Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Heliyon ; 10(2): e24590, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38312695

RESUMO

Hereditary transthyretin (TTR) amyloidosis (ATTRv amyloidosis) is autosomal dominant and caused by mutation of TTR gene. Heterozygous ATTR Tyr114Cys (p.Tyr134Cys) amyloidosis is a lethal disease with a life expectancy of about 10 years after onset of the disease. However, the molecular pathogenesis of ATTR Tyr114Cys amyloidosis is still largely unknown. In this study, we took advantage of disease-specific induced pluripotent stem (iPS) cells and generated & characterized the heterozygous ATTR Tyr114Cys amyloidosis-specific iPS cells (Y114C iPS cells), to determine whether Y114C iPS cells could be useful for elucidating the pathogenesis of ATTR Tyr114Cys amyloidosis. We successfully differentiated heterozygous Y114C iPS cells into hepatocyte like cells (HLCs) mainly producing TTR protein. On day 27 after differentiation, the expression of hepatocyte maker albumin was detected, and TTR expression was significantly increased in HLCs differentiated from Y114C iPS cells. LC-MS/MS analysis showed that both WT TTR & ATTR Y114C protein were indeed expressed in the HLCs differentiated from Y114C iPS cells. Notably, the number of detected peptides derived from ATTR Y114C protein was lower than that of WT TTR protein, indeed indicating the clinical phenotype of ATTR Tyr114Cys amyloidosis. Taken together, we first reported the heterozygous Y114C iPS cells generated from patient with ATTR Tyr114Cys amyloidosis, and suggested that Y114C iPS cells could be a potential pathological tool, which may contribute to elucidating the molecular pathogenesis of heterozygous ATTR Tyr114Cys amyloidosis.

2.
J Virol ; 96(9): e0035622, 2022 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-35420440

RESUMO

Human endogenous retroviruses (HERVs) occupy approximately 8% of the human genome. HERVs, transcribed in early embryos, are epigenetically silenced in somatic cells, except under pathological conditions. HERV-K is thought to protect embryos from exogenous viral infection. However, uncontrolled HERV-K expression in somatic cells has been implicated in several diseases. Here, we show that SOX2, which plays a key role in maintaining the pluripotency of stem cells, is critical for HERV-K LTR5Hs. HERV-K undergoes retrotransposition within producer cells in the absence of Env expression. Furthermore, we identified new HERV-K integration sites in long-term culture of induced pluripotent stem cells that express SOX2. These results suggest that the strict dependence of HERV-K on SOX2 has allowed HERV-K to protect early embryos during evolution while limiting the potentially harmful effects of HERV-K retrotransposition on host genome integrity in these early embryos. IMPORTANCE Human endogenous retroviruses (HERVs) account for approximately 8% of the human genome; however, the physiological role of HERV-K remains unknown. This study found that HERV-K LTR5Hs and LTR5B were transactivated by SOX2, which is essential for maintaining and reestablishing pluripotency. HERV-K can undergo retrotransposition within producer cells without env expression, and new integration sites may affect cell proliferation. In induced pluripotent stem cells (iPSCs), genomic impairment due to HERV-K retrotransposition has been identified, but it is a rare event. Considering the retention of SOX2-responsive elements in the HERV-K long terminal repeat (LTR) for over 20 million years, we conclude that HERV-K may play important physiological roles in SOX2-expressing cells.


Assuntos
Retrovirus Endógenos , Células-Tronco Pluripotentes Induzidas , Fatores de Transcrição SOXB1 , Retrovirus Endógenos/genética , Humanos , Células-Tronco Pluripotentes Induzidas/virologia , Fatores de Transcrição SOXB1/genética , Sequências Repetidas Terminais/genética , Integração Viral
3.
EMBO J ; 41(8): e109463, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35229328

RESUMO

In order to support bone marrow regeneration after myeloablation, hematopoietic stem cells (HSCs) actively divide to provide both stem and progenitor cells. However, the mechanisms regulating HSC function and cell fate choice during hematopoietic recovery remain unclear. We herein provide novel insights into HSC regulation during regeneration by focusing on mitochondrial metabolism and ATP citrate lyase (ACLY). After 5-fluorouracil-induced myeloablation, HSCs highly expressing endothelial protein C receptor (EPCRhigh ) were enriched within the stem cell fraction at the expense of more proliferative EPCRLow HSCs. These EPCRHigh HSCs were initially more primitive than EPCRLow HSCs and enabled stem cell expansion by enhancing histone acetylation, due to increased activity of ACLY in the early phase of hematopoietic regeneration. In the late phase of recovery, HSCs enhanced differentiation potential by increasing the accessibility of cis-regulatory elements in progenitor cell-related genes, such as CD48. In conditions of reduced mitochondrial metabolism and ACLY activity, these HSCs maintained stem cell phenotypes, while ACLY-dependent histone acetylation promoted differentiation into CD48+ progenitor cells. Collectively, these results indicate that the dynamic control of ACLY-dependent metabolism and epigenetic alterations is essential for HSC regulation during hematopoietic regeneration.


Assuntos
ATP Citrato (pro-S)-Liase , Medula Óssea , ATP Citrato (pro-S)-Liase/genética , ATP Citrato (pro-S)-Liase/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Histonas/metabolismo
4.
Am J Med Genet A ; 188(6): 1863-1867, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35178855

RESUMO

The diagnosis of mosaicism is challenging in patients with neurofibromatosis type 2 (NF2) subset due to low variant allele frequency. In this study, we generated induced pluripotent stem cells (iPSCs) were generated from a patient clinically diagnosed with NF2 based on multiple schwannomas, including bilateral vestibular schwannomas and meningiomas. Genetic analysis of the patient's mononuclear cells (MNCs) from peripheral blood failed to detect NF2 alteration but successfully found p.Q65X (c.193C>T) mutation in all separate tumors with three intracranial meningiomas and one intraorbital schwannoma, and confirming mosaicism diagnosis in NF2 alteration using deep sequencing. Five different clones with patient-derived iPSCs were established from MNCs in peripheral blood, which showed sufficient expression of pluripotent markers. Genetic analysis showed that one of five generated iPSC lines from MNCs had the same p.Q65X mutation as that found in NF2. There was no significant difference in the expression of genes related to NF2 between iPSC clones with the wild-type and mutant NF2. In this case, clonal expansion of mononuclear bone marrow-derived stem cells recapitulated mosaicism's genetic alteration in NF2. Patient-derived iPSCs from mosaic NF2 would contribute to further functional research of NF2 alteration.


Assuntos
Células-Tronco Pluripotentes Induzidas , Neoplasias Meníngeas , Meningioma , Neurofibromatose 2 , Células Clonais/patologia , Genes da Neurofibromatose 2 , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Neoplasias Meníngeas/genética , Meningioma/genética , Mutação , Neurofibromatose 2/diagnóstico , Neurofibromatose 2/genética
5.
Redox Biol ; 41: 101921, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33706170

RESUMO

Mitochondria participate in various metabolic pathways, and their dysregulation results in multiple disorders, including aging-related diseases. However, the metabolic changes and mechanisms of mitochondrial disorders are not fully understood. Here, we found that induced pluripotent stem cells (iPSCs) from a patient with mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) showed attenuated proliferation and survival when glycolysis was inhibited. These deficits were rescued by taurine administration. Metabolomic analyses showed that the ratio of the reduced (GSH) to oxidized glutathione (GSSG) was decreased; whereas the levels of cysteine, a substrate of GSH, and oxidative stress markers were upregulated in MELAS iPSCs. Taurine normalized these changes, suggesting that MELAS iPSCs were affected by the oxidative stress and taurine reduced its influence. We also analyzed the retinal pigment epithelium (RPE) differentiated from MELAS iPSCs by using a three-dimensional culture system and found that it showed epithelial mesenchymal transition (EMT), which was suppressed by taurine. Therefore, mitochondrial dysfunction caused metabolic changes, accumulation of oxidative stress that depleted GSH, and EMT in the RPE that could be involved in retinal pathogenesis. Because all these phenomena were sensitive to taurine treatment, we conclude that administration of taurine may be a potential new therapeutic approach for mitochondria-related retinal diseases.


Assuntos
Células-Tronco Pluripotentes Induzidas , Epitélio Pigmentado da Retina , Transição Epitelial-Mesenquimal , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Mitocôndrias , Epitélio Pigmentado da Retina/metabolismo , Taurina
6.
Sci Rep ; 11(1): 1552, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33452387

RESUMO

Moyamoya disease (MMD) is characterized by progressive bilateral stenotic changes in the terminal portion of the internal carotid arteries. Although RNF213 was identified as a susceptibility gene for MMD, the exact pathogenesis remains unknown. Immunohistochemical analysis of autopsy specimens from a patient with MMD revealed marked accumulation of hyaluronan and chondroitin sulfate (CS) in the thickened intima of occlusive lesions of MMD. Hyaluronan synthase 2 was strongly expressed in endothelial progenitor cells in the thickened intima. Furthermore, MMD lesions showed minimal staining for CS and hyaluronan in the endothelium, in contrast to control endothelium showing positive staining for both. Glycosaminoglycans of endothelial cells derived from MMD and control induced pluripotent stem cells demonstrated a decreased amount of CS, especially sulfated CS, in MMD. A computational fluid dynamics model showed highest wall shear stress values in the terminal portion of the internal carotid artery, which is the predisposing region in MMD. Because the peri-endothelial extracellular matrix plays an important role in protection, cell adhesion and migration, an altered peri-endothelial matrix in MMD may contribute to endothelial vulnerability to wall shear stress. Invading endothelial progenitor cells repairing endothelial injury would produce excessive hyaluronan and CS in the intima, and cause vascular stenosis.


Assuntos
Células Endoteliais/metabolismo , Doença de Moyamoya/fisiopatologia , Adenosina Trifosfatases/metabolismo , Adolescente , Idoso , Fenômenos Biomecânicos/fisiologia , Artéria Carótida Interna/patologia , Espessura Intima-Media Carotídea , Sulfatos de Condroitina/análise , Células Endoteliais/fisiologia , Endotélio/metabolismo , Feminino , Predisposição Genética para Doença , Humanos , Ácido Hialurônico/análise , Hidrodinâmica , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Doença de Moyamoya/metabolismo , Resistência ao Cisalhamento/fisiologia , Estresse Mecânico , Ubiquitina-Proteína Ligases/metabolismo
7.
Sci Rep ; 10(1): 13927, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32811847

RESUMO

Many human diseases ranging from cancer to hereditary disorders are caused by single-nucleotide mutations in critical genes. Repairing these mutations would significantly improve the quality of life for patients with hereditary diseases. However, current procedures for repairing deleterious single-nucleotide mutations are not straightforward, requiring multiple steps and taking several months to complete. In the current study, we aimed to repair pathogenic allele-specific single-nucleotide mutations using a single round of genome editing. Using high-fidelity, site-specific nuclease AsCas12a/Cpf1, we attempted to repair pathogenic single-nucleotide variants (SNVs) in disease-specific induced pluripotent stem cells. As a result, we achieved repair of the Met918Thr SNV in human oncogene RET with the inclusion of a single-nucleotide marker, followed by absolute markerless, scarless repair of the RET SNV with no detected off-target effects. The markerless method was then confirmed in human type VII collagen-encoding gene COL7A1. Thus, using this One-SHOT method, we successfully reduced the number of genetic manipulations required for genome repair from two consecutive events to one, resulting in allele-specific repair that can be completed within 3 weeks, with or without a single-nucleotide marker. Our findings suggest that One-SHOT can be used to repair other types of mutations, with potential beyond human medicine.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Associadas a CRISPR/metabolismo , Endodesoxirribonucleases/metabolismo , Edição de Genes/métodos , Polimorfismo de Nucleotídeo Único/genética , Alelos , Proteínas de Bactérias/genética , Proteínas Associadas a CRISPR/genética , Sistemas CRISPR-Cas/genética , Colágeno Tipo VII/genética , Colágeno Tipo VII/metabolismo , Endodesoxirribonucleases/genética , Endonucleases/genética , Genoma Humano/genética , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Mutação/genética , Nucleotídeos/genética , Células-Tronco Pluripotentes/fisiologia , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo
8.
Stem Cell Reports ; 13(2): 322-337, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31378669

RESUMO

Kidney formation is regulated by the balance between maintenance and differentiation of nephron progenitor cells (NPCs). Now that directed differentiation of NPCs from human induced pluripotent stem cells (iPSCs) can be achieved, maintenance and propagation of NPCs in vitro should be beneficial for regenerative medicine. Although WNT and FGF signals were previously shown to be essential for NPC propagation, the requirement for BMP/TGFß signaling remains controversial. Here we reveal that activin has superior effects to BMP7 on maintenance efficiency of human iPSC-derived NPCs. Activin expanded ITGA8+/PDGFRA-/SIX2-GFP+ NPCs by 5-fold per week at 80%-90% efficiency, and the propagated cells possessed robust capacity for nephron formation both in vitro and in vivo. The expanded cells also maintained their nephron-forming potential after freezing. Furthermore, the protocol was applicable to multiple non-GFP-tagged iPSC lines. Thus, our activin-based protocol will be applicable to a variety of research fields including disease modeling and drug screening.


Assuntos
Ativinas/farmacologia , Proteína Morfogenética Óssea 7/farmacologia , Proliferação de Células/efeitos dos fármacos , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Diferenciação Celular , Reprogramação Celular , Edição de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Camundongos , Néfrons/citologia , Néfrons/metabolismo , Néfrons/patologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Podócitos/metabolismo , Podócitos/patologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo
9.
Stem Cell Reports ; 11(3): 727-740, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30174315

RESUMO

Mutations in the NPHS1 gene, which encodes NEPHRIN, cause congenital nephrotic syndrome, resulting from impaired slit diaphragm (SD) formation in glomerular podocytes. However, methods for SD reconstitution have been unavailable, thereby limiting studies in the field. In the present study, we established human induced pluripotent stem cells (iPSCs) from a patient with an NPHS1 missense mutation, and reproduced the SD formation process using iPSC-derived kidney organoids. The mutant NEPHRIN failed to become localized on the cell surface for pre-SD domain formation in the induced podocytes. Upon transplantation, the mutant podocytes developed foot processes, but exhibited impaired SD formation. Genetic correction of the single amino acid mutation restored NEPHRIN localization and phosphorylation, colocalization of other SD-associated proteins, and SD formation. Thus, these kidney organoids from patient-derived iPSCs identified SD abnormalities in the podocytes at the initial phase of congenital nephrotic disease.


Assuntos
Células-Tronco Pluripotentes Induzidas/patologia , Proteínas de Membrana/análise , Síndrome Nefrótica/patologia , Organoides/patologia , Podócitos/patologia , Animais , Células Cultivadas , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Rim/metabolismo , Rim/patologia , Proteínas de Membrana/genética , Camundongos SCID , Mutação de Sentido Incorreto , Síndrome Nefrótica/genética , Organoides/metabolismo , Podócitos/metabolismo
10.
PLoS One ; 13(7): e0200790, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30044827

RESUMO

Mesenchymal stem cells (MSCs) isolated from adult human tissues are capable of proliferating in vitro and maintaining their multipotency, making them attractive cell sources for regenerative medicine. However, the availability and capability of self-renewal under current preparation regimes are limited. Induced pluripotent stem cells (iPSCs) now offer an alternative, similar cell source to MSCs. Herein, we established new methods for differentiating hiPSCs into MSCs via mesoderm-like and neuroepithelium-like cells. Both derived MSC populations exhibited self-renewal and multipotency, as well as therapeutic potential in mouse models of skin wounds, pressure ulcers, and osteoarthritis. Interestingly, the therapeutic effects differ between the two types of MSCs in the disease models, suggesting that the therapeutic effect depends on the cell origin. Our results provide valuable basic insights for the clinical application of such cells.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Medicina Regenerativa/métodos , Adipócitos/citologia , Animais , Diferenciação Celular , Modelos Animais de Doenças , Humanos , Masculino , Mesoderma/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Osteogênese , Pele/metabolismo
11.
Development ; 145(2)2018 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-29378823

RESUMO

Mesenchymal stem cells (MSCs) are somatic stem cells that can be derived from adult bone marrow (BM) and white adipose tissue (WAT), and that display multipotency and self-renewal capacity. Although MSCs are essential for tissue formation and have already been used in clinical therapy, the origins and markers of these cells remain unknown. In this study, we first investigated the developmental process of MSCs in mouse embryos using the gene encoding platelet-derived growth factor receptor α (Pdgfra) as a marker. We then traced cells expressing Pdgfra and other genes (brachyury, Sox1 and Pmx1) in various mutant mouse embryos until the adult stage. This tracing of MSC origins and destinies indicates that embryonic MSCs emerge in waves and that almost all adult BM MSCs and WAT MSCs originate from mesoderm and embryonic Pdgfrα-positive cells. Furthermore, we demonstrate that adult Pdgfrα-positive cells are involved in some pathological conditions.


Assuntos
Tecido Adiposo Branco/citologia , Tecido Adiposo Branco/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Tecido Adiposo Branco/crescimento & desenvolvimento , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Feminino , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Gravidez , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
12.
Sci Rep ; 8(1): 1634, 2018 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-29374279

RESUMO

Recently, we reported that bacterial incorporation induces cellular transdifferentiation of human fibroblasts. However, the bacterium-intrinsic cellular- transdifferentiation factor remained unknown. Here, we found that cellular transdifferentiation is caused by ribosomes. Ribosomes, isolated from both prokaryotic and eukaryotic cells, induce the formation of embryoid body-like cell clusters. Numerous ribosomes are incorporated into both the cytoplasm and nucleus through trypsin-activated endocytosis, which leads to cell-cluster formation. Although ribosome-induced cell clusters (RICs) express several stemness markers and differentiate into derivatives of all three germ layers in heterogeneous cell populations, RICs fail to proliferate, alter the methylation states of pluripotent genes, or contribute to teratoma or chimera formation. However, RICs express markers of epithelial-mesenchymal transition without altering the cell cycle, despite their proliferation obstruction. These findings demonstrate that incorporation of ribosomes into host cells induces cell transdifferentiation and alters cellular plasticity.


Assuntos
Transdiferenciação Celular , Fibroblastos/fisiologia , Ribossomos/metabolismo , Bactérias/metabolismo , Células Cultivadas , Endocitose , Humanos
13.
Sci Transl Med ; 9(391)2017 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-28539470

RESUMO

Amyotrophic lateral sclerosis (ALS), a fatal disease causing progressive loss of motor neurons, still has no effective treatment. We developed a phenotypic screen to repurpose existing drugs using ALS motor neuron survival as readout. Motor neurons were generated from induced pluripotent stem cells (iPSCs) derived from an ALS patient with a mutation in superoxide dismutase 1 (SOD1). Results of the screen showed that more than half of the hits targeted the Src/c-Abl signaling pathway. Src/c-Abl inhibitors increased survival of ALS iPSC-derived motor neurons in vitro. Knockdown of Src or c-Abl with small interfering RNAs (siRNAs) also rescued ALS motor neuron degeneration. One of the hits, bosutinib, boosted autophagy, reduced the amount of misfolded mutant SOD1 protein, and attenuated altered expression of mitochondrial genes. Bosutinib also increased survival in vitro of ALS iPSC-derived motor neurons from patients with sporadic ALS or other forms of familial ALS caused by mutations in TAR DNA binding protein (TDP-43) or repeat expansions in C9orf72 Furthermore, bosutinib treatment modestly extended survival of a mouse model of ALS with an SOD1 mutation, suggesting that Src/c-Abl may be a potentially useful target for developing new drugs to treat ALS.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Mutação/genética , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
14.
Stem Cells ; 35(5): 1162-1175, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28100021

RESUMO

Induction of pluripotency with defined factors (octamer-binding transcription factor 4 [Oct4], SRY (sex determining region Y)-box 2 [Sox2], Kruppel-like factor 4 [Klf4], c-Myc) raises hopes for successful clinical trials. Despite considerable efforts, the molecular mechanism of reprogramming remains poorly understood. The aim of the present study was to identify the role of calcineurin/nuclear factor of activated T cells (NFAT) in reprogramming. Our results demonstrated a biphasic role for calcineurin/NFAT signaling during reprogramming. In the early phase of reprogramming, calcineurin activity is required to maintain proper cell cycle division and for mesenchymal-epithelial transition. In the late phase, calcineurin exerts a negative effect that is mediated by NFATc2. NFATc2 interacts with Hdac3, Ezh2, and Suv39h1 to increase H3K9me3 and H3K27me3 over the Sox2 enhancer and Klf2 promoter, respectively, resulting in the downregulation of their expression. Moreover, Gαq was identified as a positive upstream regulator for calcineurin. The Gαq/calcineurin/NFATc2 axis negatively regulates the late step of reprogramming. By inhibiting NFATc2 or calcineurin, induced pluripotent stem cells could be established without exogenous Sox2. Thus, the present study revealed another regulatory level of reprogramming, and proposes a biological axis that could be useful for cancer therapy. Stem Cells 2017;35:1162-1175.


Assuntos
Calcineurina/metabolismo , Reprogramação Celular , Fatores de Transcrição NFATC/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Animais , Biomarcadores/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Técnicas de Silenciamento de Genes , Heterocromatina/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Receptores Acoplados a Proteínas G/metabolismo
15.
Cell Stem Cell ; 19(5): 587-592, 2016 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-27814480

RESUMO

Cell types more advanced in development than embryonic stem cells, such as EpiSCs, fail to contribute to chimeras when injected into pre-implantation-stage blastocysts, apparently because the injected cells undergo apoptosis. Here we show that transient promotion of cell survival through expression of the anti-apoptotic gene BCL2 enables EpiSCs and Sox17+ endoderm progenitors to integrate into blastocysts and contribute to chimeric embryos. Upon injection into blastocyst, BCL2-expressing EpiSCs contributed to all bodily tissues in chimeric animals while Sox17+ endoderm progenitors specifically contributed in a region-specific fashion to endodermal tissues. In addition, BCL2 expression enabled rat EpiSCs to contribute to mouse embryonic chimeras, thereby forming interspecies chimeras that could survive to adulthood. Our system therefore provides a method to overcome cellular compatibility issues that typically restrict chimera formation. Application of this type of approach could broaden the use of embryonic chimeras, including region-specific chimeras, for basic developmental biology research and regenerative medicine.


Assuntos
Apoptose , Quimera/metabolismo , Embrião de Mamíferos/citologia , Animais , Células-Tronco Embrionárias/citologia , Camundongos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fatores de Transcrição SOX/metabolismo
16.
J Dermatol Sci ; 84(2): 186-196, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27510999

RESUMO

BACKGROUND: We recently generated induced pluripotent stem cells (iPSCs) from cultured dermal fibroblasts of systemic sclerosis (SSc-iPSC) to study the disease mechanisms. OBJECTIVE: In the present study, we have performed gene expression analysis using cultured SSc dermal fibroblasts, SSc-iPSC, and fibroblasts re-differentiated from SSc-iPSC (SSc-iPSC-FB). METHODS: mRNA and protein levels of collagen and integrins were analyzed using PCR array, PCR, immunoblotting, and immunofluorescence. RESULTS: We compared expression pattern of TGF-ß-related genes between normal iPSC (NS-iPSC) and SSc-iPSC by PCR array, and found constitutive and significant down-regulation of S100A8, Smad6, and TGF-ß2 in SSc-iPSC. The expression of these genes was not altered in cultured SSc fibroblasts or SSc-iPSC-FB compared to NS fibroblasts or NS-iPSC-FB, respectively. On the other hand, the expression of collagen, integrin α and ß was up-regulated in SSc fibroblasts, while SSc-iPSC-FB showed normalized levels of collagen and integrin ß. CONCLUSIONS: So far, there have been no reports investigating disease-derived iPSCs of SSc. Our results suggest that S100A8, Smad6, and TGF-ß2 may be the key molecules of this disease. On the other hand, the normalization of collagen and integrins by iPSC reprogramming suggests that epigenetic modifications of genes may play a role in the mechanism of collagen accumulation seen in SSc fibroblasts, and that gene reprogramming may become novel therapeutic approach. As the limitation of this study, we established only one iPSC line from each patient, which may not be enough to discuss disease-specific phenotypes. Larger studies including increased number of iPSC lines are needed in the future.


Assuntos
Perfilação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/citologia , Escleroderma Sistêmico/metabolismo , Idoso , Animais , Biópsia , Antígenos CD18/metabolismo , Calgranulina A/metabolismo , Diferenciação Celular , Colágeno/metabolismo , Fibroblastos/metabolismo , Humanos , Camundongos , Pessoa de Meia-Idade , Fenótipo , Reação em Cadeia da Polimerase , Escleroderma Sistêmico/genética , Análise de Sequência de RNA , Pele/metabolismo , Pele/patologia , Proteína Smad6/metabolismo , Teratoma/metabolismo , Fator de Crescimento Transformador beta2/metabolismo
17.
Methods Mol Biol ; 1357: 403-13, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25520286

RESUMO

Human induced pluripotent stem (hiPS) cells are very attractive tools for modeling diseases and regenerative medicine. However, to achieve them, the efficient differentiation methods of hiPS cells into aimed cell type in vitro are necessary. Because mesoderm cells are useful in particular, we have developed the differentiation of mouse embryonic stem (mES) cells into mesoderm cells previously. In this time, these methods were improved for hiPS cells and now human mesoderm cells are able to be obtained efficiently. It is certain that the new methods are applicable to various studies and therapies.


Assuntos
Técnicas de Cultura de Células/métodos , Técnicas de Reprogramação Celular/métodos , Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Mesoderma/citologia , Animais , Antígenos de Diferenciação/análise , Diferenciação Celular , Linhagem da Célula , Condrogênese , Ensaio de Unidades Formadoras de Colônias , Meios de Cultura/farmacologia , Fibroblastos/citologia , Humanos , Mesoderma/química , Camundongos , Osteogênese , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/análise , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/análise
18.
Anticancer Res ; 35(8): 4479-85, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26168490

RESUMO

BACKGROUND/AIM: Neuroblastoma is a pediatric solid tumor refractory to eradication by chemotherapy. To determine whether Aloe vera (AV), a pontial anticancer reagent, could be useful in neuroblastoma therapy, we investigated the anti-proliferative effects of an AV protein extract. MATERIALS AND METHODS: Human neuroblastoma cell lines (IMR-32, TGW, CHP-126 and NBL-S) were cultured with AV protein extract and proliferation status was assessed by cell counting, Ki-67 staining and gene expression. RESULTS: Among tested lines, the number of viable, AV-treated IMR-32 cells significantly decreased 1.98-fold by day 2 and 1.33-fold by day 5 of culture relative to untreated controls (p<0.05). Treatment also decreased the number of Ki-67(+) IMR-32 cells by 13% by day 5 (p<0.05) and, unlike untreated controls, CCND2 mRNA expression levels became undetectable by day 1. CONCLUSION: AV-protein extract suppresses human IMR-32 neuroblastoma cell proliferation, possibly by suppressing CCND2 transcript levels in vitro.


Assuntos
Aloe/metabolismo , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Extratos Vegetais/farmacologia , Animais , Linhagem Celular Tumoral , Ciclina D2/genética , Humanos , Camundongos , RNA Mensageiro/biossíntese
19.
Genesis ; 53(5): 329-36, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25884589

RESUMO

Platelet-derived growth factor (PDGF) and its receptor play an important role in embryogenesis. PDGF receptor α (PDGFRα) is expressed specifically in the embryonic day 7.5 (E7.5) mesoderm and in the E9.5 neural crest among other tissues. PDGFRα-expressing cells and their descendants are involved in the formation of various tissues. To trace PDGFRα-expressing cells in vivo, we generated a knock-in mouse line that expressed a fusion protein of green fluorescent protein (GFP), Cre recombinase (Cre), and mutated estrogen receptor ligand-binding domain (ERT2) under the control of the PDGFRα promoter. In these mice, Cre activity in PDGFRα-expressing cells could be induced by tamoxifen treatment. Taken together, our results suggest that the knock-in mouse line generated here could be useful for studying PDGFRα-expressing cells and their descendants in vivo at various stages of development.


Assuntos
Proteínas de Fluorescência Verde/genética , Integrases/genética , Domínios e Motivos de Interação entre Proteínas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptores de Estrogênio/genética , Animais , Imunofluorescência , Expressão Gênica , Técnicas de Introdução de Genes , Ordem dos Genes , Marcação de Genes , Genes Reporter , Loci Gênicos , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Transgênicos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Estrogênio/química , Transgenes
20.
Stem Cell Res ; 12(2): 574-83, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24531302

RESUMO

Familial amyloidotic polyneuropathy (FAP) is a hereditary amyloidosis induced by amyloidogenic transthyretin (ATTR). Because most transthyretin (TTR) in serum is synthesized by the liver, liver transplantation (LT) is today the only treatment available to halt the progression of FAP, even though LT is associated with several problems. Despite the urgent need to develop alternatives to LT, the detailed pathogenesis of FAP is still unknown; also, no model fully represents the relevant processes in patients with FAP. The induction of induced pluripotent stem (iPS) cells has allowed development of pluripotent cells specific for patients and has led to useful models of human diseases. Because of the need for a tool to elucidate the molecular pathogenesis of FAP, in this study we sought to establish heterozygous ATTR mutant iPS cells, and were successful, by using a Sendai virus vector mixture containing four transcription factors (Oct3/4, Sox2, Klf4, and c-Myc) to reprogram dermal fibroblasts derived from FAP patients. Moreover, FAP-specific iPS cells had the potential to differentiate into hepatocyte-like cells and indeed expressed ATTR. FAP-specific iPS cells demonstrated the possibility of serving as a pathological tool that will contribute to understanding the pathogenesis of FAP and development of FAP treatments.


Assuntos
Neuropatias Amiloides Familiares/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Pré-Albumina/metabolismo , Neuropatias Amiloides Familiares/genética , Animais , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA