Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Stem Cell Rev Rep ; 20(1): 67-87, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37768523

RESUMO

Polycystic ovary syndrome (PCOS) is the most prevalent endocrine condition among women with pleiotropic sequelae possessing reproductive, metabolic, and psychological characteristics. Although the exact origin of PCOS is elusive, it is known to be a complex multigenic disorder with a genetic, epigenetic, and environmental background. However, the pathogenesis of PCOS, and the role of genetic variants in increasing the risk of the condition, are still unknown due to the lack of an appropriate study model. Since the debut of induced pluripotent stem cell (iPSC) technology, the ability of reprogrammed somatic cells to self-renew and their potential for multidirectional differentiation have made them excellent tools to study different disease mechanisms. Recently, researchers have succeeded in establishing human in vitro PCOS disease models utilizing iPSC lines from heterogeneous PCOS patient groups (iPSCPCOS). The current review sets out to summarize, for the first time, our current knowledge of the implications and challenges of iPSC technology in comprehending PCOS pathogenesis and tissue-specific disease mechanisms. Additionally, we suggest that the analysis of polygenic risk prediction based on genome-wide association studies (GWAS) could, theoretically, be utilized when creating iPSC lines as an additional research tool to identify women who are genetically susceptible to PCOS. Taken together, iPSCPCOS may provide a new paradigm for the exploration of PCOS tissue-specific disease mechanisms.


Assuntos
Células-Tronco Pluripotentes Induzidas , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Estudo de Associação Genômica Ampla , Diferenciação Celular
2.
J Ovarian Res ; 16(1): 194, 2023 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-37726790

RESUMO

The nuclear receptor subfamily 5 group A member 1 (NR5A1), encoding steroidogenic factor 1 (SF-1), has been identified as a critical factor in gonadal development in animal studies. A previous study of ours suggested that upregulation of NR5A1 during early gonadal differentiation in male (46,XY) human pluripotent stem cells steers the cells into a more mature gonadal cell type. However, the detailed role of NR5A1 in female gonadal differentiation has yet to be determined. In this study, by combining the processes of gonadal differentiation and conditional gene activation, we show that NR5A1 induction predominantly upregulates the female gonadal marker inhibin subunit α (INHA) and steroidogenic markers steroidogenic acute regulatory protein (STAR), cytochrome P450 family 11 subfamily A member 1 (CYP11A1), cytochrome P450 family 17 subfamily A member 1 (CYP17A1), hydroxy-delta-5-steroid dehydrogenase (HSD3B2) and hydroxysteroid 17-beta dehydrogenase 1 (HSD17B1). In contrast, NR5A1 induction did not seem to affect the bipotential gonadal markers gata binding protein 4 (GATA4) and Wilms' tumour suppressor 1 (WT1) nor the female gonadal markers r-spondin 1 (RSPO1) and wnt family member 4 (WNT4). Differentially expressed genes were highly associated with adrenal and ovarian steroidogenesis pathways. Moreover, time-series analysis revealed different dynamic changes between male and female induced samples, where continuously upregulated genes in female gonadal differentiation were mostly associated with adrenal steroidogenesis. Thus, in contrast to male gonadal differentiation, NR5A1 is necessary but not sufficient to steer human embryonic stem cell (hESC)-derived bipotential gonadal-like cells towards a more mature somatic, female cell fate. Instead, it seems to direct bipotential gonadal-like cells more towards a steroidogenic-like cell population. The information obtained in this study helps in elucidating the role of NR5A1 in gonadal differentiation of a female stem cell line.


Assuntos
Células-Tronco Embrionárias Humanas , Animais , Humanos , Feminino , Masculino , Sistemas CRISPR-Cas , Fator Esteroidogênico 1/genética , Diferenciação Celular/genética , Família 17 do Citocromo P450
3.
Reprod Biomed Online ; 47(4): 103287, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37603956

RESUMO

RESEARCH QUESTION: Are age-normalized reference values for human ovarian cortical follicular density adequate for tissue quality control in fertility preservation? DESIGN: Published quantitative data on the number of follicles in samples without known ovarian pathology were converted into cortical densities to create reference values. Next, a sample cohort of 126 girls (age 1-24 years, mean ± SD 11 ± 6) with cancer, severe haematological disease or Turner syndrome were used to calculate Z-scores for cortical follicular density based on the reference values. RESULTS: No difference was observed between Z-scores in samples from untreated patients (0.3 ± 3.5, n = 30) and patients treated with (0.5 ± 2.9, n = 48) and without (0.1 ± 1.3, n = 6) alkylating chemotherapy. Z-scores were not correlated with increasing cumulative exposure to cytostatics. Nevertheless, Z-scores in young treated patients (0-2 years -2.1 ± 3.1, n = 10, P = 0.04) were significantly lower than Z-scores in older treated patients (11-19 years, 2 ± 1.9, n = 15). Samples from patients with Turner syndrome differed significantly from samples from untreated patients (-5.2 ± 5.1, n = 24, P = 0.003), and a Z-score of -1.7 was identified as a cut-off showing good diagnostic value for identification of patients with Turner syndrome with reduced ovarian reserve. When this cut-off was applied to other patients, analysis showed that those with indications for reduced ovarian reserve (n = 15) were significantly younger (5.9 ± 4.2 versus 10.7 ± 5.9 years, P = 0.004) and, when untreated, more often had non-malignant haematologic diseases compared with those with normal ovarian reserve (n = 24, 100% versus 19%, P = 0.009). CONCLUSION: Z-scores allow the estimation of genetic- and treatment-related effects on follicular density in cortical tissue from young patients stored for fertility preservation. Understanding the quality of cryopreserved tissue facilitates its use during patient counselling. More research is needed regarding the cytostatic effects found in this study.


Assuntos
Síndrome de Turner , Feminino , Humanos , Idoso , Lactente , Pré-Escolar , Criança , Adolescente , Adulto Jovem , Adulto , Ovário , Padrões de Referência , Controle de Qualidade , Antineoplásicos Alquilantes
4.
Stem Cells ; 40(12): 1107-1121, 2022 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-36153707

RESUMO

Hypothalamic gonadotropin-releasing hormone (GnRH) neurons lay the foundation for human development and reproduction; however, the critical cell populations and the entangled mechanisms underlying the development of human GnRH neurons remain poorly understood. Here, by using our established human pluripotent stem cell-derived GnRH neuron model, we decoded the cellular heterogeneity and differentiation trajectories at the single-cell level. We found that a glutamatergic neuron population, which generated together with GnRH neurons, showed similar transcriptomic properties with olfactory sensory neuron and provided the migratory path for GnRH neurons. Through trajectory analysis, we identified a specific gene module activated along the GnRH neuron differentiation lineage, and we examined one of the transcription factors, DLX5, expression in human fetal GnRH neurons. Furthermore, we found that Wnt inhibition could increase DLX5 expression and improve the GnRH neuron differentiation efficiency through promoting neurogenesis and switching the differentiation fates of neural progenitors into glutamatergic neurons/GnRH neurons. Our research comprehensively reveals the dynamic cell population transition and gene regulatory network during GnRH neuron differentiation.


Assuntos
Hormônio Liberador de Gonadotropina , Células-Tronco Pluripotentes , Humanos , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Via de Sinalização Wnt/genética , Neurônios/metabolismo , Diferenciação Celular/genética , Células-Tronco Pluripotentes/metabolismo
5.
Dis Model Mech ; 15(8)2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35833364

RESUMO

Fibroblast growth factor 8 (FGF8), acting through the fibroblast growth factor receptor 1 (FGFR1), has an important role in the development of gonadotropin-releasing hormone-expressing neurons (GnRH neurons). We hypothesized that FGF8 regulates differentiation of human GnRH neurons in a time- and dose-dependent manner via FGFR1. To investigate this further, human pluripotent stem cells were differentiated during 10 days of dual-SMAD inhibition into neural progenitor cells, followed either by treatment with FGF8 at different concentrations (25 ng/ml, 50 ng/ml or 100 ng/ml) for 10 days or by treatment with 100 ng/ml FGF8 for different durations (2, 4, 6 or 10 days); cells were then matured through DAPT-induced inhibition of Notch signaling for 5 days into GnRH neurons. FGF8 induced expression of GNRH1 in a dose-dependent fashion and the duration of FGF8 exposure correlated positively with gene expression of GNRH1 (P<0.05, Rs=0.49). However, cells treated with 100 ng/ml FGF8 for 2 days induced the expression of genes, such as FOXG1, ETV5 and SPRY2, and continued FGF8 treatment induced the dynamic expression of several other genes. Moreover, during exposure to FGF8, FGFR1 localized to the cell surface and its specific inhibition with the FGFR1 inhibitor PD166866 reduced expression of GNRH1 (P<0.05). In neurons, FGFR1 also localized to the nucleus. Our results suggest that dose- and time-dependent FGF8 signaling via FGFR1 is indispensable for human GnRH neuron ontogeny. This article has an associated First Person interview with the first author of the paper.


Assuntos
Hormônio Liberador de Gonadotropina , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Fator 8 de Crescimento de Fibroblasto/metabolismo , Fator 8 de Crescimento de Fibroblasto/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Neurônios/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo
6.
Nat Commun ; 12(1): 5603, 2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34556652

RESUMO

Although the first dissection of the human ovary dates back to the 17th century, the biophysical characteristics of the ovarian cell microenvironment are still poorly understood. However, this information is vital to deciphering cellular processes such as proliferation, morphology and differentiation, as well as pathologies like tumor progression, as demonstrated in other biological tissues. Here, we provide the first readout of human ovarian fiber morphology, interstitial and perifollicular fiber orientation, pore geometry, topography and surface roughness, and elastic and viscoelastic properties. By determining differences between healthy prepubertal, reproductive-age, and menopausal ovarian tissue, we unravel and elucidate a unique biophysical phenotype of reproductive-age tissue, bridging biophysics and female fertility. While these data enable to design of more biomimetic scaffolds for the tissue-engineered ovary, our analysis pipeline is applicable for the characterization of other organs in physiological or pathological states to reveal their biophysical markers or design their bioinspired analogs.


Assuntos
Ovário/anatomia & histologia , Ovário/fisiologia , Adulto , Fatores Etários , Idoso , Bioengenharia , Criança , Pré-Escolar , Tecido Elástico/anatomia & histologia , Tecido Elástico/metabolismo , Elasticidade , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Feminino , Hormônios/metabolismo , Humanos , Pessoa de Meia-Idade , Folículo Ovariano/crescimento & desenvolvimento , Reserva Ovariana , Ovário/citologia , Viscosidade , Adulto Jovem
7.
Dis Model Mech ; 13(3)2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-31996360

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons provide a fundamental signal for the onset of puberty and subsequent reproductive functions by secretion of gonadotropin-releasing hormone. Their disrupted development or function leads to congenital hypogonadotropic hypogonadism (CHH). To model the development of human GnRH neurons, we generated a stable GNRH1-TdTomato reporter cell line in human pluripotent stem cells (hPSCs) using CRISPR-Cas9 genome editing. RNA-sequencing of the reporter clone, differentiated into GnRH neurons by dual SMAD inhibition and FGF8 treatment, revealed 6461 differentially expressed genes between progenitors and GnRH neurons. Expression of the transcription factor ISL1, one of the top 50 most upregulated genes in the TdTomato-expressing GnRH neurons, was confirmed in 10.5 gestational week-old human fetal GnRH neurons. Among the differentially expressed genes, we detected 15 genes that are implicated in CHH and several genes that are implicated in human puberty timing. Finally, FGF8 treatment in the neuronal progenitor pool led to upregulation of 37 genes expressed both in progenitors and in TdTomato-expressing GnRH neurons, which suggests upstream regulation of these genes by FGF8 signaling during GnRH neuron differentiation. These results illustrate how hPSC-derived human GnRH neuron transcriptomic analysis can be utilized to dissect signaling pathways and gene regulatory networks involved in human GnRH neuron development.This article has an associated First Person interview with the first author of the paper.


Assuntos
Genes Reporter , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Células-Tronco Pluripotentes/metabolismo , Transcriptoma/genética , Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Linhagem Celular , Feto/citologia , Fator 8 de Crescimento de Fibroblasto/farmacologia , Humanos , Hipogonadismo/genética , Proteínas com Homeodomínio LIM/metabolismo , Neurônios/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
8.
Mol Cell Endocrinol ; 479: 103-109, 2019 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-30287399

RESUMO

While human chorionic gonadotropin (hCG) appears to have an essential role in early pregnancy, it is controversial whether the hyperglycosylated form of hCG (hCG-h), which is the major hCG isoform during the first 4-5 weeks of pregnancy, is able to activate LH/hCG receptor (LHCGR). To address this, we utilized different extensively characterized hCG and hCGß reference reagents, cell culture- and urine-derived hCG-h preparations, and an in vitro reporter system for LHCGR activation. The WHO hCG reference reagent (99/688) was found to activate LHCGR with an EC50-value of 3.3 ±â€¯0.6 pmol/L (n = 9). All three studied hCG-h preparations were also able to activate LHCGR, but with a lower potency (EC50-values between 7.1 ±â€¯0.5 and 14 ±â€¯3 pmol/L, n = 5-11, for all P < 0.05 as compared to the hCG reference). The activities of commercial urinary hCG (Pregnyl) and recombinant hCG (Ovitrelle) preparations were intermediate between those of the hCG reference and the hCG-h. These results strongly suggest that the hCG-h is functionally similar to hCG, although it has lower potency for LHCGR activation. Whether this explains the reduced proportion of hCG-h to hCG reported in patients developing early onset pre-eclampsia or those having early pregnancy loss remains to be determined.


Assuntos
Gonadotropina Coriônica/farmacologia , Hormônio Luteinizante/metabolismo , Receptores do LH/metabolismo , Animais , Gonadotropina Coriônica Humana Subunidade beta/farmacologia , Cães , Glicosilação , Humanos , Células Madin Darby de Rim Canino
9.
Stem Cell Reports ; 7(2): 149-57, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27426041

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons regulate human puberty and reproduction. Modeling their development and function in vitro would be of interest for both basic research and clinical translation. Here, we report a three-step protocol to differentiate human pluripotent stem cells (hPSCs) into GnRH-secreting neurons. Firstly, hPSCs were differentiated to FOXG1, EMX2, and PAX6 expressing anterior neural progenitor cells (NPCs) by dual SMAD inhibition. Secondly, NPCs were treated for 10 days with FGF8, which is a key ligand implicated in GnRH neuron ontogeny, and finally, the cells were matured with Notch inhibitor to bipolar TUJ1-positive neurons that robustly expressed GNRH1 and secreted GnRH decapeptide into the culture medium. The protocol was reproducible both in human embryonic stem cells and induced pluripotent stem cells, and thus provides a translational tool for investigating the mechanisms of human puberty and its disorders.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Células-Tronco Pluripotentes/citologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Humanos , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurônios/efeitos dos fármacos , Nariz/citologia , Células-Tronco Pluripotentes/metabolismo , Receptores Notch/metabolismo , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/metabolismo
10.
PLoS One ; 10(3): e0116668, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25822230

RESUMO

Small RNA molecules, including microRNAs (miRNAs), play critical roles in regulating pluripotency, proliferation and differentiation of embryonic stem cells. miRNA-offset RNAs (moRNAs) are similar in length to miRNAs, align to miRNA precursor (pre-miRNA) loci and are therefore believed to derive from processing of the pre-miRNA hairpin sequence. Recent next generation sequencing (NGS) studies have reported the presence of moRNAs in human neurons and cancer cells and in several tissues in mouse, including pluripotent stem cells. In order to gain additional knowledge about human moRNAs and their putative development-related expression, we applied NGS of small RNAs in human embryonic stem cells (hESCs) and fibroblasts. We found that certain moRNA isoforms are notably expressed in hESCs from loci coding for stem cell-selective or cancer-related miRNA clusters. In contrast, we observed only sparse moRNAs in fibroblasts. Consistent with earlier findings, most of the observed moRNAs derived from conserved loci and their expression did not appear to correlate with the expression of the adjacent miRNAs. We provide here the first report of moRNAs in hESCs, and their expression profile in comparison to fibroblasts. Moreover, we expand the repertoire of hESC miRNAs. These findings provide an expansion on the known repertoire of small non-coding RNA contents in hESCs.


Assuntos
Expressão Gênica , Células-Tronco Embrionárias Humanas/metabolismo , MicroRNAs/genética , Pequeno RNA não Traduzido/genética , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Biologia Computacional , Perfilação da Expressão Gênica , Biblioteca Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , MicroRNAs/química , Anotação de Sequência Molecular , Dados de Sequência Molecular , Pequeno RNA não Traduzido/química , Alinhamento de Sequência
11.
PLoS One ; 8(10): e76205, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098444

RESUMO

Correct interactions with extracellular matrix are essential to human pluripotent stem cells (hPSC) to maintain their pluripotent self-renewal capacity during in vitro culture. hPSCs secrete laminin 511/521, one of the most important functional basement membrane components, and they can be maintained on human laminin 511 and 521 in defined culture conditions. However, large-scale production of purified or recombinant laminin 511 and 521 is difficult and expensive. Here we have tested whether a commonly available human choriocarcinoma cell line, JAR, which produces high quantities of laminins, supports the growth of undifferentiated hPSCs. We were able to maintain several human pluripotent stem cell lines on decellularized matrix produced by JAR cells using a defined culture medium. The JAR matrix also supported targeted differentiation of the cells into neuronal and hepatic directions. Importantly, we were able to derive new human induced pluripotent stem cell (hiPSC) lines on JAR matrix and show that adhesion of the early hiPSC colonies to JAR matrix is more efficient than to matrigel. In summary, JAR matrix provides a cost-effective and easy-to-prepare alternative for human pluripotent stem cell culture and differentiation. In addition, this matrix is ideal for the efficient generation of new hiPSC lines.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes/citologia , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular , Coriocarcinoma/genética , Coriocarcinoma/metabolismo , Coriocarcinoma/patologia , Células-Tronco Embrionárias , Matriz Extracelular/metabolismo , Células Alimentadoras , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Laminina/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Células-Tronco Pluripotentes/metabolismo
12.
Mol Endocrinol ; 27(7): 1128-41, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23660593

RESUMO

The granulosa cells in the mammalian ovarian follicle respond to gonadotropin signaling and are involved in the processes of folliculogenesis and oocyte maturation. Studies on gene expression and regulation in human granulosa cells are of interest due to their potential for estimating the oocyte viability and in vitro fertilization success. However, the posttranscriptional gene expression studies on micro-RNA (miRNA) level in the human ovary have been scarce. The current study determined the miRNA profile by deep sequencing of the 2 intrafollicular somatic cell types: mural and cumulus granulosa cells (MGCs and CGCs, respectively) isolated from women undergoing controlled ovarian stimulation and in vitro fertilization. Altogether, 936 annotated and 9 novel miRNAs were identified. Ninety of the annotated miRNAs were differentially expressed between MGCs and CGCs. Bioinformatic prediction revealed that TGFß, ErbB signaling, and heparan sulfate biosynthesis were targeted by miRNAs in both granulosa cell populations, whereas extracellular matrix remodeling, Wnt, and neurotrophin signaling pathways were enriched among miRNA targets in MGCs. Two of the nine novel miRNAs found were of intronic origin: one from the aromatase and the other from the FSH receptor gene. The latter miRNA was predicted to target the activin signaling pathway. In addition to revealing the genome-wide miRNA signature in human granulosa cells, our results suggest that posttranscriptional regulation of gene expression by miRNAs could play an important role in the modification of gonadotropin signaling. miRNA expression studies could therefore lead to new prognostic markers in assisted reproductive technologies.


Assuntos
Aromatase/genética , Células da Granulosa/enzimologia , MicroRNAs/metabolismo , Receptores do FSH/genética , Análise de Sequência de RNA , Adulto , Aromatase/metabolismo , Sequência de Bases , Células do Cúmulo/metabolismo , Feminino , Perfilação da Expressão Gênica , Ontologia Genética , Humanos , MicroRNAs/genética , Anotação de Sequência Molecular , Dados de Sequência Molecular , Receptores do FSH/metabolismo , Alinhamento de Sequência , Transdução de Sinais/genética , Regulação para Cima/genética
13.
Stem Cell Res Ther ; 1(4): 33, 2010 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-20974014

RESUMO

Pluripotent stem cells are able to form any terminally differentiated cell. They have opened new doors for experimental and therapeutic studies to understand early development and to cure degenerative diseases in a way not previously possible. Nevertheless, it remains important to resolve and define the mechanisms underlying pluripotent stem cells, as that understanding will impact strongly on future medical applications. The capture of pluripotent stem cells in a dish is bound to several landmark discoveries, from the initial culture and phenotyping of pluripotent embryonal carcinoma cells to the recent induction of pluripotency in somatic cells. On this developmental time line, key transcription factors, such as Oct4, Sox2 or Nanog, have been revealed not only to regulate but also to functionally induce pluripotency. These early master regulators of development control developmental signalling pathways that affect the cell cycle, regulate gene expression, modulate the epigenetic state and repair DNA damage. Besides transcription factors, microRNAs have recently been shown to play important roles in gene expression and are embedded into the regulatory network orchestrating cellular development. However, there are species-specific differences in pluripotent cells, such as surface marker expression and growth factor requirements. Such differences and their underlying developmental pathways require clear definition and have major impacts on the preclinical test bed of pluripotent cells.


Assuntos
Ciclo Celular/genética , Reparo do DNA/genética , Regulação da Expressão Gênica/genética , Células-Tronco Pluripotentes/metabolismo , Animais , Diferenciação Celular , Reprogramação Celular , Dano ao DNA/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteína Homeobox Nanog , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição SOXB1/metabolismo
14.
Nat Biotechnol ; 28(4): 371-7, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20351689

RESUMO

Prolonged culture of human embryonic stem cells (hESCs) can lead to adaptation and the acquisition of chromosomal abnormalities, underscoring the need for rigorous genetic analysis of these cells. Here we report the highest-resolution study of hESCs to date using an Affymetrix SNP 6.0 array containing 906,600 probes for single nucleotide polymorphisms (SNPs) and 946,000 probes for copy number variations (CNVs). Analysis of 17 different hESC lines maintained in different laboratories identified 843 CNVs of 50 kb-3 Mb in size. We identified, on average, 24% of the loss of heterozygosity (LOH) sites and 66% of the CNVs changed in culture between early and late passages of the same lines. Thirty percent of the genes detected within CNV sites had altered expression compared to samples with normal copy number states, of which >44% were functionally linked to cancer. Furthermore, LOH of the q arm of chromosome 16, which has not been observed previously in hESCs, was detected.


Assuntos
Variações do Número de Cópias de DNA/genética , Análise Mutacional de DNA/métodos , DNA/genética , Células-Tronco Embrionárias/classificação , Células-Tronco Embrionárias/fisiologia , Variação Genética/genética , Análise de Sequência de DNA/métodos , Sequência de Bases , Técnicas de Cultura de Células/métodos , Humanos , Dados de Sequência Molecular
15.
Stem Cells ; 25(1): 197-202, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17008421

RESUMO

Human embryonic and mesenchymal stem cell therapies may offer significant benefit to a large number of patients. Recently, however, human embryonic stem cell lines cultured on mouse feeder cells were reported to be contaminated by the xeno-carbohydrate N-glycolylneuraminic acid (Neu5Gc) and considered potentially unfit for human therapy. To determine the extent of the problem of Neu5Gc contamination for the development of stem cell therapies, we investigated whether it also occurs in cells cultured on human feeder cells and in mesenchymal stem cells, what are the sources of contamination, and whether the contamination is reversible. We found that N-glycolylneuraminic acid was present in embryonic stem cells cultured on human feeder cells, correlating with the presence of Neu5Gc in components of the commercial serum replacement culture medium. Similar contamination occurred in mesenchymal stem cells cultured in the presence of fetal bovine serum. The results suggest that the Neu5Gc is present in both glycoprotein and lipid-linked glycans, as detected by mass spectrometric analysis and monoclonal antibody staining, respectively. Significantly, the contamination was largely reversible in the progeny of both cell types, suggesting that decontaminated cells may be derived from existing stem cell lines. Although major complications have not been reported in the clinical trials with mesenchymal stem cells exposed to fetal bovine serum, the immunogenic contamination may potentially be reflected in the viability and efficacy of the transplanted cells and thus bias the published results. Definition of safe culture conditions for stem cells is essential for future development of cellular therapies.


Assuntos
Antígenos Heterófilos/farmacologia , Células-Tronco Embrionárias/fisiologia , Células-Tronco Mesenquimais/fisiologia , Ácidos Neuramínicos/imunologia , Ácidos Neuramínicos/farmacologia , Anticorpos/farmacologia , Especificidade de Anticorpos , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Citometria de Fluxo , Homeostase , Humanos , Imuno-Histoquímica , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Fenótipo , Polissacarídeos/química , Polissacarídeos/isolamento & purificação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
16.
BMC Dev Biol ; 6: 40, 2006 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-16895598

RESUMO

BACKGROUND: Individual differences between human embryonic stem cell (hESC) lines are poorly understood. Here, we describe the derivation of five hESC lines (called FES 21, 22, 29, 30 and 61) from frozen-thawed human embryos and compare their individual differentiation characteristic. RESULTS: The cell lines were cultured either on human or mouse feeder cells. The cells grew significantly faster and could be passaged enzymatically only on mouse feeders. However, this was found to lead to chromosomal instability after prolonged culture. All hESC lines expressed the established markers of pluripotent cells as well as several primordial germ cell (PGC) marker genes in a uniform manner. However, the cell lines showed distinct features in their spontaneous differentiation patterns. The embryoid body (EB) formation frequency of FES 30 cell line was significantly lower than that of other lines and cells within the EBs differentiated less readily. Likewise, teratomas derived from FES 30 cells were constantly cystic and showed only minor solid tissue formation with a monotonous differentiation pattern as compared with the other lines. CONCLUSION: hESC lines may differ substantially in their differentiation properties although they appear similar in the undifferentiated state.


Assuntos
Diferenciação Celular , Linhagem Celular , Embrião de Mamíferos/citologia , Indução Embrionária/fisiologia , Células-Tronco/metabolismo , Animais , Biomarcadores/análise , Técnicas de Cultura de Células/métodos , Células Germinativas/metabolismo , Células Germinativas/ultraestrutura , Humanos , Cariotipagem , Masculino , Camundongos , Células-Tronco/ultraestrutura , Teratoma/patologia
17.
Reproduction ; 131(4): 641-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16595715

RESUMO

The receptor tyrosine c-Kit and its cognate ligand, c-Kit ligand (KL, stem cell factor, SCF), are involved in ovarian follicular development in several animal species. We studied the expression of KL and c-Kit using in situ hybridization and immunohistochemistry in donated human ovarian cortical tissue. The KL transcripts were expressed in granulosa cells of primary follicles, whereas the expression of c-Kit was confined to the oocyte and granulosa cells in primary and secondary follicles. We employed an ovarian organ culture using firstly serum-containing and then serum-free medium to study the effects of KL and an anti-c-Kit antibody, ACK2, on the development and survival of ovarian follicles in vitro. Culture of ovarian cortical slices for 7 days resulted in a 37% increase in the number of primary follicles and a 6% increase in secondary follicles. The proportion of viable follicles decreased in all cultures. The addition of KL (1, 10 and 100 ng/ml) into the culture media did not affect the developmental stages of the follicles or the proportion of atretic follicles. Inclusion of ACK2 (800 ng/ml) in the culture medium significantly increased the proportion of atretic follicles on days 7 (49 vs 28% in control cultures) and 14 (62 vs 38%) of culture. In conclusion, c-Kit and KL are expressed in human ovaries during follicular development. Blocking the c-Kit receptor induces follicular atresia. The KL/c-Kit signaling system is likely to control the survival of human ovarian follicles during early follicular development.


Assuntos
Folículo Ovariano/metabolismo , Proteínas Proto-Oncogênicas c-kit/análise , Fator de Células-Tronco/análise , Anticorpos Monoclonais/farmacologia , Northern Blotting/métodos , Feminino , Atresia Folicular , Humanos , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Técnicas de Cultura de Órgãos , Proteínas Proto-Oncogênicas c-kit/imunologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fator de Células-Tronco/metabolismo
18.
Hum Reprod ; 20(6): 1534-40, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15695312

RESUMO

BACKGROUND: The purpose has been to develop an in vitro oocyte maturation (IVM) method for a wide range of patients. METHODS: A total of 239 cycles with immature oocyte retrieval (IOC) were carried out without hormonal priming. Patients with regular cycles and normal or polycystic ovaries (PCO) and anovulatory cycles with PCOS were included. Insemination or intracytoplasmic sperm injection (ICSI) according to sperm quality was alternatively used in fertilization of the matured oocytes. RESULTS: A total of 971 immature oocytes (mean 8.0 +/- 5.2) were collected in 122 IVM-IVF cycles and 851 oocytes (mean 7.3 +/- 4.4) in 117 IVM-ICSI cycles. The oocyte maturation and fertilization rate was 62.6% and 37.7% after insemination, and 53.9% and 69.3% after ICSI, respectively. The mean number of embryos transferred was 1.6. Clinical pregnancy rate per IOC was 23.8% in IVM-IVF and 17.1% in IVM-ICSI (ns). Implantation rate was higher in IVM-IVF (24.2%) than in IVM-ICSI (14.8%) (P < 0.05). CONCLUSIONS: Insemination of IVM oocytes functions well, resulting in comparable pregnancy rates per IOC between IVM-IVF and IVM-ICSI. Good pregnancy results can be achieved both in patients with regular cycles and with PCO(S) by transferring only one or two embryos at a time.


Assuntos
Fertilização in vitro/métodos , Oócitos/fisiologia , Taxa de Gravidez , Células Cultivadas , Feminino , Humanos , Recém-Nascido , Infertilidade Feminina/terapia , Ovário/fisiologia , Indução da Ovulação/métodos , Síndrome do Ovário Policístico/complicações , Gravidez , Resultado da Gravidez , Valores de Referência
19.
J Assist Reprod Genet ; 21(8): 301-6, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15568331

RESUMO

PURPOSE: To test the effect of cyclic adenosine 3', 5'-monophosphate (cAMP) on early human ovarian follicles during prolonged culture period. METHODS: Donated ovarian biopsies from 16 women undergoing gynecological laparoscopy were cut into slices and cultured in parallel for 1, 2, or 3 weeks in the presence and the absence of 0.5 mM 8-bromo-cAMP. The developmental stages, sizes, and viability of the follicles were recorded from histological sections of all samples. RESULTS: On day 14, cortical slices cultured with 8-bromo-cAMP showed a significantly higher proportion of secondary follicles (50.0% vs. 20.0%) and a lower proportion of primordial follicles (9.7% vs. 26.7%) when compared with those cultured without 8-bromo-cAMP. On day 21, the proportion of viable follicles in cortical slices with 8-bromo-cAMP treatment was significantly higher than that without 8-bromo-cAMP treatment (79.6% vs. 55.2%). CONCLUSION: CyclicAMP promoted folliculogenesis and follicle survival during 14-21 days' culture of human ovarian cortical slices.


Assuntos
8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Folículo Ovariano/citologia , Técnicas de Cultura de Tecidos/métodos , Feminino , Humanos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/crescimento & desenvolvimento
20.
Fertil Steril ; 82 Suppl 3: 1077-85, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15474077

RESUMO

OBJECTIVE: To investigate the effects of T, dihydrotestosterone (DHT), and 17beta-estradiol on human ovarian stromal tissue survival in culture and to identify steroids capable of inhibiting cell death in vitro. DESIGN: Prospective study. SETTING: Academic research setting. PATIENT(S): Thirty women, aged 18-38 years, undergoing gynecological operations for benign conditions and eight women, aged 27-36 years, undergoing IVF because of tubal obstruction or male factor infertility. INTERVENTION(S): Cultured tissue was exposed to T, DHT, 17beta-estradiol, or the anti-androgen casodex. MAIN OUTCOME MEASURE(S): Immunohistochemistry for androgen receptor (AR), Southern blot analysis of DNA fragmentation, histology, and in situ end labeling of apoptotic DNA. RESULT(S): Androgen receptors were detected in the ovarian stroma and granulosa cells of the primordial follicles, although they were more clearly seen in primary follicles and more advanced-stage follicles. Testosterone only marginally suppressed ovarian tissue apoptosis in vitro. DHT was more effective than T, whereas 17beta-estradiol had no notable effect on the viability of the tissue. The effects of androgens on the ovarian tissue may be mediated through ARs, since blocking the receptors with an AR antagonist reversed the suppressive effect of DHT. CONCLUSION(S): DHT may be useful for enhancing human ovarian tissue survival in vitro.


Assuntos
Di-Hidrotestosterona/farmacologia , Estradiol/farmacologia , Ovário/efeitos dos fármacos , Ovário/fisiologia , Testosterona/farmacologia , Adulto , Antagonistas de Androgênios/farmacologia , Antagonistas de Receptores de Andrógenos , Anilidas/farmacologia , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cultura , Feminino , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/fisiologia , Humanos , Nitrilas , Ovário/metabolismo , Estudos Prospectivos , Receptores Androgênicos/metabolismo , Sobrevivência de Tecidos/efeitos dos fármacos , Compostos de Tosil
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA