Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
2.
Cell Stem Cell ; 31(1): 25-38.e8, 2024 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-38086390

RESUMO

Human embryonic stem cell (hESC)-derived midbrain dopaminergic (mDA) cell transplantation is a promising therapeutic strategy for Parkinson's disease (PD). Here, we present the derivation of high-purity mDA progenitors from clinical-grade hESCs on a large scale under rigorous good manufacturing practice (GMP) conditions. We also assessed the toxicity, biodistribution, and tumorigenicity of these cells in immunodeficient rats in good laboratory practice (GLP)-compliant facilities. Various doses of mDA progenitors were transplanted into hemi-parkinsonian rats, and a significant dose-dependent behavioral improvement was observed with a minimal effective dose range of 5,000-10,000 mDA progenitor cells. These results provided insights into determining a low cell dosage (3.15 million cells) for human clinical trials. Based on these results, approval for a phase 1/2a clinical trial for PD cell therapy was obtained from the Ministry of Food and Drug Safety in Korea, and a clinical trial for treating patients with PD has commenced.


Assuntos
Células-Tronco Embrionárias Humanas , Doença de Parkinson , Humanos , Ratos , Animais , Doença de Parkinson/terapia , Distribuição Tecidual , Neurônios Dopaminérgicos , Transplante de Células-Tronco/métodos , Mesencéfalo , Dopamina , Diferenciação Celular
3.
J Yeungnam Med Sci ; 40(Suppl): S47-S55, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37641557

RESUMO

BACKGROUND: This study aimed to elucidate the association between total lean muscle mass and the incidence of non-alcoholic fatty liver disease (NAFLD) in the adult Korean population. METHODS: Utilizing data derived from the 18-year prospective cohort of the Korean Genome and Epidemiology Study, NAFLD was diagnosed via the hepatic steatosis index with an established cutoff value of 36. Lean muscle mass was assessed via bioelectrical impedance analysis and subsequently divided into tertiles. A generalized mixed model with a logit link was employed for repeated measures data analysis, accounting for potential confounders. RESULTS: Analysis encompassed 7,794 participants yielding 49,177 measurements. The findings revealed a markedly increased incidence of NAFLD in the lower tertiles of muscle mass, specifically, tertile 1 (odds ratio [OR], 20.65; 95% confidence interval [CI], 9.66-44.11) and tertile 2 (OR, 4.57; 95% CI, 2.11-9.91), in comparison to tertile 3. Age-dependent decreases in the OR were observed within the tertile 1 group, with ORs of 10.12 at age of 40 years and 4.96 at age of 80 years. Moreover, each 1%-point increment in total muscle mass corresponded with an estimated OR of 0.87 (95% CI, 0.82-0.93) for NAFLD resolution. CONCLUSIONS: The study demonstrates a significant association between total muscle mass and NAFLD prevalence among Korean adults. Given the potential endocrine role of muscle mass in NAFLD pathogenesis, interventions aimed at enhancing muscle mass might serve as an effective public health strategy for mitigating NAFLD prevalence.

5.
Tissue Eng Regen Med ; 19(6): 1349-1358, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36036887

RESUMO

BACKGROUND: Spinal cord injury (SCI) results in permanent impairment of motor and sensory functions at and below the lesion site. There is no therapeutic option to the functional recovery of SCI involving diverse injury responses of different cell types in the lesion that limit endogenous nerve regeneration. In this regard, cell replacement therapy utilizing stem cells or their derivatives has become a highly promising approach to promote locomotor recovery. For this reason, the demand for a safe and efficient multipotent cell source that can differentiate into various neural cells is increasing. In this study, we evaluated the efficacy and safety of human polysialylated-neural cell adhesion molecule (PSA-NCAM)-positive neural precursor cells (hNPCsPSA-NCAM+) as a treatment for SCI. METHODS: One hundred thousand hNPCsPSA-NCAM+ isolated from human embryonic stem cell-derived NPCs were transplanted into the lesion site by microinjection 7 days after contusive SCI at the thoracic level. We examined the histological characteristics of the graft and behavioral improvement in the SCI rats 10 weeks after transplantation. RESULTS: Locomotor activity improvement was estimated by the Basso-Beattie-Bresnahan locomotor rating scale. Behavioral tests revealed that the transplantation of the hNPCsPSA-NCAM+ into the injured spinal cords of rats significantly improved locomotor function. Histological examination showed that hNPCsPSA-NCAM+ had differentiated into neural cells and successfully integrated into the host tissue with no evidence of tumor formation. We investigated cytokine expressions, which led to the early therapeutic effect of hNPCsPSA-NCAM+, and found that some undifferentiated NPCs still expressed midkine, a well-known neurotrophic factor involved in neural development and inflammatory responses, 10 weeks after transplantation. CONCLUSION: Our results demonstrate that hNPCsPSA-NCAM+ serve as a safe and efficient cell source which has the potential to improve impaired motor function following SCI.


Assuntos
Células-Tronco Embrionárias Humanas , Células-Tronco Neurais , Traumatismos da Medula Espinal , Ratos , Animais , Humanos , Moléculas de Adesão de Célula Nervosa/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/patologia , Células-Tronco Embrionárias Humanas/transplante , Traumatismos da Medula Espinal/terapia , Modelos Animais de Doenças
6.
Biomaterials ; 283: 121429, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35217482

RESUMO

The bleeding disorder hemophilia A (HA) is caused by a single-gene (F8) defect and its clinical symptom can be substantially improved by a small increase in the plasma coagulation factor VIII (FVIII) level. In this study, we used F8-defective human induced pluripotent stem cells from an HA patient (F8d-HA hiPSCs) and F8-corrected (F8c) HA hiPSCs produced by CRISPR/Cas9 genome engineering of F8d-HA hiPSCs. We obtained a highly enriched population of CD157+ cells from CRISPR/Cas9-edited F8c-HA hiPSCs. These cells exhibited multiple cellular and functional phenotypes of endothelial cells (ECs) with significant levels of FVIII activity, which was not observed in F8d-HA hiPSC-ECs. After transplantation, the engineered F8c-HA hiPSC-ECs dramatically changed bleeding episodes in HA animals and restored plasma FVIII activity. Notably, grafting a high dose of ECs substantially reduced the bleeding time during multiple consecutive bleeding challenges in HA mice, demonstrating a robust hemostatic effect (90% survival). Furthermore, the engrafted ECs survived more than 3 months in HA mice and reversed bleeding phenotypes against lethal wounding challenges. We also produced F8c-HA hiPSC-derived 3D liver organoids by assembling three different cell types in microwell devices and confirmed its therapeutic effect in HA animals. Our data demonstrate that the combination of genome-engineering and iPSC technologies represents a novel modality that allows autologous cell-mediated gene therapy for treating HA.


Assuntos
Hemofilia A , Células-Tronco Pluripotentes Induzidas , Animais , Sistemas CRISPR-Cas/genética , Células Endoteliais/metabolismo , Hemofilia A/genética , Hemofilia A/metabolismo , Hemofilia A/terapia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Organoides/metabolismo
7.
Stem Cell Res ; 49: 102026, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33038747

RESUMO

Factor VII (FVII) deficiency is the most common among the rare bleeding disorders, which is caused by mutations in coagulation factor VII. Clinical features caused by FVII deficiency vary from mild or asymptomatic to fatal cerebral hemorrhage. We generated an induced pluripotent stem cell (iPSC) line, YCMi002-A, from FVII deficiency patient-derived fibroblasts. YCMi002-A cells are characterized by novel compound heterozygous mutations. The c.345C > A; p.C115X is well known and the second one, c.1276C > T; p.Q426X, remains novel. YCMi002-A cells may help researchers to understand correlation between these mutations and the symptoms of FVII deficiency.


Assuntos
Deficiência do Fator VII , Células-Tronco Pluripotentes Induzidas , Fator VII/genética , Deficiência do Fator VII/genética , Humanos , Mutação
8.
Stem Cell Reports ; 12(6): 1242-1249, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31105049

RESUMO

Hemophilia A (HA) is caused by genetic mutations in the blood coagulation factor VIII (FVIII) gene. Genome-editing approaches can be used to target the mutated site itself in patient-derived induced pluripotent stem cells (iPSCs). However, these approaches can be hampered by difficulty in preparing thousands of editing platforms for each corresponding variant found in HA patients. Here, we report a universal approach to correct the various mutations in HA patient iPSCs by the targeted insertion of the FVIII gene into the human H11 site via CRISPR/Cas9. We derived corrected clones from two types of patient iPSCs with frequencies of up to 64% and 66%, respectively, without detectable unwanted off-target mutations. Moreover, we demonstrated that endothelial cells differentiated from the corrected iPSCs successfully secreted functional protein. This strategy may provide a universal therapeutic method for correcting all genetic variants found in HA patients.


Assuntos
Sistemas CRISPR-Cas , Fator VIII , Edição de Genes , Hemofilia A , Células-Tronco Pluripotentes Induzidas , Mutação , Fator VIII/biossíntese , Fator VIII/genética , Hemofilia A/genética , Hemofilia A/metabolismo , Hemofilia A/patologia , Hemofilia A/terapia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia
9.
Exp Mol Med ; 51(4): 1-9, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30996250

RESUMO

Target-specific genome editing, using engineered nucleases zinc finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN), and type II clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9), is considered a promising approach to correct disease-causing mutations in various human diseases. In particular, hemophilia A can be considered an ideal target for gene modification via engineered nucleases because it is a monogenic disease caused by a mutation in coagulation factor VIII (FVIII), and a mild restoration of FVIII levels in plasma can prevent disease symptoms in patients with severe hemophilia A. In this study, we describe a universal genome correction strategy to restore FVIII expression in induced pluripotent stem cells (iPSCs) derived from a patient with hemophilia A by the human elongation factor 1 alpha (EF1α)-mediated normal FVIII gene expression in the FVIII locus of the patient. We used the CRISPR/Cas9-mediated homology-directed repair (HDR) system to insert the B-domain deleted from the FVIII gene with the human EF1α promoter. After gene targeting, the FVIII gene was correctly inserted into iPSC lines at a high frequency (81.81%), and these cell lines retained pluripotency after knock-in and neomycin resistance cassette removal. More importantly, we confirmed that endothelial cells from the gene-corrected iPSCs could generate functionally active FVIII protein from the inserted FVIII gene. This is the first demonstration that the FVIII locus is a suitable site for integration of the normal FVIII gene and can restore FVIII expression by the EF1α promoter in endothelial cells differentiated from the hemophilia A patient-derived gene-corrected iPSCs.


Assuntos
Fator VIII/metabolismo , Hemofilia A/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Cultivadas , Éxons/genética , Fator VIII/genética , Edição de Genes , Engenharia Genética/métodos , Células HEK293 , Hemofilia A/genética , Humanos , Regiões Promotoras Genéticas/genética , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo
10.
Exp Mol Med ; 50(4): 1-13, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29650976

RESUMO

The canonical Wnt signal pathway plays a pivotal role in anteroposterior patterning and midbrain specification during early neurogenesis. Activating Wnt signal has been a strategy for differentiating human pluripotent stem cells (PSCs) into midbrain dopaminergic (DA) neurons; however, the underlying molecular mechanism(s) of how the Wnt signal drives posterior fate remained unclear. In this study, we found that activating the canonical Wnt signal significantly upregulated the expression of EN1, a midbrain-specific marker, in a fibroblast growth factor signal-dependent manner in human PSC-derived neural precursor cells (NPCs). The EN1 promoter region contains a putative TCF4-binding site that directly interacts with the ß-catenin/TCF complex upon Wnt signal activation. Once differentiated, NPCs treated with a Wnt signal agonist gave rise to functional midbrain neurons including glutamatergic, GABAergic, and DA neurons. Our results provide a potential molecular mechanism that underlies midbrain specification of human PSC-derived NPCs by Wnt activation, as well as a differentiation paradigm for generating human midbrain neurons that may serve as a cellular platform for studying the ontogenesis of midbrain neurons and neurological diseases relevant to the midbrain.


Assuntos
Proteínas de Homeodomínio/genética , Mesencéfalo/metabolismo , Células-Tronco Pluripotentes/metabolismo , Regiões Promotoras Genéticas , Fator de Transcrição 4/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Mesencéfalo/citologia , Células-Tronco Pluripotentes/citologia , Ligação Proteica , RNA Interferente Pequeno/genética
11.
Nat Protoc ; 11(11): 2154-2169, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27711053

RESUMO

Genome engineering technology using engineered nucleases has been rapidly developing, enabling the efficient correction of simple mutations. However, the precise correction of structural variations (SVs) such as large inversions remains limited. Here we describe a detailed procedure for the modeling or correction of large chromosomal rearrangements and short nucleotide repeat expansions using engineered nucleases in human induced pluripotent stem cells (hiPSCs) from a healthy donor and patients with SVs. This protocol includes the delivery of engineered nucleases with no donor template to hiPSCs, and genotyping and derivation/characterization of gene-manipulated hiPSC clones. With engineered nucleases, genomic inversions, reversions, and deletions of short nucleotide expansions can be identified in 2 weeks, and desired clones can be generated in as little as 3-4 weeks. This protocol enables the correction of large inverted segments and short nucleotide repeat expansions in diseases such as hemophilia A, fragile X syndrome, Hunter syndrome, and Friedreich's ataxia.


Assuntos
Sistemas CRISPR-Cas/genética , Engenharia Genética/métodos , Variação Genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Genéticos , Sequência de Bases , Reparo do DNA por Junção de Extremidades/genética , Rearranjo Gênico/genética , Genoma Humano/genética , Humanos , Sequências Repetitivas de Ácido Nucleico/genética
12.
Hum Genet ; 135(9): 977-81, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27357631

RESUMO

Hemophilia is caused by various mutations in blood coagulation factor genes, including factor VIII (FVIII) and factor IX (FIX), that encode key proteins in the blood clotting pathway. Although the addition of therapeutic genes or infusion of clotting factors may be used to remedy hemophilia's symptoms, no permanent cure for the disease exists. Moreover, patients often develop neutralizing antibodies or experience adverse effects that limit the therapy's benefits. However, targeted gene therapy involving the precise correction of these mutated genes at the genome level using programmable nucleases is a promising strategy. These nucleases can induce double-strand breaks (DSBs) on genomes, and repairs of such induced DSBs by the two cellular repair systems enable a targeted gene correction. Going beyond cultured cell systems, we are now entering the age of direct gene correction in vivo using various delivery tools. Here, we describe the current status of in vivo and ex vivo genome-editing technology related to potential hemophilia gene correction and the prominent issues surrounding its application in patients with monogenic diseases.


Assuntos
Edição de Genes , Terapia Genética , Hemofilia A/terapia , Humanos
13.
Trends Biotechnol ; 34(7): 548-561, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27016031

RESUMO

The analysis of chromosomal structural variations (SVs), such as inversions and translocations, was made possible by the completion of the human genome project and the development of genome-wide sequencing technologies. SVs contribute to genetic diversity and evolution, although some SVs can cause diseases such as hemophilia A in humans. Genome engineering technology using programmable nucleases (e.g., ZFNs, TALENs, and CRISPR/Cas9) has been rapidly developed, enabling precise and efficient genome editing for SV research. Here, we review advances in modeling and gene correction of SVs, focusing on inversion, translocation, and nucleotide repeat expansion.


Assuntos
Aberrações Cromossômicas , Cromossomos Humanos , Edição de Genes/métodos , Terapia Genética/métodos , Variação Genética , Genoma Humano , Humanos
14.
Cell Rep ; 13(2): 234-41, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26440889

RESUMO

Fragile X syndrome (FXS) is the most common form of inherited intellectual disability, resulting from a CGG repeat expansion in the fragile X mental retardation 1 (FMR1) gene. Here, we report a strategy for CGG repeat correction using CRISPR/Cas9 for targeted deletion in both embryonic stem cells and induced pluripotent stem cells derived from FXS patients. Following gene correction in FXS induced pluripotent stem cells, FMR1 expression was restored and sustained in neural precursor cells and mature neurons. Strikingly, after removal of the CGG repeats, the upstream CpG island of the FMR1 promoter showed extensive demethylation, an open chromatin state, and transcription initiation. These results suggest a silencing maintenance mechanism for the FMR1 promoter that is dependent on the existence of the CGG repeat expansion. Our strategy for deletion of trinucleotide repeats provides further insights into the molecular mechanisms of FXS and future therapies of trinucleotide repeat disorders.


Assuntos
Metilação de DNA , Proteína do X Frágil de Retardo Mental/genética , Síndrome do Cromossomo X Frágil/genética , Inativação Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/metabolismo , Repetições de Trinucleotídeos , Sistemas CRISPR-Cas , Células Cultivadas , Ilhas de CpG , Proteína do X Frágil de Retardo Mental/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/citologia , Regiões Promotoras Genéticas
15.
Cell Stem Cell ; 17(2): 213-20, 2015 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-26212079

RESUMO

Hemophilia A is an X-linked genetic disorder caused by mutations in the F8 gene, which encodes the blood coagulation factor VIII. Almost half of all severe hemophilia A cases result from two gross (140-kbp or 600-kbp) chromosomal inversions that involve introns 1 and 22 of the F8 gene, respectively. We derived induced pluripotent stem cells (iPSCs) from patients with these inversion genotypes and used CRISPR-Cas9 nucleases to revert these chromosomal segments back to the WT situation. We isolated inversion-corrected iPSCs with frequencies of up to 6.7% without detectable off-target mutations based on whole-genome sequencing or targeted deep sequencing. Endothelial cells differentiated from corrected iPSCs expressed the F8 gene and functionally rescued factor VIII deficiency in an otherwise lethal mouse model of hemophilia. Our results therefore provide a proof of principle for functional correction of large chromosomal rearrangements in patient-derived iPSCs and suggest potential therapeutic applications.


Assuntos
Sistemas CRISPR-Cas/genética , Inversão Cromossômica/genética , Fator VII/genética , Hemofilia A/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Sequência de Bases , Células Clonais , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular
16.
Stem Cell Reports ; 4(5): 821-34, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25937368

RESUMO

Tumorigenic potential of human pluripotent stem cells (hPSCs) is an important issue in clinical applications. Despite many efforts, PSC-derived neural precursor cells (NPCs) have repeatedly induced tumors in animal models even though pluripotent cells were not detected. We found that polysialic acid-neural cell adhesion molecule (PSA-NCAM)(-) cells among the early NPCs caused tumors, whereas PSA-NCAM(+) cells were nontumorigenic. Molecular profiling, global gene analysis, and multilineage differentiation of PSA-NCAM(-) cells confirm that they are multipotent neural crest stem cells (NCSCs) that could differentiate into both ectodermal and mesodermal lineages. Transplantation of PSA-NCAM(-) cells in a gradient manner mixed with PSA-NCAM(+) cells proportionally increased mesodermal tumor formation and unwanted grafts such as PERIPHERIN(+) cells or pigmented cells in the rat brain. Therefore, we suggest that NCSCs are a critical target for tumor prevention in hPSC-derived NPCs, and removal of PSA-NCAM(-) cells eliminates the tumorigenic potential originating from NCSCs after transplantation.


Assuntos
Molécula L1 de Adesão de Célula Nervosa/metabolismo , Crista Neural/metabolismo , Células-Tronco Pluripotentes/citologia , Ácidos Siálicos/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Ectoderma/citologia , Ectoderma/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Humanos , Imuno-Histoquímica , Masculino , Mesoderma/citologia , Mesoderma/metabolismo , Neoplasias/etiologia , Neoplasias/metabolismo , Molécula L1 de Adesão de Célula Nervosa/genética , Crista Neural/citologia , Crista Neural/transplante , Periferinas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Ratos , Ratos Sprague-Dawley , Ácidos Siálicos/genética , Transcriptoma
17.
Cancer Lett ; 351(1): 72-80, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-24887560

RESUMO

TGF-ß signaling plays an important role in breast cancer progression and metastasis. Epithelial-mesenchymal transition (EMT) is an important step in the progression of solid tumors to metastatic disease. We previously reported that IN-1130, a novel transforming growth factor-ß type I receptor kinase (ALK5) inhibitor, suppressed renal fibrosis in obstructive nephropathy (Moon et al., 2006). Here, we show that IN-1130 suppressed EMT and the lung metastasis of mammary tumors in mouse models. Treating human and mouse cell lines with IN-1130 inhibited TGF-ß-mediated transcriptional activation, the phosphorylation and nuclear translocation of Smad2, and TGF-ß-induced-EMT, which induces morphological changes in epithelial cells. Additionally, we demonstrated that IN-1130 blocked TGF-ß-induced 4T1 mammary cancer cell migration and invasion. The TGF-ß-mediated increase in matrix metalloproteinase (MMP)-2 and MMP-9 expression was restored by IN-1130 co-treatment with TGF-ß in human epithelial cells and in 4T1 cells. Furthermore, we found that lung metastasis from primary breast cancer was inhibited by IN-1130 in both 4T1-xenografted BALB/c mice and MMTV/c-Neu transgenic mice without any change in primary tumor volume. IN-1130 prolonged the life span of tumor-bearing mice. In summary, this study indicated that IN-1130 has therapeutic potential for preventing breast cancer metastasis to the lung.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Imidazóis/farmacologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Mamárias Experimentais/tratamento farmacológico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Quinoxalinas/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Movimento Celular , Núcleo Celular/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Pulmão/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Transplante de Neoplasias , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Carga Tumoral/efeitos dos fármacos
18.
Proc Natl Acad Sci U S A ; 111(25): 9253-8, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24927536

RESUMO

Hemophilia A, one of the most common genetic bleeding disorders, is caused by various mutations in the blood coagulation factor VIII (F8) gene. Among the genotypes that result in hemophilia A, two different types of chromosomal inversions that involve a portion of the F8 gene are most frequent, accounting for almost half of all severe hemophilia A cases. In this study, we used a transcription activator-like effector nuclease (TALEN) pair to invert a 140-kbp chromosomal segment that spans the portion of the F8 gene in human induced pluripotent stem cells (iPSCs) to create a hemophilia A model cell line. In addition, we reverted the inverted segment back to its normal orientation in the hemophilia model iPSCs using the same TALEN pair. Importantly, we detected the F8 mRNA in cells derived from the reverted iPSCs lines, but not in those derived from the clones with the inverted segment. Thus, we showed that TALENs can be used both for creating disease models associated with chromosomal rearrangements in iPSCs and for correcting genetic defects caused by chromosomal inversions. This strategy provides an iPSC-based novel therapeutic option for the treatment of hemophilia A and other genetic diseases caused by chromosomal inversions.


Assuntos
Inversão Cromossômica , Desoxirribonucleases/biossíntese , Fator VIII/genética , Marcação de Genes/métodos , Hemofilia A , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Desoxirribonucleases/genética , Fator VIII/metabolismo , Células HEK293 , Hemofilia A/genética , Hemofilia A/metabolismo , Hemofilia A/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/patologia
19.
PLoS One ; 8(2): e56242, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23437103

RESUMO

X-linked adrenoleukodystrophy (X-ALD) is a peroxisomal disorder caused by mutations in the ABCD1 gene that encodes the peroxisomal ATP-binding cassette (ABC) transporter subfamily D member 1 protein (ABCD1), which is referred to as the adrenoleukodystrophy protein (ALDP). Induction of the ABCD2 gene, the closest homolog of ABCD1, has been mentioned as a possible therapeutic option for the defective ABCD1 protein in X-ALD. However, little is known about the transcriptional regulation of ABCD2 gene expression. Here, through in silico analysis, we found two putative TCF-4 binding elements between nucleotide positions -360 and -260 of the promoter region of the ABCD2 gene. The transcriptional activity of the ABCD2 promoter was strongly increased by ectopic expression of ß-catenin and TCF-4. In addition, mutation of either or both TCF-4 binding elements by site-directed mutagenesis decreased promoter activity. This was further validated by the finding that ß-catenin and the promoter of the ABCD2 gene were pulled down with a ß-catenin antibody in a chromatin immunoprecipitation assay. Moreover, real-time PCR analysis revealed that ß-catenin and TCF-4 increased mRNA levels of ABCD2 in both a hepatocellular carcinoma cell line and primary fibroblasts from an X-ALD patient. Interestingly, we found that the levels of very long chain fatty acids were decreased by ectopic expression of ABCD2-GFP as well as ß-catenin and TCF-4. Taken together, our results demonstrate for the first time the direct regulation of ABCD2 by ß-catenin and TCF-4.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Adrenoleucodistrofia/genética , Adrenoleucodistrofia/terapia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Terapia de Alvo Molecular , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo , Subfamília D de Transportador de Cassetes de Ligação de ATP , Adrenoleucodistrofia/patologia , Sequência de Bases , Sítios de Ligação/genética , Ácidos Graxos/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Inativação Gênica , Proteínas de Fluorescência Verde/metabolismo , Células Hep G2 , Humanos , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição 4 , Transcrição Gênica , Ativação Transcricional/genética , Regulação para Cima/genética
20.
Exp Mol Med ; 44(3): 202-13, 2012 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-22179105

RESUMO

The generation of disease-specific induced pluripotent stem cell (iPSC) lines from patients with incurable diseases is a promising approach for studying disease mechanisms and drug screening. Such innovation enables to obtain autologous cell sources in regenerative medicine. Herein, we report the generation and characterization of iPSCs from fibroblasts of patients with sporadic or familial diseases, including Parkinson's disease (PD), Alzheimer's disease (AD), juvenile-onset, type I diabetes mellitus (JDM), and Duchenne type muscular dystrophy (DMD), as well as from normal human fibroblasts (WT). As an example to modeling disease using disease-specific iPSCs, we also discuss the previously established childhood cerebral adrenoleukodystrophy (CCALD)- and adrenomyeloneuropathy (AMN)-iPSCs by our group. Through DNA fingerprinting analysis, the origins of generated disease-specific iPSC lines were identified. Each iPSC line exhibited an intense alkaline phosphatase activity, expression of pluripotent markers, and the potential to differentiate into all three embryonic germ layers: the ectoderm, endoderm, and mesoderm. Expression of endogenous pluripotent markers and downregulation of retrovirus-delivered transgenes [OCT4 (POU5F1), SOX2, KLF4, and c-MYC] were observed in the generated iPSCs. Collectively, our results demonstrated that disease-specific iPSC lines characteristically resembled hESC lines. Furthermore, we were able to differentiate PD-iPSCs, one of the disease-specific-iPSC lines we generated, into dopaminergic (DA) neurons, the cell type mostly affected by PD. These PD-specific DA neurons along with other examples of cell models derived from disease-specific iPSCs would provide a powerful platform for examining the pathophysiology of relevant diseases at the cellular and molecular levels and for developing new drugs and therapeutic regimens.


Assuntos
Doença de Alzheimer/patologia , Diabetes Mellitus Tipo 1/patologia , Descoberta de Drogas/métodos , Células-Tronco Pluripotentes Induzidas/patologia , Distrofia Muscular de Duchenne/patologia , Doença de Parkinson/patologia , Doença de Alzheimer/genética , Diferenciação Celular , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Distrofia Muscular de Duchenne/genética , Doença de Parkinson/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...