-banding revealed acquisition of characteristic karyotype abnormalities, e.g., trisomy 7 and monosomy X, in two independently immortalized and cultured populations after 80-100 population doublings. Interestingly, the detected aneuploidies resemble some of the genetic events observed in hepatocellular cancer. However, these genetic changes were not sufficient to induce oncogenic transformation reflected by absence of anchorage-independent growth. Furthermore, long-term cultured telomerase-immortalized cells preserved p53 expression levels and effective p53-mediated damage response.


Assuntos
Transformação Celular Neoplásica , Aberrações Cromossômicas , Hepatócitos/metabolismo , Telômero/metabolismo , Western Blotting , Ciclo Celular , Células Cultivadas , Senescência Celular , Dano ao DNA , Feto , Regulação da Expressão Gênica , Hepatócitos/citologia , Humanos , Cariotipagem , Mutação , Telomerase/metabolismo , Transdução Genética , Transfecção , Proteína Supressora de Tumor p53/genética
20.
Am J Med Genet A ; 117A(3): 289-94, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12599195

RESUMO

Zimmermann-Laband syndrome (ZLS) is a rare disorder characterized by gingival fibromatosis, abnormalities of the nose and/or ears, and absence or hypoplasia of nails or terminal phalanges of hands and feet. Other more variable features include hyperextensibility of joints, hepatosplenomegaly, mild hirsutism, and mental retardation. The genetic basis of ZLS is unknown; autosomal dominant inheritance has been suggested. We report an apparently balanced chromosomal aberration, 46,XX, t(3;8)(p13-p21.2;q24.1-q24.3), in a family with an affected mother and daughter. Using fluorescence in situ hybridization with BAC clones, we refined the breakpoints to 3p21.2 and 8q24.3 and, thereby, narrowed down both breakpoint regions to approximately 1.5 Mb. Our data provide additional support to the assumption that ZLS follows autosomal dominant inheritance. The 3;8 translocation described here represents a powerful resource to identify the causative gene for ZLS that maps most likely to one of the breakpoints.


Assuntos
Anormalidades Múltiplas/genética , Cromossomos Humanos Par 3/genética , Cromossomos Humanos Par 8/genética , Translocação Genética , Anormalidades Múltiplas/patologia , Pré-Escolar , Bandeamento Cromossômico , Quebra Cromossômica/genética , Transtornos Cromossômicos/genética , Transtornos Cromossômicos/patologia , Ossos Faciais/anormalidades , Saúde da Família , Feminino , Fibromatose Gengival/patologia , Dedos/anormalidades , Humanos , Hibridização in Situ Fluorescente , Linhagem , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA