Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Adv Healthc Mater ; : e2400501, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38817106

RESUMO

In response to the increasing demand for spheroid-based cancer research, the importance of developing integrated platforms that can simultaneously facilitate high-throughput spheroid production and multiplexed analysis is emphasized. In addition, the understanding of how the size and cellular composition of tumors directly influence their internal structures and functionalities underlines the critical need to produce spheroids of diverse sizes and compositions on a large scale. To address this rising demand, this work presents a configurable and linkable in vitro three-dimensional (3D) cell culture kit (CLiCK) for spheroids, termed CLiCK-Spheroid. This platform consists of three primary components: a hanging drop microarray (HDMA), a concave pillar microarray (CPMA), and gradient blocks. The HDMA alone produces a homogeneous spheroid array, while its combination with the gradient block enables one-step generation of a size-gradient spheroid array. Using the size-gradient spheroid arrays, the occurrence of necrotic cores based on spheroid size is demonstrated. Additionally, spheroids in a single batch can be conveniently compartmentalized and regrouped using a CPMA, enhancing the versatility of spheroid arrays and enabling multiplexed drug treatments. By combining the different assembly methods, this work has achieved high-throughput production of cell composition-gradient spheroid arrays, with noticeable variations in morphology and vascularization based on cell compositions.

2.
Immunity ; 56(9): 2105-2120.e13, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37527657

RESUMO

Childhood neglect and/or abuse can induce mental health conditions with unknown mechanisms. Here, we identified stress hormones as strong inducers of astrocyte-mediated synapse phagocytosis. Using in vitro, in vivo, and human brain organoid experiments, we showed that stress hormones increased the expression of the Mertk phagocytic receptor in astrocytes through glucocorticoid receptor (GR). In post-natal mice, exposure to early social deprivation (ESD) specifically activated the GR-MERTK pathway in astrocytes, but not in microglia. The excitatory post-synaptic density in cortical regions was reduced in ESD mice, and there was an increase in the astrocytic engulfment of these synapses. The loss of excitatory synapses, abnormal neuronal network activities, and behavioral abnormalities in ESD mice were largely prevented by ablating GR or MERTK in astrocytes. Our work reveals the critical roles of astrocytic GR-MERTK activation in evoking stress-induced abnormal behaviors in mice, suggesting GR-MERTK signaling as a therapeutic target for stress-induced mental health conditions.


Assuntos
Astrócitos , Fagocitose , Estresse Psicológico , Animais , Criança , Humanos , Camundongos , Astrócitos/metabolismo , c-Mer Tirosina Quinase/genética , Hormônios/metabolismo , Sinapses/metabolismo , Estresse Psicológico/metabolismo
3.
Mol Ther ; 31(4): 1002-1016, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-36755495

RESUMO

Fabry disease (FD), a lysosomal storage disorder, is caused by defective α-galactosidase (GLA) activity, which results in the accumulation of globotriaosylceramide (Gb3) in endothelial cells and leads to life-threatening complications such as left ventricular hypertrophy (LVH), renal failure, and stroke. Enzyme replacement therapy (ERT) results in Gb3 clearance; however, because of a short half-life in the body and the high immunogenicity of FD patients, ERT has a limited therapeutic effect, particularly in patients with late-onset disease or progressive complications. Because vascular endothelial cells (VECs) derived from FD-induced pluripotent stem cells display increased thrombospondin-1 (TSP1) expression and enhanced SMAD2 signaling, we screened for chemical compounds that could downregulate TSP1 and SMAD2 signaling. Fasudil reduced the levels of p-SMAD2 and TSP1 in FD-VECs and increased the expression of angiogenic factors. Furthermore, fasudil downregulated the endothelial-to-mesenchymal transition (EndMT) and mitochondrial function of FD-VECs. Oral administration of fasudil to FD mice alleviated several FD phenotypes, including LVH, renal fibrosis, anhidrosis, and heat insensitivity. Our findings demonstrate that fasudil is a novel candidate for FD therapy.


Assuntos
Doença de Fabry , Animais , Camundongos , Doença de Fabry/tratamento farmacológico , Doença de Fabry/genética , Células Endoteliais/metabolismo , alfa-Galactosidase/genética , Fenótipo , Terapia de Reposição de Enzimas
4.
Stem Cell Res ; 66: 103001, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36516658

RESUMO

Fabry disease (FD) is a lysosomal storage disorder caused by mutations in GLA gene. Here, GLA mutation (1268fs*1 (c.803_806del)) of FD iPSCs was corrected using the CRISPR-Cas9 gene editing system. The corrected (cor) FD-iPSCs retained normal morphology, karyotype, expression of pluripotency-associated markers, trilineage differentiation potential, and GLA activity. Thus, FD(cor)-iPSCs can be used as valuable tools to study the mechanism how GLA mutation1268fs*1 induces various pathophysiologic phenotypes in FD patients.


Assuntos
Doença de Fabry , Células-Tronco Pluripotentes Induzidas , Humanos , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo , Doença de Fabry/genética , Sistemas CRISPR-Cas/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação/genética
5.
Stem Cell Reports ; 16(8): 1985-1998, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34242618

RESUMO

Costello syndrome (CS) is an autosomal dominant disorder caused by mutations in HRAS. Although CS patients have skeletal abnormalities, the role of mutated HRAS in bone development remains unclear. Here, we use CS induced pluripotent stem cells (iPSCs) undergoing osteogenic differentiation to investigate how dysregulation of extracellular matrix (ECM) remodeling proteins contributes to impaired osteogenesis. Although CS patient-derived iPSCs develop normally to produce mesenchymal stem cells (MSCs), the resulting CS MSCs show defective osteogenesis with reduced alkaline phosphatase activity and lower levels of bone mineralization. We found that hyperactivation of SMAD3 signaling during the osteogenic differentiation of CS MSCs leads to aberrant expression of ECM remodeling proteins such as MMP13, TIMP1, and TIMP2. CS MSCs undergoing osteogenic differentiation also show reduced ß-catenin signaling. Knockdown of TIMPs permits normal differentiation of CS MSCs into osteoblasts and enhances ß-catenin signaling in a RUNX2-independent manner. Thus, this study demonstrates that enhanced TIMP expression induced by hyperactivated SMAD3 signaling impairs the osteogenic development of CS MSCs via an inactivation of ß-catenin signaling.


Assuntos
Diferenciação Celular/genética , Síndrome de Costello/genética , Proteínas da Matriz Extracelular/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese/genética , Fosfatase Alcalina/metabolismo , Calcificação Fisiológica/genética , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Síndrome de Costello/metabolismo , Síndrome de Costello/patologia , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Humanos , Osteoblastos/citologia , Osteoblastos/metabolismo , Transdução de Sinais/genética , Proteína Smad3/genética , Proteína Smad3/metabolismo , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
6.
J Med Genet ; 58(11): 767-777, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33051312

RESUMO

BACKGROUND: ARID2 belongs to the Switch/sucrose non-fermenting complex, in which the genetic defects have been found in patients with dysmorphism, short stature and intellectual disability (ID). As the phenotypes of patients with ARID2 mutations partially overlap with those of RASopathy, this study evaluated the biochemical association between ARID2 and RAS-MAPK pathway. METHODS: The phenotypes of 22 patients with either an ARID2 heterozygous mutation or haploinsufficiency were reviewed. Comprehensive molecular analyses were performed using somatic and induced pluripotent stem cells (iPSCs) of a patient with ARID2 haploinsufficiency as well as using the mouse model of Arid2 haploinsufficiency by CRISPR/Cas9 gene editing. RESULTS: The phenotypic characteristics of ARID2 deficiency include RASopathy, Coffin-Lowy syndrome or Coffin-Siris syndrome or undefined syndromic ID. Transient ARID2 knockout HeLa cells using an shRNA increased ERK1 and ERK2 phosphorylation. Impaired neuronal differentiation with enhanced RAS-MAPK activity was observed in patient-iPSCs. In addition, Arid2 haploinsufficient mice exhibited reduced body size and learning/memory deficit. ARID2 haploinsufficiency was associated with reduced IFITM1 expression, which interacts with caveolin-1 (CAV-1) and inhibits ERK activation. DISCUSSION: ARID2 haploinsufficiency is associated with enhanced RAS-MAPK activity, leading to reduced IFITM1 and CAV-1 expression, thereby increasing ERK activity. This altered interaction might lead to abnormal neuronal development and a short stature.


Assuntos
Nanismo/genética , Deficiência Intelectual/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Fatores de Transcrição/genética , Anormalidades Múltiplas/etiologia , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Encéfalo/anormalidades , Encéfalo/fisiopatologia , Caveolina 1/genética , Caveolina 1/metabolismo , Criança , Pré-Escolar , Face/anormalidades , Feminino , Deformidades Congênitas da Mão/etiologia , Haploinsuficiência , Heterozigoto , Humanos , Deficiência Intelectual/etiologia , Masculino , Camundongos Knockout , Micrognatismo/etiologia , Mutação , Pescoço/anormalidades , Fatores de Transcrição/metabolismo , Adulto Jovem , Proteínas ras/genética , Proteínas ras/metabolismo
7.
EBioMedicine ; 52: 102633, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31981984

RESUMO

BACKGROUND: Fabry disease (FD) is a recessive X-linked lysosomal storage disorder caused by α-galactosidase A (GLA) deficiency. Although the mechanism is unclear, GLA deficiency causes an accumulation of globotriaosylceramide (Gb3), leading to vasculopathy. METHODS: To explore the relationship between the accumulation of Gb3 and vasculopathy, induced pluripotent stem cells generated from four Fabry patients (FD-iPSCs) were differentiated into vascular endothelial cells (VECs). Genome editing using CRISPR-Cas9 system was carried out to correct the GLA mutation or to delete Thrombospondin-1 (TSP-1). Global transcriptomes were compared between wild-type (WT)- and FD-VECs by RNA-sequencing analysis. FINDINGS: Here, we report that overexpression of TSP-1 contributes to the dysfunction of VECs in FD. VECs originating from FD-iPSCs (FD-VECs) showed aberrant angiogenic functionality even upon treatment with recombinant α-galactosidase. Intriguingly, FD-VECs produced more p-SMAD2 and TSP-1 than WT-VECs. We also found elevated TSP-1 in the peritubular capillaries of renal tissues biopsied from FD patients. Inhibition of SMAD2 signaling or knock out of TSP-1 (TSP-1-/-) rescues normal vascular functionality in FD-VECs, like in gene-corrected FD-VECs. In addition, the enhanced oxygen consumption rate is reduced in TSP-1-/- FD-VECs. INTERPRETATION: The overexpression of TSP-1 secondary to Gb3 accumulation is primarily responsible for the observed FD-VEC dysfunction. Our findings implicate dysfunctional VEC angiogenesis in the peritubular capillaries in some of the complications of Fabry disease. FUNDING: This study was supported by grant 2018M3A9H1078330 from the National Research Foundation of the Republic of Korea.


Assuntos
Células Endoteliais/metabolismo , Doença de Fabry/genética , Doença de Fabry/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Trombospondina 1/genética , Adulto , Alelos , Animais , Sistemas CRISPR-Cas , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular , Ativação Enzimática , Doença de Fabry/diagnóstico , Edição de Genes , Expressão Gênica , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Imuno-Histoquímica , Imunofenotipagem , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Modelos Biológicos , Mutação , Estresse Oxidativo , Fenótipo , Trombospondina 1/metabolismo , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo
8.
ACS Biomater Sci Eng ; 6(2): 813-821, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33464850

RESUMO

Human iPSC-derived mesenchymal stem cells (iMSCs) are an alternative to primary mesenchymal stem cells (MSCs), which have been a limited supply, and have attracted a great deal of interest as a promising cell source in cell-based therapy. However, despite their enormous therapeutic potential, it has been difficult to translate this potential into clinical applications due to the short viability duration of transplanted iMSCs. Therefore, to maximize the therapeutic effects of iMSCs, it is extremely important to extend their retention rate during and even after the transplantation. In this study, we developed a new extracellular matrix (ECM)-coating method involving the mild reduction of the cell surface. The reduction of disulfide bonds around the cell membrane enhanced the coating efficiency without a decrease in the viability and differentiation potential of iMSCs. We then induced ECM-coated single iMSCs to form three-dimensional spheroids via self-assembly of the aggregates within a physically confined microenvironment. The spheroids exhibited longer maintenance of the survival rate. Nanometric ECM coating of the cell membrane is a new approach as a key for resolving the conventional challenges of cell-based therapy.


Assuntos
Matriz Extracelular , Células-Tronco Pluripotentes Induzidas , Células-Tronco Mesenquimais , Diferenciação Celular , Humanos , Fosfinas
9.
Int J Mol Sci ; 18(12)2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29194391

RESUMO

Cardiofaciocutaneous (CFC) syndrome is a rare genetic disorder caused by mutations in the extracellular signal-regulated kinase (ERK) signaling. However, little is known about how aberrant ERK signaling is associated with the defective bone development manifested in most CFC syndrome patients. In this study, induced pluripotent stem cells (iPSCs) were generated from dermal fibroblasts of a CFC syndrome patient having rapidly accelerated fibrosarcoma kinase B (BRAF) gain-of-function mutation. CFC-iPSCs were differentiated into mesenchymal stem cells (CFC-MSCs) and further induced to osteoblasts in vitro. The osteogenic defects of CFC-MSCs were revealed by alkaline phosphatase activity assay, mineralization assay, quantitative real-time polymerase chain reaction (qRT-PCR), and western blotting. Osteogenesis of CFC-MSCs was attenuated compared to wild-type (WT)-MSCs. In addition to activated ERK signaling, increased p-SMAD2 and decreased p-SMAD1 were observed in CFC-MSCs during osteogenesis. The defective osteogenesis of CFC-MSCs was rescued by inhibition of ERK signaling and SMAD2 signaling or activation of SMAD1 signaling. Importantly, activation of ERK signaling and SMAD2 signaling or inhibition of SMAD1 signaling recapitulated the impaired osteogenesis in WT-MSCs. Our findings indicate that SMAD2 signaling and SMAD1 signaling as well as ERK signaling are responsible for defective early bone development in CFC syndrome, providing a novel insight on the pathological mechanism and therapeutic targets.


Assuntos
Displasia Ectodérmica/patologia , Insuficiência de Crescimento/patologia , Cardiopatias Congênitas/patologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Proteínas Proto-Oncogênicas B-raf/genética , Animais , Diferenciação Celular , Linhagem Celular , Displasia Ectodérmica/genética , Displasia Ectodérmica/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fácies , Insuficiência de Crescimento/genética , Insuficiência de Crescimento/metabolismo , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Mutação , Osteoblastos/metabolismo , Osteogênese , Fosforilação , Transdução de Sinais , Proteína Smad1/metabolismo , Proteína Smad2/metabolismo
10.
Sci Rep ; 6: 35145, 2016 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-27731367

RESUMO

Insulin secretion is elaborately modulated in pancreatic ß cells within islets of three-dimensional (3D) structures. Using human pluripotent stem cells (hPSCs) to develop islet-like structures with insulin-producing ß cells for the treatment of diabetes is challenging. Here, we report that pancreatic islet-like clusters derived from hESCs are functionally capable of glucose-responsive insulin secretion as well as therapeutic effects. Pancreatic hormone-expressing endocrine cells (ECs) were differentiated from hESCs using a step-wise protocol. The hESC-derived ECs expressed pancreatic endocrine hormones, such as insulin, somatostatin, and pancreatic polypeptide. Notably, dissociated ECs autonomously aggregated to form islet-like, 3D structures of consistent sizes (100-150 µm in diameter). These EC clusters (ECCs) enhanced insulin secretion in response to glucose stimulus and potassium channel inhibition in vitro. Furthermore, ß cell-deficient mice transplanted with ECCs survived for more than 40 d while retaining a normal blood glucose level to some extent. The expression of pancreatic endocrine hormones was observed in tissues transplanted with ECCs. In addition, ECCs could be generated from human induced pluripotent stem cells. These results suggest that hPSC-derived, islet-like clusters may be alternative therapeutic cell sources for treating diabetes.


Assuntos
Glucose/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Glicemia/metabolismo , Agregação Celular , Diferenciação Celular , Células Cultivadas , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/terapia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Transplante das Ilhotas Pancreáticas , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Organoides/citologia , Organoides/metabolismo
11.
Stem Cells Dev ; 25(8): 636-47, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26914390

RESUMO

Citrin deficiency (CD) is a recessive genetic disorder caused by mutations in the citrin gene SLC25A13. CD causes various symptoms related to nutrient metabolism such as urea cycle failure, abnormal amino acid levels, and fatty liver. To understand the pathophysiology of CD, the molecular phenotypes were investigated using induced pluripotent stem cells derived from fibroblasts of CD patient (CD-iPSCs). In this study, we demonstrate that aberrant mitochondrial ß-oxidation may lead to fatty liver in CD patients. CD-iPSCs normally differentiated into hepatocytes, similar to wild-type iPSCs (WT-iPSCs). However, hepatocytes derived from CD-iPSCs (CD-HLCs) did not exhibit ureogenesis. Cellular triglyceride and lipid granule levels were significantly increased in CD-HLCs compared with WT-HLCs. Peroxisome proliferator-activated receptor-α (PPAR-α) and its target genes which are involved in mitochondrial ß-oxidation were downregulated in CD-HLCs, and treatment with a PPAR-α agonist partially reduced the lipid accumulation in CD-HLCs. In addition, the mitochondria in CD-HLCs exhibited abnormal morphologies. Based on these observations, we conclude that the lipid accumulation in CD-HLCs results from dysfunctional mitochondrial ß-oxidation and abnormal mitochondrial structure.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Hepatócitos/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Mitocôndrias Hepáticas/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Humanos , Metabolismo dos Lipídeos , Camundongos Nus , Oxirredução
12.
Stem Cell Res Ther ; 6: 160, 2015 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-26347346

RESUMO

INTRODUCTION: Bone abnormalities, one of the primary manifestations of Menkes disease (MD), include a weakened bone matrix and low mineral density. However, the molecular and cellular mechanisms underlying these bone defects are poorly understood. METHODS: We present in vitro modeling for impaired osteogenesis in MD using human induced pluripotent stem cells (iPSCs) with a mutated ATP7A gene. MD-iPSC lines were generated from two patients harboring different mutations. RESULTS: The MD-iPSCs showed a remarkable retardation in CD105 expression with morphological anomalies during development to mesenchymal stem cells (MSCs) compared with wild-type (WT)-iPSCs. Interestingly, although prolonged culture enhanced CD105 expression, mature MD-MSCs presented with low alkaline phosphatase activity, reduced calcium deposition in the extracellular matrix, and downregulated osteoblast-specific genes during osteoblast differentiation in vitro. Knockdown of ATP7A also impaired osteogenesis in WT-MSCs. Lysyl oxidase activity was also decreased in MD-MSCs during osteoblast differentiation. CONCLUSIONS: Our findings indicate that ATP7A dysfunction contributes to retardation in MSC development and impairs osteogenesis in MD.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Síndrome dos Cabelos Torcidos/metabolismo , Osteogênese , Adenosina Trifosfatases/genética , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Proteínas de Transporte de Cátions/genética , Células Cultivadas , ATPases Transportadoras de Cobre , Endoglina , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/transplante , Lactente , Recém-Nascido , Síndrome dos Cabelos Torcidos/genética , Síndrome dos Cabelos Torcidos/patologia , Camundongos , Mutação , Proteína-Lisina 6-Oxidase/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo
13.
PLoS One ; 10(7): e0132992, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26177506

RESUMO

Human pluripotent stem cell-derived hepatocytes have the potential to provide in vitro model systems for drug discovery and hepatotoxicity testing. However, these cells are currently unsuitable for drug toxicity and efficacy testing because of their limited expression of genes encoding drug-metabolizing enzymes, especially cytochrome P450 (CYP) enzymes. Transcript levels of major CYP genes were much lower in human embryonic stem cell-derived hepatocytes (hESC-Hep) than in human primary hepatocytes (hPH). To verify the mechanism underlying this reduced expression of CYP genes, including CYP1A1, CYP1A2, CYP1B1, CYP2D6, and CYP2E1, we investigated their epigenetic regulation in terms of DNA methylation and histone modifications in hESC-Hep and hPH. CpG islands of CYP genes were hypermethylated in hESC-Hep, whereas they had an open chromatin structure, as represented by hypomethylation of CpG sites and permissive histone modifications, in hPH. Inhibition of DNA methyltransferases (DNMTs) during hepatic maturation induced demethylation of the CpG sites of CYP1A1 and CYP1A2, leading to the up-regulation of their transcription. Combinatorial inhibition of DNMTs and histone deacetylases (HDACs) increased the transcript levels of CYP1A1, CYP1A2, CYP1B1, and CYP2D6. Our findings suggest that limited expression of CYP genes in hESC-Hep is modulated by epigenetic regulatory factors such as DNMTs and HDACs.


Assuntos
Metilases de Modificação do DNA/genética , Epigênese Genética , Hepatócitos/metabolismo , Histona Desacetilases/genética , Processamento de Proteína Pós-Traducional , Diferenciação Celular , Linhagem Celular , Cromatina/metabolismo , Cromatina/ultraestrutura , Ilhas de CpG , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP1B1/genética , Citocromo P-450 CYP1B1/metabolismo , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP2E1/genética , Citocromo P-450 CYP2E1/metabolismo , Metilação de DNA , Metilases de Modificação do DNA/metabolismo , Hepatócitos/citologia , Histona Desacetilases/metabolismo , Histonas/genética , Histonas/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Especificidade de Órgãos , Cultura Primária de Células , Transdução de Sinais , Transcrição Gênica
14.
Stem Cells ; 33(5): 1447-55, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25639853

RESUMO

Cardio-facio-cutaneous (CFC) syndrome is a developmental disorder caused by constitutively active ERK signaling manifesting mainly from BRAF mutations. Little is known about the role of elevated ERK signaling in CFC syndrome during early development. Here, we show that both SMAD1 and ERK signaling pathways may contribute to the developmental defects in CFC syndrome. Induced pluripotent stem cells (iPSCs) derived from dermal fibroblasts of a CFC syndrome patient (CFC-iPSCs) revealed early developmental defects in embryoid body (EB) development, ß-catenin localization, and neuronal differentiation. Both SMAD1 and ERK signalings were significantly activated in CFC-iPSCs during EB formation. Most of the ß-catenin was dissociated from the membrane and preferentially localized into the nucleus in CFC-EBs. Furthermore, activation of SMAD1 signaling recapitulated early developmental defects in wild-type iPSCs. Intriguingly, inhibition of SMAD1 signaling in CFC-iPSCs rescued aberrant EB morphology, impaired neuronal differentiation, and altered ß-catenin localization. These results suggest that SMAD1 signaling may be a key pathway contributing the pathogenesis of CFC syndrome during early development.


Assuntos
Displasia Ectodérmica/metabolismo , Displasia Ectodérmica/patologia , Insuficiência de Crescimento/metabolismo , Insuficiência de Crescimento/patologia , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Corpos Embrioides/metabolismo , Fácies , Humanos , Masculino , Neurônios/patologia , Transporte Proteico , beta Catenina/metabolismo
15.
Cell Stem Cell ; 15(6): 735-49, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25479749

RESUMO

LIN28-mediated processing of the microRNA (miRNA) let-7 has emerged as a multilevel program that controls self-renewal in embryonic stem cells. LIN28A is believed to act primarily in the cytoplasm together with TUT4/7 to prevent final maturation of let-7 by Dicer, whereas LIN28B has been suggested to preferentially act on nuclear processing of let-7. Here, we find that SET7/9 monomethylation in a putative nucleolar localization region of LIN28A increases its nuclear retention and protein stability. In the nucleoli of human embryonic stem cells, methylated LIN28A sequesters pri-let-7 and blocks its processing independently of TUT4/7. The nuclear form of LIN28A regulates transcriptional changes in MYC-pathway targets, thereby maintaining stemness programs and inhibiting expression of early lineage-specific markers. These findings provide insight into the molecular mechanism underlying the posttranslational methylation of nuclear LIN28A and its ability to modulate pluripotency by repressing let-7 miRNA expression in human embryonic stem cells.


Assuntos
Nucléolo Celular/metabolismo , Células-Tronco Embrionárias/fisiologia , MicroRNAs/metabolismo , Células-Tronco Pluripotentes/fisiologia , Proteínas de Ligação a RNA/metabolismo , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Proteínas de Ligação a DNA/metabolismo , Genes myc/fisiologia , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Metilação , MicroRNAs/genética , Multimerização Proteica , Transporte Proteico , Proteínas de Ligação a RNA/genética
16.
PLoS One ; 9(4): e94888, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24728509

RESUMO

OBJECTIVES: Kidney disease is emerging as a critical medical problem worldwide. Because of limited treatment options for the damaged kidney, stem cell treatment is becoming an alternative therapeutic approach. Of many possible human stem cell sources, pluripotent stem cells are most attractive due to their self-renewal and pluripotent capacity. However, little is known about the derivation of renal lineage cells from human pluripotent stem cells (hPSCs). In this study, we developed a novel protocol for differentiation of nephron progenitor cells (NPCs) from hPSCs in a serum- and feeder-free system. MATERIALS AND METHODS: We designed step-wise protocols for differentiation of human pluripotent stem cells toward primitive streak, intermediate mesoderm and NPCs by recapitulating normal nephrogenesis. Expression of key marker genes was examined by RT-PCR, real time RT-PCR and immunocytochemistry. Each experiment was independently performed three times to confirm its reproducibility. RESULTS: After modification of culture period and concentration of exogenous factors, hPSCs can differentiate into NPCs that markedly express specific marker genes such as SIX2, GDNF, HOXD11, WT1 and CITED1 in addition to OSR1, PAX2, SALL1 and EYA1. Moreover, NPCs possess the potential of bidirectional differentiation into both renal tubular epithelial cells and glomerular podocytes in defined culture conditions. In particular, approximately 70% of SYN-positive cells were obtained from hPSC-derived NPCs after podocytes induction. NPCs can also form in vitro tubule-like structures in three dimensional culture systems. CONCLUSIONS: Our novel protocol for hPSCs differentiation into NPCs can be useful for producing alternative sources of cell replacement therapy and disease modeling for human kidney diseases.


Assuntos
Técnicas de Cultura de Células , Diferenciação Celular , Néfrons/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco/citologia , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células Epiteliais/citologia , Humanos , Células-Tronco Pluripotentes Induzidas , Túbulos Renais/citologia , Mesoderma/citologia , Podócitos/citologia , Linha Primitiva/citologia
17.
Biochem Biophys Res Commun ; 444(3): 311-8, 2014 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-24468087

RESUMO

Menkes disease (MD) is a copper-deficient neurodegenerative disorder that manifests severe neurologic symptoms such as seizures, lethargic states, and hypotonia. Menkes disease is due to a dysfunction of ATP7A, but the pathophysiology of neurologic manifestation is poorly understood during embryonic development. To understand the pathophysiology of neurologic symptoms, molecular and cellular phenotypes were investigated in Menkes disease-derived induced pluripotent stem cells (MD-iPSCs). MD-iPSCs were generated from fibroblasts of a Menkes disease patient. Abnormal reticular distribution of ATP7A was observed in MD-fibroblasts and MD-iPSCs, respectively. MD-iPSCs showed abnormal morphology in appearance during embryoid body (EB) formation as compared with wild type (WT)-iPSCs. Intriguingly, aberrant switch of E-cadherin (E-cad) to N-cadherin (N-cad) and impaired neural rosette formation were shown in MD-iPSCs during early differentiation. When extracellular copper was chelated in WT-iPSCs by treatment with bathocuprione sulfate, aberrant switch of E-cad to N-cad and impaired neuronal differentiation were observed, like in MD-iPSCs. Our results suggest that neurological defects in Menkes disease patients may be responsible for aberrant cadherin transition and impaired neuronal differentiation during early developmental stage.


Assuntos
Síndrome dos Cabelos Torcidos/fisiopatologia , Modelos Biológicos , Células-Tronco Pluripotentes/citologia , Adenosina Trifosfatases/genética , Sequência de Bases , Caderinas/metabolismo , Proteínas de Transporte de Cátions/genética , Diferenciação Celular , Células Cultivadas , Pré-Escolar , Cobre/metabolismo , ATPases Transportadoras de Cobre , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Neurônios/patologia
18.
FEBS J ; 281(4): 1029-45, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24341592

RESUMO

Although octamer-binding transcription factor 4 (Oct-4) is one of the most intensively studied factors in mammalian development, no cellular genes capable of replacing Oct-4 function in embryonic stem (ES) cells have been found. Recent data show that nuclear receptor subfamily 5, group A, member 2 (Nr5a2) is able to replace Oct-4 function in the reprogramming process; however, it is unclear whether Nr5a2 can replace Oct-4 function in ES cells. In this study, the ability of Nr5a2 to maintain self-renewal and pluripotency in ES cells was investigated. Nr5a2 localized to the nucleus in ES cells, similarly to Oct-4. However, expression of Nr5a2 failed to rescue the stem cell phenotype or to maintain the self-renewal ability of ES cells. Furthermore, as compared with Oct-4-expressing ES cells, Nr5a2-expressing ES cells showed a reduced number of cells in S-phase, did not expand normally, and did not remain in an undifferentiated state. Ectopic expression of Nr5a2 in ES cells was not able to activate transcription of ES cell-specific genes, and gene expression profiling demonstrated differences between Nr5a2-expressing and Oct-4-expressing ES cells. In addition, Nr5a2-expressing ES cells were not able to form teratomas in nude mice. Taken together, these results strongly suggest that the gene regulation properties of Nr5a2 and Oct-4 and their abilities to confer self-renewal and pluripotency of ES cells differ. The present study provides strong evidence that Nr5a2 cannot replace Oct-4 function in ES cells.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiologia , Imuno-Histoquímica , Camundongos , Camundongos Nus , Fator 3 de Transcrição de Octâmero/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores Citoplasmáticos e Nucleares/genética
19.
Hum Mol Genet ; 23(7): 1802-16, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24218367

RESUMO

The extensive molecular characterization of human pluripotent stem cells (hPSCs), human embryonic stem cells (hESCs) and human-induced pluripotent stem cells (hiPSCs) is required before they can be applied in the future for personalized medicine and drug discovery. Despite the efforts that have been made with kinome analyses, we still lack in-depth insights into the molecular signatures of receptor tyrosine kinases (RTKs) that are related to pluripotency. Here, we present the first detailed and distinct repertoire of RTK characteristic for hPSC pluripotency by determining both the expression and phosphorylation profiles of RTKs in hESCs and hiPSCs using reverse transcriptase-polymerase chain reaction with degenerate primers that target conserved tyrosine kinase domains and phospho-RTK array, respectively. Among the RTKs tested, the up-regulation of EPHA1, ERBB2, FGFR4 and VEGFR2 and the down-regulation of AXL, EPHA4, PDGFRB and TYRO3 in terms of both their expression and phosphorylation levels were predominantly related to the maintenance of hPSC pluripotency. Notably, the specific inhibition of AXL was significantly advantageous in maintaining undifferentiated hESCs and hiPSCs and for the overall efficiency and kinetics of hiPSC generation. Additionally, a global phosphoproteomic analysis showed that ∼30% of the proteins (293 of 970 phosphoproteins) showed differential phosphorylation upon AXL inhibition in undifferentiated hPSCs, revealing the potential contribution of AXL-mediated phosphorylation dynamics to pluripotency-related signaling networks. Our findings provide a novel molecular signature of AXL in pluripotency control that will complement existing pluripotency-kinome networks.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Sequência de Aminoácidos , Ciclo Celular/genética , Células Cultivadas , Cromatografia Líquida , Ativação Enzimática , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Dados de Sequência Molecular , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fosforilação , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1/genética , Transdução de Sinais/genética , Espectrometria de Massas em Tandem , Receptor Tirosina Quinase Axl
20.
J Biol Chem ; 287(47): 39698-709, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23012353

RESUMO

Histone-modifying enzymes play a pivotal role in gene expression and repression. In human, DOT1L (Dot1-like) is the only known histone H3 lysine 79 methyltransferase. hDOT1L is associated with transcriptional activation, but the general mechanism connecting hDOT1L to active transcription remains largely unknown. Here, we report that hDOT1L interacts with the phosphorylated C-terminal domain of actively transcribing RNA polymerase II (RNAPII) through a region conserved uniquely in multicellular DOT1 proteins. Genome-wide profiling analyses indicate that the occupancy of hDOT1L largely overlaps with that of RNAPII at actively transcribed genes, especially surrounding transcriptional start sites, in embryonic carcinoma NCCIT cells. We also find that C-terminal domain binding or H3K79 methylations by hDOT1L is important for the expression of target genes such as NANOG and OCT4 and a marker for pluripotency in NCCIT cells. Our results indicate that a functional interaction between hDOT1L and RNAPII targets hDOT1L and subsequent H3K79 methylations to actively transcribed genes.


Assuntos
Regulação da Expressão Gênica/fisiologia , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Metiltransferases/metabolismo , RNA Polimerase II/metabolismo , Transcrição Gênica/fisiologia , Estudo de Associação Genômica Ampla , Células HEK293 , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Humanos , Metilação , Metiltransferases/genética , Ligação Proteica/fisiologia , RNA Polimerase II/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA