Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genes Cells ; 28(12): 845-856, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37844904

RESUMEN

Cytokinesis, the final process of cell division, involves the accumulation of actin and myosin II filaments at the cell's equator, forming a contractile ring that facilitates the division into two daughter cells. While light microscopy has provided valuable insights into the molecular mechanism of this process, it has limitations in examining individual filaments in vivo. In this study, we utilized transmission electron microscopy to observe actin and myosin II filaments in the contractile rings of dividing Dictyostelium cells. To synchronize cytokinesis, we developed a novel method that allowed us to visualize dividing cells undergoing cytokinesis with a frequency as high as 18%. This improvement enabled us to examine the lengths and alignments of individual filaments within the contractile rings. As the furrow constricted, the length of actin filaments gradually decreased. Moreover, both actin and myosin II filaments reoriented perpendicularly to the long axis during furrow constriction. Through experiments involving myosin II null cells, we discovered that myosin II plays a role in regulating both the lengths and alignments of actin filaments. Additionally, dynamin-like protein A was found to contribute to regulating the length of actin filaments, while cortexillins were involved in regulating their alignment.


Asunto(s)
Actomiosina , Dictyostelium , Actomiosina/metabolismo , Actinas/metabolismo , Dictyostelium/metabolismo , Citoesqueleto de Actina/metabolismo , Citocinesis , Miosina Tipo II/metabolismo
2.
Stem Cell Reports ; 18(11): 2071-2083, 2023 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-37832542

RESUMEN

UGT1A1 (UDP glucuronosyltransferase family 1 member A1) is the primary enzyme required for bilirubin conjugation, which is essential for preventing hyperbilirubinemia. Animal models lack key human organic anion transporting polypeptides with distinct epigenetic control over bilirubin metabolism, necessitating a human model to interrogate the regulatory mechanism behind UGT1A1 function. Here, we use induced pluripotent stem cells to develop human liver organoids that can emulate conjugation failure phenotype. Bilirubin conjugation assays, chromatin immunoprecipitation, and transcriptome analysis elucidated the role of glucocorticoid antagonism in UGT1A1 activation. This antagonism prevents the binding of transcriptional repressor MECP2 at the expense of NRF2 with associated off-target effects. Therefore, we introduced functional GULO (L-gulonolactone oxidase) in human organoids to augment intracellular ascorbate for NRF2 reactivation. This engineered organoid conjugated more bilirubin and protected against hyperbilirubinemia when transplanted in immunosuppressed Crigler-Najjar syndrome rat model. Collectively, we demonstrate that our organoid system serves as a manipulatable model for interrogating hyperbilirubinemia and potential therapeutic development.


Asunto(s)
Síndrome de Crigler-Najjar , Células Madre Pluripotentes , Humanos , Animales , Ratas , Bilirrubina/farmacología , Bilirrubina/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Hígado/metabolismo , Síndrome de Crigler-Najjar/genética , Síndrome de Crigler-Najjar/terapia , Hiperbilirrubinemia/genética , Hiperbilirrubinemia/metabolismo , Hiperbilirrubinemia/terapia , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Células Madre Pluripotentes/metabolismo
3.
Commun Biol ; 6(1): 854, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37770589

RESUMEN

Alport syndrome (AS) is a hereditary glomerulonephritis caused by COL4A3, COL4A4 or COL4A5 gene mutations and characterized by abnormalities of glomerular basement membranes (GBMs). Due to a lack of curative treatments, the condition proceeds to end-stage renal disease even in adolescents. Hampering drug discovery is the absence of effective in vitro methods for testing the restoration of normal GBMs. Here, we aimed to develop kidney organoid models from AS patient iPSCs for this purpose. We established iPSC-derived collagen α5(IV)-expressing kidney organoids and confirmed that kidney organoids from COL4A5 mutation-corrected iPSCs restore collagen α5(IV) protein expression. Importantly, our model recapitulates the differences in collagen composition between iPSC-derived kidney organoids from mild and severe AS cases. Furthermore, we demonstrate that a chemical chaperone, 4-phenyl butyric acid, has the potential to correct GBM abnormalities in kidney organoids showing mild AS phenotypes. This iPSC-derived kidney organoid model will contribute to drug discovery for AS.


Asunto(s)
Células Madre Pluripotentes Inducidas , Nefritis Hereditaria , Adolescente , Humanos , Nefritis Hereditaria/genética , Nefritis Hereditaria/metabolismo , Nefritis Hereditaria/terapia , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Riñón/metabolismo , Membrana Basal Glomerular
4.
Nat Commun ; 14(1): 804, 2023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36808132

RESUMEN

Induced pluripotent stem cells (iPSCs) are a promising resource for allogeneic cartilage transplantation to treat articular cartilage defects that do not heal spontaneously and often progress to debilitating conditions, such as osteoarthritis. However, to the best of our knowledge, allogeneic cartilage transplantation into primate models has never been assessed. Here, we show that allogeneic iPSC-derived cartilage organoids survive and integrate as well as are remodeled as articular cartilage in a primate model of chondral defects in the knee joints. Histological analysis revealed that allogeneic iPSC-derived cartilage organoids in chondral defects elicited no immune reaction and directly contributed to tissue repair for at least four months. iPSC-derived cartilage organoids integrated with the host native articular cartilage and prevented degeneration of the surrounding cartilage. Single-cell RNA-sequence analysis indicated that iPSC-derived cartilage organoids differentiated after transplantation, acquiring expression of PRG4 crucial for joint lubrication. Pathway analysis suggested the involvement of SIK3 inactivation. Our study outcomes suggest that allogeneic transplantation of iPSC-derived cartilage organoids may be clinically applicable for the treatment of patients with chondral defects of the articular cartilage; however further assessment of functional recovery long term after load bearing injuries is required.


Asunto(s)
Cartílago Articular , Trasplante de Células Madre Hematopoyéticas , Células Madre Pluripotentes Inducidas , Animales , Cartílago Articular/patología , Primates , Organoides , Condrocitos
5.
Nat Commun ; 14(1): 488, 2023 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-36717582

RESUMEN

Induced pluripotent stem cell (iPSC) reprogramming is inefficient and understanding the molecular mechanisms underlying this inefficiency holds the key to successfully control cellular identity. Here, we report 24 reprogramming roadblock genes identified by CRISPR/Cas9-mediated genome-wide knockout (KO) screening. Of these, depletion of the predicted KRAB zinc finger protein (KRAB-ZFP) Zfp266 strongly and consistently enhances murine iPSC generation in several reprogramming settings, emerging as the most robust roadblock. We show that ZFP266 binds Short Interspersed Nuclear Elements (SINEs) adjacent to binding sites of pioneering factors, OCT4 (POU5F1), SOX2, and KLF4, and impedes chromatin opening. Replacing the KRAB co-suppressor with co-activator domains converts ZFP266 from an inhibitor to a potent facilitator of iPSC reprogramming. We propose that the SINE-KRAB-ZFP interaction is a critical regulator of chromatin accessibility at regulatory elements required for efficient cellular identity changes. In addition, this work serves as a resource to further illuminate molecular mechanisms hindering reprogramming.


Asunto(s)
Células Madre Pluripotentes Inducidas , Dedos de Zinc , Animales , Ratones , Reprogramación Celular/genética , Cromatina/genética , Cromatina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel
6.
Stem Cell Res ; 65: 102953, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36334578

RESUMEN

Glucose-6-phosphate dehydrogenase (G6PD) deficiency is a common genetic enzyme-defect disorder. In this study, CHULAi001-A was established from peripheral blood mononuclear cells of a G6PD-deficient subject who carried the Viangchan variant (871 G > A). Episomal plasmids expressing OCT3/4, SOX2, KLF4, L-MYC, LIN28, and shRNA against p53 were introduced into parental cells by electroporation and cultured under feeder-free conditions to reprogram iPSCs. Embryonic pluripotency, in vitro differentiation capacity, and episomal vector integration of the established CHULAi001-A were verified. The CHULAi001-A iPSCs retained a normal karyotype.


Asunto(s)
Glucosafosfato Deshidrogenasa , Células Madre Pluripotentes Inducidas , Humanos , Glucosafosfato Deshidrogenasa/genética , Leucocitos Mononucleares , Mutación/genética
7.
Stem Cells Dev ; 31(9-10): 250-257, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35316100

RESUMEN

Induced pluripotent stem cells (iPSCs) can serve as a biological resource for functional and conservation research for various species. This realization has led to the generation of iPSCs from many species, including those identified as endangered. However, the understanding of species variation in mammalian iPSCs remains largely unknown. To gain insight into species variation in iPSCs, we generated iPSCs from a new species Grevy's zebra (Equus grevyi; gz-iPSCs), which has been listed as endangered in the IUCN (International Union for Conservation of Nature) Red List. We isolated primary fibroblast cells from an individual and successfully reprogrammed them into iPSCs. The generated gz-iPSCs continued to grow under primed-type culture condition and showed pluripotency and differentiation potential. To describe the molecular characteristics of gz-iPSCs, we performed RNA sequencing analysis. The gz-iPSC transcriptome showed robust expression of pluripotency-associated genes reported in human and mouse, suggesting evolutionary conservation among the species. This study provides insight into the iPSCs from a rare species and helps the understanding of the gene expression basis underlying mammalian pluripotent stem cells.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Animales , Diferenciación Celular/genética , Reprogramación Celular , Equidae/genética , Ratones , Transcriptoma/genética
8.
Cell Rep ; 37(5): 109909, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34731633

RESUMEN

Human induced pluripotent stem cells (hiPSCs) show variable differentiation potential due to their epigenomic heterogeneity, whose extent/attributes remain unclear, except for well-studied elements/chromosomes such as imprints and the X chromosomes. Here, we show that seven hiPSC lines with variable germline potential exhibit substantial epigenomic heterogeneity, despite their uniform transcriptomes. Nearly a quarter of autosomal regions bear potentially differential chromatin modifications, with promoters/CpG islands for H3K27me3/H2AK119ub1 and evolutionarily young retrotransposons for H3K4me3. We identify 145 large autosomal blocks (≥100 kb) with differential H3K9me3 enrichment, many of which are lamina-associated domains (LADs) in somatic but not in embryonic stem cells. A majority of these epigenomic heterogeneities are independent of genetic variations. We identify an X chromosome state with chromosome-wide H3K9me3 that stably prevents X chromosome erosion. Importantly, the germline potential of female hiPSCs correlates with X chromosome inactivation. We propose that inherent genomic properties, including CpG density, transposons, and LADs, engender epigenomic heterogeneity in hiPSCs.


Asunto(s)
Cromosomas Humanos X , Epigénesis Genética , Epigenoma , Heterogeneidad Genética , Histonas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Inactivación del Cromosoma X , Diferenciación Celular , Línea Celular , Ensamble y Desensamble de Cromatina , Islas de CpG , Elementos Transponibles de ADN , Epigenómica , Evolución Molecular , Humanos , Metilación , Lámina Nuclear/genética , Lámina Nuclear/metabolismo , Procesamiento Proteico-Postraduccional , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
9.
Biology (Basel) ; 10(10)2021 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-34681161

RESUMEN

The SCN5A R1623Q mutation is one of the most common genetic variants associated with severe congenital long QT syndrome 3 (LQT3) in fetal and neonatal patients. To investigate the properties of the R1623Q mutation, we established an induced pluripotent stem cell (iPSC) cardiomyocyte (CM) model from a patient with LQTS harboring a heterozygous R1623Q mutation. The properties and pharmacological responses of iPSC-CMs were characterized using a multi-electrode array system. The biophysical characteristic analysis revealed that R1623Q increased open probability and persistent currents of sodium channel, indicating a gain-of-function mutation. In the pharmacological study, mexiletine shortened FPDcF in R1623Q-iPSC-CMs, which exhibited prolonged field potential duration corrected by Fridericia's formula (FPDcF, analogous to QTcF). Meanwhile, E4031, a specific inhibitor of human ether-a-go-go-related gene (hERG) channel, significantly increased the frequency of arrhythmia-like early after depolarization (EAD) events. These characteristics partly reflect the patient phenotypes. To further analyze the effect of neonatal isoform, which is predominantly expressed in the fetal period, on the R1623Q mutant properties, we transfected adult form and neonatal isoform SCN5A of control and R1623Q mutant SCN5A genes to 293T cells. Whole-cell automated patch-clamp recordings revealed that R1623Q increased persistent Na+ currents, indicating a gain-of-function mutation. Our findings demonstrate the utility of LQT3-associated R1623Q mutation-harboring iPSC-CMs for assessing pharmacological responses to therapeutic drugs and improving treatment efficacy. Furthermore, developmental switching of neonatal/adult Nav1.5 isoforms may be involved in the pathological mechanisms underlying severe long QT syndrome in fetuses and neonates.

10.
Sci Rep ; 11(1): 9123, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33907292

RESUMEN

PAX2 is a transcription factor essential for kidney development and the main causative gene for renal coloboma syndrome (RCS). The mechanisms of PAX2 action during kidney development have been evaluated in mice but not in humans. This is a critical gap in knowledge since important differences have been reported in kidney development in the two species. In the present study, we hypothesized that key human PAX2-dependent kidney development genes are differentially expressed in nephron progenitor cells from induced pluripotent stem cells (iPSCs) in patients with RCS relative to healthy individuals. Cap analysis of gene expression revealed 189 candidate promoters and 71 candidate enhancers that were differentially activated by PAX2 in this system in three patients with RCS with PAX2 mutations. By comparing this list with the list of candidate Pax2-regulated mouse kidney development genes obtained from the Functional Annotation of the Mouse/Mammalian (FANTOM) database, we prioritized 17 genes. Furthermore, we ranked three genes-PBX1, POSTN, and ITGA9-as the top candidates based on closely aligned expression kinetics with PAX2 in the iPSC culture system and susceptibility to suppression by a Pax2 inhibitor in cultured mouse embryonic kidney explants. Identification of these genes may provide important information to clarify the pathogenesis of RCS, human kidney development, and kidney regeneration.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Riñón/crecimiento & desarrollo , Factor de Transcripción PAX2/genética , Adulto , Animales , Moléculas de Adhesión Celular/genética , Linaje de la Célula , Coloboma/patología , Femenino , Humanos , Células Madre Pluripotentes Inducidas , Integrinas/genética , Riñón/citología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Persona de Mediana Edad , Nefronas/citología , Nefronas/fisiología , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Insuficiencia Renal/patología
12.
Sci Rep ; 10(1): 14859, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32908229

RESUMEN

Shwachman-Diamond syndrome (SDS), an autosomal recessive disorder characterized by bone marrow failure, exocrine pancreatic insufficiency, and skeletal abnormalities, is caused by mutations in the Shwachman-Bodian-Diamond syndrome (SBDS) gene, which plays a role in ribosome biogenesis. Although the causative genes of congenital disorders frequently involve regulation of embryogenesis, the role of the SBDS gene in early hematopoiesis remains unclear, primarily due to the lack of a suitable experimental model for this syndrome. In this study, we established induced pluripotent stem cells (iPSCs) from patients with SDS (SDS-iPSCs) and analyzed their in vitro hematopoietic and endothelial differentiation potentials. SDS-iPSCs generated hematopoietic and endothelial cells less efficiently than iPSCs derived from healthy donors, principally due to the apoptotic predisposition of KDR+CD34+ common hemoangiogenic progenitors. By contrast, forced expression of SBDS gene in SDS-iPSCs or treatment with a caspase inhibitor reversed the deficiency in hematopoietic and endothelial development, and decreased apoptosis of their progenitors, mainly via p53-independent mechanisms. Patient-derived iPSCs exhibited the hematological abnormalities associated with SDS even at the earliest hematopoietic stages. These findings will enable us to dissect the pathogenesis of multiple disorders associated with ribosomal dysfunction.


Asunto(s)
Diferenciación Celular , Células Endoteliales , Hematopoyesis , Células Madre Hematopoyéticas , Células Madre Pluripotentes Inducidas , Síndrome de Shwachman-Diamond , Apoptosis/genética , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Japón , Masculino , Mutación , Proteínas/genética , Síndrome de Shwachman-Diamond/metabolismo , Síndrome de Shwachman-Diamond/patología
13.
Biochem Biophys Res Commun ; 529(4): 1186-1194, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32819584

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is a hereditary disorder which manifests progressive renal cyst formation and leads to end-stage kidney disease. Around 85% of cases are caused by PKD1 heterozygous mutations, exhibiting relatively poorer renal outcomes than those with mutations in other causative gene PKD2. Although many disease models have been proposed for ADPKD, the pre-symptomatic pathology of the human disease remains unknown. To unveil the mechanisms of early cytogenesis, robust and genetically relevant human models are needed. Here, we report a novel ADPKD model using kidney organoids derived from disease-specific human induced pluripotent stem cells (hiPSCs). Importantly, we found that kidney organoids differentiated from gene-edited heterozygous PKD1-mutant as well as ADPKD patient-derived hiPSCs can reproduce renal cysts. Further, we demonstrated the possibility of ADPKD kidney organoids serving as drug screening platforms. This newly developed model will contribute to identifying novel therapeutic targets, extending the field of ADPKD research.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Riñón/patología , Modelos Biológicos , Organoides/patología , Riñón Poliquístico Autosómico Dominante/patología , Secuencia de Aminoácidos , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Línea Celular , Colforsina/farmacología , Evaluación Preclínica de Medicamentos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Mutación/genética , Fenotipo , Canales Catiónicos TRPP/química , Canales Catiónicos TRPP/genética
14.
Stem Cell Res ; 45: 101815, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32361464

RESUMEN

Juvenile nephronophthisis is an inherited renal ciliopathy, causing cystic kidney disease, renal fibrosis, and end-stage renal failure. Human induced pluripotent stem cell (hiPSC) lines, derived from two Juvenile nephronophthisis patients, were generated from peripheral blood mononuclear cells by episomal plasmid vectors. Generated hiPSC lines showed self-renewal and pluripotency and carried a large deletion in NPHP1 (Nephrocystin 1) gene. Since the molecular pathogenesis caused by NPHP1 dysfunction remains unclear, these cell resources provide useful tools to establish disease models and to develop new therapies for juvenile nephronophthisis.


Asunto(s)
Células Madre Pluripotentes Inducidas , Proteínas Adaptadoras Transductoras de Señales , Proteínas del Citoesqueleto , Fibrosis , Humanos , Enfermedades Renales Quísticas/congénito , Leucocitos Mononucleares , Proteínas de la Membrana/genética
15.
Stem Cell Reports ; 12(5): 1113-1128, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31056481

RESUMEN

CpG islands (CGIs) including those at imprinting control regions (ICRs) are protected from de novo methylation in somatic cells. However, many cancers often exhibit CGI hypermethylation, implying that the machinery is impaired in cancer cells. Here, we conducted a comprehensive analysis of CGI methylation during somatic cell reprogramming. Although most CGIs remain hypomethylated, a small subset of CGIs, particularly at several ICRs, was often de novo methylated in reprogrammed pluripotent stem cells (PSCs). Such de novo ICR methylation was linked with the silencing of reprogramming factors, which occurs at a late stage of reprogramming. The ICR-preferred CGI hypermethylation was similarly observed in human PSCs. Mechanistically, ablation of Dnmt3a prevented PSCs from de novo ICR methylation. Notably, the ICR-preferred CGI hypermethylation was observed in pediatric cancers, while adult cancers exhibit genome-wide CGI hypermethylation. These results may have important implications in the pathogenesis of pediatric cancers and the application of PSCs.


Asunto(s)
Reprogramación Celular/genética , Metilación de ADN/genética , Impresión Genómica/genética , Células Madre Pluripotentes/metabolismo , Adulto , Animales , Células Cultivadas , Islas de CpG/genética , Epigénesis Genética/genética , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos ICR , Células Madre Pluripotentes/citología
16.
Cell Stem Cell ; 24(4): 566-578.e7, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30853558

RESUMEN

Induced pluripotent stem cells (iPSCs) have strong potential in regenerative medicine applications; however, immune rejection caused by HLA mismatching is a concern. B2M gene knockout and HLA-homozygous iPSC stocks can address this issue, but the former approach may induce NK cell activity and fail to present antigens, and it is challenging to recruit rare donors for the latter method. Here, we show two genome-editing strategies for making immunocompatible donor iPSCs. First, we generated HLA pseudo-homozygous iPSCs with allele-specific editing of HLA heterozygous iPSCs. Second, we generated HLA-C-retained iPSCs by disrupting both HLA-A and -B alleles to suppress the NK cell response while maintaining antigen presentation. HLA-C-retained iPSCs could evade T cells and NK cells in vitro and in vivo. We estimated that 12 lines of HLA-C-retained iPSCs combined with HLA-class II knockout are immunologically compatible with >90% of the world's population, greatly facilitating iPSC-based regenerative medicine applications.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica , Antígenos HLA/genética , Histocompatibilidad/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Línea Celular , Femenino , Antígenos HLA/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos NOD
17.
Physiol Rev ; 99(1): 79-114, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30328784

RESUMEN

The discovery of somatic cell nuclear transfer proved that somatic cells can carry the same genetic code as the zygote, and that activating parts of this code are sufficient to reprogram the cell to an early developmental state. The discovery of induced pluripotent stem cells (iPSCs) nearly half a century later provided a molecular mechanism for the reprogramming. The initial creation of iPSCs was accomplished by the ectopic expression of four specific genes (OCT4, KLF4, SOX2, and c-Myc; OSKM). iPSCs have since been acquired from a wide range of cell types and a wide range of species, suggesting a universal molecular mechanism. Furthermore, cells have been reprogrammed to iPSCs using a myriad of methods, although OSKM remains the gold standard. The sources for iPSCs are abundant compared with those for other pluripotent stem cells; thus the use of iPSCs to model the development of tissues, organs, and other systems of the body is increasing. iPSCs also, through the reprogramming of patient samples, are being used to model diseases. Moreover, in the 10 years since the first report, human iPSCs are already the basis for new cell therapies and drug discovery that have reached clinical application. In this review, we examine the generation of iPSCs and their application to disease and development.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos , Células Madre Pluripotentes/clasificación , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Humanos , Factor 4 Similar a Kruppel
18.
Stem Cell Reports ; 11(1): 142-156, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-30008324

RESUMEN

We show that a human pluripotent stem cell (hPSC) population cultured on a low-adhesion substrate developed two hPSC subtypes with different colony morphologies: flat and domed. Notably, the dome-like cells showed higher active proliferation capacity and increased several pluripotent genes' expression compared with the flat monolayer cells. We further demonstrated that cell-matrix adhesion mediates the interaction between cell morphology and expression of KLF4 and KLF5 through a serum response factor (SRF)-based regulatory double loop. Our results provide a mechanistic view on the coupling among adhesion, stem cell morphology, and pluripotency, shedding light on the critical role of cell-matrix adhesion in the induction and maintenance of hPSC.


Asunto(s)
Uniones Célula-Matriz/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Biomarcadores , Adhesión Celular/genética , Diferenciación Celular , Proliferación Celular , Autorrenovación de las Células/genética , Expresión Génica , Humanos , Inmunofenotipificación , Cariotipo , Factor 4 Similar a Kruppel , Modelos Biológicos
19.
Nat Commun ; 9(1): 1387, 2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29643333

RESUMEN

Multicellular organisms consist of multiple cell types. The identity of these cells is primarily maintained by cell-type-specific gene expression programs; however, mechanisms that suppress these programs are poorly defined. Here we show that serum response factor (Srf), a transcription factor that is activated by various extracellular stimuli, can repress cell-type-specific genes and promote cellular reprogramming to pluripotency. Manipulations that decrease ß-actin monomer quantity result in the nuclear accumulation of Mkl1 and the activation of Srf, which downregulate cell-type-specific genes and alter the epigenetics of regulatory regions and chromatin organization. Mice overexpressing Srf exhibit various pathologies including an ulcerative colitis-like symptom and a metaplasia-like phenotype in the pancreas. Our results demonstrate an unexpected function of Srf via a mechanism by which extracellular stimuli actively destabilize cell identity and suggest Srf involvement in a wide range of diseases.


Asunto(s)
Cromatina/química , Colitis Ulcerosa/genética , Células Madre Pluripotentes Inducidas/metabolismo , Metaplasia/genética , Células-Madre Neurales/metabolismo , Páncreas/metabolismo , Factor de Respuesta Sérica/genética , Actinas/genética , Actinas/metabolismo , Animales , Diferenciación Celular , Reprogramación Celular/genética , Cromatina/metabolismo , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/patología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Masculino , Metaplasia/metabolismo , Metaplasia/patología , Ratones , Ratones Transgénicos , Células-Madre Neurales/citología , Páncreas/patología , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Transactivadores/genética , Transactivadores/metabolismo
20.
Biochem Biophys Res Commun ; 497(2): 719-725, 2018 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-29462620

RESUMEN

AK2 is an adenylate phosphotransferase that localizes at the intermembrane spaces of the mitochondria, and its mutations cause a severe combined immunodeficiency with neutrophil maturation arrest named reticular dysgenesis (RD). Although the dysfunction of hematopoietic stem cells (HSCs) has been implicated, earlier developmental events that affect the fate of HSCs and/or hematopoietic progenitors have not been reported. Here, we used RD-patient-derived induced pluripotent stem cells (iPSCs) as a model of AK2-deficient human cells. Hematopoietic differentiation from RD-iPSCs was profoundly impaired. RD-iPSC-derived hemoangiogenic progenitor cells (HAPCs) showed decreased ATP distribution in the nucleus and altered global transcriptional profiles. Thus, AK2 has a stage-specific role in maintaining the ATP supply to the nucleus during hematopoietic differentiation, which affects the transcriptional profiles necessary for controlling the fate of multipotential HAPCs. Our data suggest that maintaining the appropriate energy level of each organelle by the intracellular redistribution of ATP is important for controlling the fate of progenitor cells.


Asunto(s)
Adenosina Trifosfato/metabolismo , Adenilato Quinasa/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/patología , Células Madre Pluripotentes Inducidas/patología , Leucopenia/patología , Inmunodeficiencia Combinada Grave/patología , Adenilato Quinasa/genética , Células Cultivadas , Metabolismo Energético , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Leucopenia/genética , Leucopenia/metabolismo , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA