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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 571(7766): 515-520, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31341297

RESUMEN

The mitochondrial ADP/ATP carrier (AAC) is a major transport protein of the inner mitochondrial membrane. It exchanges mitochondrial ATP for cytosolic ADP and controls cellular production of ATP. In addition, it has been proposed that AAC mediates mitochondrial uncoupling, but it has proven difficult to demonstrate this function or to elucidate its mechanisms. Here we record AAC currents directly from inner mitochondrial membranes from various mouse tissues and identify two distinct transport modes: ADP/ATP exchange and H+ transport. The AAC-mediated H+ current requires free fatty acids and resembles the H+ leak via the thermogenic uncoupling protein 1 found in brown fat. The ADP/ATP exchange via AAC negatively regulates the H+ leak, but does not completely inhibit it. This suggests that the H+ leak and mitochondrial uncoupling could be dynamically controlled by cellular ATP demand and the rate of ADP/ATP exchange. By mediating two distinct transport modes, ADP/ATP exchange and H+ leak, AAC connects coupled (ATP production) and uncoupled (thermogenesis) energy conversion in mitochondria.


Asunto(s)
Mitocondrias/metabolismo , Translocasas Mitocondriales de ADP y ATP/metabolismo , Protones , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Coenzimas/metabolismo , Ácidos Grasos/metabolismo , Transporte Iónico , Masculino , Ratones , Consumo de Oxígeno
2.
Hum Mol Genet ; 28(14): 2365-2377, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31267131

RESUMEN

MEGF10 myopathy is a rare inherited muscle disease that is named after the causative gene, MEGF10. The classic phenotype, early onset myopathy, areflexia, respiratory distress and dysphagia, is severe and immediately life-threatening. There are no disease-modifying therapies. We performed a small molecule screen and follow-up studies to seek a novel therapy. A primary in vitro drug screen assessed cellular proliferation patterns in Megf10-deficient myoblasts. Secondary evaluations were performed on primary screen hits using myoblasts derived from Megf10-/- mice, induced pluripotent stem cell-derived myoblasts from MEGF10 myopathy patients, mutant Drosophila that are deficient in the homologue of MEGF10 (Drpr) and megf10 mutant zebrafish. The screen yielded two promising candidates that are both selective serotonin reuptake inhibitors (SSRIs), sertraline and escitalopram. In depth follow-up analyses demonstrated that sertraline was highly effective in alleviating abnormalities across multiple models of the disease including mouse myoblast, human myoblast, Drosophila and zebrafish models. Sertraline also restored deficiencies of Notch1 in disease models. We conclude that SSRIs show promise as potential therapeutic compounds for MEGF10 myopathy, especially sertraline. The mechanism of action may involve the Notch pathway.


Asunto(s)
Proteínas de la Membrana/genética , Enfermedades Musculares/tratamiento farmacológico , Mioblastos/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Sertralina/uso terapéutico , Animales , Línea Celular , Movimiento Celular , Proliferación Celular , Citalopram/farmacología , Citalopram/uso terapéutico , Drosophila/efectos de los fármacos , Drosophila/genética , Evaluación Preclínica de Medicamentos , Humanos , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Mutación , Mioblastos/metabolismo , Receptor Notch1/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Sertralina/farmacología , Transducción de Señal , Pez Cebra/genética , Pez Cebra/metabolismo
3.
Am J Physiol Cell Physiol ; 319(5): C825-C838, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32783656

RESUMEN

Hypertension (HTN) is a polyfactorial disease that can manifest severe cardiovascular pathologies such as heart failure or stroke. Genome-wide association studies (GWAS) of HTN indicate that single-nucleotide polymorphisms (SNPs) contribute to increased risk for HTN and resistance to some HTN drug regimens (Hiltunen TP et al., J Am Heart Assoc 4: e001521, 2015; Le MT et al., PLoS One 8: e52062, 2013; McDonough CW et al., J Hypertens 31: 698-704, 2013; Vandell AG et al., Hypertension 60: 957-964, 2012). However, cellular mechanistic insights of such SNPs remain largely unknown. Using a bank of induced pluripotent stem cells (iPSCs) derived from patients with HTN and CRISPR/Cas9-mediated gene-editing approach, we investigated the effects of a female HTN risk-associated SNP (rs1154431) of the G protein-coupled estrogen receptor (GPER) (Bassuk SS, Manson JE., Clin Chem 60: 68-77, 2014) in vascular endothelial cells. Although GPER1 deletion reduced endothelial nitric oxide synthase (eNOS) activation in iPSC-derived endothelial cells (iECs), the polymorphism itself did not significantly affect eNOS and NO production in a comparison of isogenic hemizygous iECs expressing either normal (P16) or HTN-associated (L16) GPER. Interestingly, we demonstrate for the first time that GPER plays a role in regulation of adhesion molecule expression and monocyte adhesion to iECs. Moreover, the L16 iECs had higher expression of inflammation genes than P16 iECs, implying that the risk variant may affect carrier individuals through increased inflammatory activity. This study further indicates that iPSCs are a useful platform for exploring mechanistic insights underlying hypertension GWAS endeavors.


Asunto(s)
Células Endoteliales/metabolismo , Hipertensión/genética , Células Madre Pluripotentes Inducidas/metabolismo , Polimorfismo de Nucleótido Simple , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Adulto , Antígenos CD/genética , Antígenos CD/metabolismo , Secuencia de Bases , Sistemas CRISPR-Cas , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular , Diferenciación Celular , Ingeniería Celular/métodos , Células Endoteliales/patología , Femenino , Edición Génica/métodos , Regulación de la Expresión Génica , Humanos , Hipertensión/metabolismo , Hipertensión/fisiopatología , Células Madre Pluripotentes Inducidas/patología , Modelos Biológicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Cultivo Primario de Células , Receptores de Estrógenos/deficiencia , Receptores Acoplados a Proteínas G/deficiencia , Factores de Riesgo , Células THP-1 , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
4.
Biotechnol Bioeng ; 117(3): 816-831, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31814110

RESUMEN

Intracellular delivery of functional proteins is of great interest for basic biological research as well as for clinical applications. Transfection is the most commonly used method, however, it is not applicable to large-scale manipulation and inefficient in important cell types implicated in biomedical applications, such as epithelial, immune and pluripotent stem cells. In this study, we explored a bacterial type III secretion system (Bac-T3SS)-mediated proteofection method to overcome these limitations. An attenuated Pseudomonas aeruginosa vector was constructed, which has features of low toxicity, high T3SS activity, and self-limiting growth. Compared to the method of transfection, the Bac-T3SS showed significantly higher efficiencies of Cre recombinase translocation and target site recombination for hard-to-transfect human cell lines. Furthermore, through the delivery of ß-lactamase in live animals, we demonstrated the feasibility and biosafety of in vivo application of the Bac-T3SS. This study provided an efficient and low-cost proteofection strategy for laboratory use as well as for application in large-scale cell manipulations.


Asunto(s)
Integrasas/genética , Proteínas Recombinantes de Fusión/genética , Transfección/métodos , Sistemas de Secreción Tipo III/genética , Animales , Proteínas Bacterianas/genética , Reactores Biológicos , Línea Celular , Vectores Genéticos/genética , Humanos , Ratones , Células Madre Pluripotentes , Pseudomonas aeruginosa/genética , Proteínas Recombinantes de Fusión/metabolismo , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
5.
Mol Ther ; 26(11): 2617-2630, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30274788

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by a CTG nucleotide repeat expansion within the 3' UTR of the Dystrophia Myotonica protein kinase gene. In this study, we explored therapeutic genome editing using CRISPR/Cas9 via targeted deletion of expanded CTG repeats and targeted insertion of polyadenylation signals in the 3' UTR upstream of the CTG repeats to eliminate toxic RNA CUG repeats. We found paired SpCas9 or SaCas9 guide RNA induced deletion of expanded CTG repeats. However, this approach incurred frequent inversion in both the mutant and normal alleles. In contrast, the insertion of polyadenylation signals in the 3' UTR upstream of the CTG repeats eliminated toxic RNA CUG repeats, which led to phenotype reversal in differentiated neural stem cells, forebrain neurons, cardiomyocytes, and skeletal muscle myofibers. We concluded that targeted insertion of polyadenylation signals in the 3' UTR is a viable approach to develop therapeutic genome editing for DM1.


Asunto(s)
Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica/genética , Células-Madre Neurales/fisiología , Expansión de Repetición de Trinucleótido/genética , Regiones no Traducidas 3' , Sistemas CRISPR-Cas/genética , Diferenciación Celular/genética , Edición Génica/métodos , Terapia Genética/métodos , Células HEK293 , Humanos , Músculo Esquelético/crecimiento & desarrollo , Miocitos Cardíacos/fisiología , Distrofia Miotónica/patología , Distrofia Miotónica/terapia , Neuronas/fisiología , Señales de Poliadenilación de ARN 3'/genética , ARN Guía de Kinetoplastida , Transfección
6.
Stem Cells ; 35(3): 545-550, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28100040

RESUMEN

Recent advances in DNA sequencing technologies are revealing how human genetic variations associate with differential health risks, disease susceptibilities, and drug responses. Such information is now expected to help evaluate individual health risks, design personalized health plans and treat patients with precision. It is still challenging, however, to understand how such genetic variations cause the phenotypic alterations in pathobiologies and treatment response. Human induced pluripotent stem cell (iPSC) technologies are emerging as a promising strategy to fill the knowledge gaps between genetic association studies and underlying molecular mechanisms. Breakthroughs in genome editing technologies and continuous improvement in iPSC differentiation techniques are particularly making this research direction more realistic and practical. Pioneering studies have shown that iPSCs derived from a variety of monogenic diseases can faithfully recapitulate disease phenotypes in vitro when differentiated into disease-relevant cell types. It has been shown possible to partially recapitulate disease phenotypes, even with late onset and polygenic diseases. More recently, iPSCs have been shown to validate effects of disease and treatment-related single nucleotide polymorphisms identified through genome wide association analysis. In this review, we will discuss how iPSC research will further contribute to human health in the coming era of precision medicine. Stem Cells 2017;35:545-550.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Medicina de Precisión , Investigación con Células Madre , Animales , Estudio de Asociación del Genoma Completo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Fenotipo , Polimorfismo de Nucleótido Simple/genética
7.
Lab Invest ; 97(10): 1126-1132, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28759008

RESUMEN

Induced pluripotent stem cell (iPSC) technology was originally developed in 2006. Essentially, it converts somatic cells into pluripotent stem cells by transiently expressing a few transcriptional factors. Once generated, these iPSCs can differentiate into all the cell types of our body, theoretically, which has attracted great attention for clinical research including disease pathobiology studies. Could this technology then become an additional research or diagnostic tool widely available to practicing pathologists? Here we summarize progress in iPSC research toward disease pathobiology studies, its future potential, and remaining problems from a pathologist's perspective. A particular focus will be on introducing the effort to recapitulate disease-related morphological changes through three-dimensional culture of stem cells such as organoid differentiation.


Asunto(s)
Células Madre Pluripotentes Inducidas , Patología Clínica , Investigación con Células Madre , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Patólogos
8.
Mol Ther ; 24(8): 1378-87, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27203440

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by expanded Cytosine-Thymine-Guanine (CTG) repeats in the 3'-untranslated region (3' UTR) of the Dystrophia myotonica protein kinase (DMPK) gene, for which there is no effective therapy. The objective of this study is to develop genome therapy in human DM1 induced pluripotent stem (iPS) cells to eliminate mutant transcripts and reverse the phenotypes for developing autologous stem cell therapy. The general approach involves targeted insertion of polyA signals (PASs) upstream of DMPK CTG repeats, which will lead to premature termination of transcription and elimination of toxic mutant transcripts. Insertion of PASs was mediated by homologous recombination triggered by site-specific transcription activator-like effector nuclease (TALEN)-induced double-strand break. We found genome-treated DM1 iPS cells continue to maintain pluripotency. The insertion of PASs led to elimination of mutant transcripts and complete disappearance of nuclear RNA foci and reversal of aberrant splicing in linear-differentiated neural stem cells, cardiomyocytes, and teratoma tissues. In conclusion, genome therapy by insertion of PASs upstream of the expanded DMPK CTG repeats prevented the production of toxic mutant transcripts and reversal of phenotypes in DM1 iPS cells and their progeny. These genetically-treated iPS cells will have broad clinical application in developing autologous stem cell therapy for DM1.


Asunto(s)
Terapia Genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Trasplante de Células Madre , Animales , Diferenciación Celular , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Marcación de Gen , Sitios Genéticos , Humanos , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Distrofia Miotónica/terapia , Proteína Quinasa de Distrofia Miotónica/genética , Poli A , Unión Proteica , Empalme del ARN , Teratoma/genética , Teratoma/metabolismo , Teratoma/patología , Nucleasas de los Efectores Tipo Activadores de la Transcripción , Trasplante Autólogo , Repeticiones de Trinucleótidos
9.
Stem Cells ; 33(6): 1829-38, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25702800

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by expanded CTG repeats in the 3'-untranslated region (3' UTR) of the DMPK gene. Correcting the mutation in DM1 stem cells would be an important step toward autologous stem cell therapy. The objective of this study is to demonstrate in vitro genome editing to prevent production of toxic mutant transcripts and reverse phenotypes in DM1 stem cells. Genome editing was performed in DM1 neural stem cells (NSCs) derived from human DM1 induced pluripotent stem (iPS) cells. An editing cassette containing SV40/bGH polyA signals was integrated upstream of the CTG repeats by TALEN-mediated homologous recombination (HR). The expression of mutant CUG repeats transcript was monitored by nuclear RNA foci, the molecular hallmarks of DM1, using RNA fluorescence in situ hybridization. Alternative splicing of microtubule-associated protein tau (MAPT) and muscleblind-like (MBNL) proteins were analyzed to further monitor the phenotype reversal after genome modification. The cassette was successfully inserted into DMPK intron 9 and this genomic modification led to complete disappearance of nuclear RNA foci. MAPT and MBNL 1, 2 aberrant splicing in DM1 NSCs were reversed to normal pattern in genome-modified NSCs. Genome modification by integration of exogenous polyA signals upstream of the DMPK CTG repeat expansion prevents the production of toxic RNA and leads to phenotype reversal in human DM1 iPS-cells derived stem cells. Our data provide proof-of-principle evidence that genome modification may be used to generate genetically modified progenitor cells as a first step toward autologous cell transfer therapy for DM1.


Asunto(s)
Genoma Humano , Células Madre Pluripotentes Inducidas/citología , Proteínas Asociadas a Microtúbulos/metabolismo , Distrofia Miotónica/metabolismo , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Humanos , Distrofia Miotónica/patología , Fenotipo
10.
Lab Invest ; 95(1): 4-13, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25089770

RESUMEN

Approximately 3 years ago, we assessed how patient induced pluripotent stem cell (iPSC) research could potentially impact human pathobiology studies in the future. Since then, the field has grown considerably with numerous technical developments, and the idea of modeling diseases 'in a dish' is becoming increasingly popular in biomedical research. Likely, it is even acceptable to include patient iPSCs as one of the standard research tools for disease mechanism studies, just like knockout mice. However, as the field matures, we acknowledge there remain many practical limitations and obstacles for their genuine application to understand diseases, and accept that it has not been as straightforward to model disorders as initially proposed. A major practical challenge has been efficient direction of iPSC differentiation into desired lineages and preparation of the large numbers of specific cell types required for study. Another even larger obstacle is the limited value of in vitro outcomes, which often do not closely represent disease conditions. To overcome the latter issue, many new approaches are underway, including three-dimensional organoid cultures from iPSCs, xenotransplantation of human cells to animal models and in vitro interaction of multiple cell types derived from isogenic iPSCs. Here we summarize the areas where patient iPSC studies have provided truly valuable information beyond existing skepticism, discuss the desired technologies to overcome current limitations and include practical guidance for how to utilize the resources. Undoubtedly, these human patient cells are an asset for experimental pathology studies. The future rests on how wisely we use them.


Asunto(s)
Investigación Biomédica/métodos , Células Madre Pluripotentes/citología , Humanos
11.
J Cell Sci ; 126(Pt 16): 3638-48, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23750005

RESUMEN

Induced pluripotent stem cells (iPSCs) hold great promise for cell therapy. However, their low efficiency of lineage-specific differentiation and tumorigenesis severely hinder clinical translation. We hypothesized that reprogramming of somatic cells into lineage-specific progenitor cells might allow for large-scale expansion, avoiding the tumorigenesis inherent with iPSCs and simultaneously facilitating lineage-specific differentiation. Here we aimed at reprogramming rat hepatic WB cells, using four Yamanaka factors, into pancreatic progenitor cells (PPCs) or intermediate (IM) cells that have characteristics of PPCs. IM clones were selected based on their specific morphology and alkaline phosphatase activity and stably passaged under defined culture conditions. IM cells did not have iPSC properties, could be stably expanded in large quantity, and expressed all 14 genes that are used to define the PPC developmental stage. Directed differentiation of IM and WB cells by Pdx1-Ngn3-MafA (PNM) into pancreatic beta-like cells revealed that the IM cells are more susceptible to directed beta cell differentiation because of their open chromatin configuration, as demonstrated by expression of key pancreatic beta cell genes, secretion of insulin in response to glucose stimulation, and easy access to exogenous PNM proteins at the rat insulin 1 and Pdx1 promoters. This notion that IM cells are superior to their parental cells is further supported by the epigenetic demonstration of accessibility of Pdx1 and insulin 1 promoters. In conclusion, we have developed a strategy to derive and expand PPC cells from hepatic WB cells using conventional cell reprogramming. This proof-of-principal study may offer a novel, safe and effective way to generate autologous pancreatic beta cells for cell therapy of diabetes.


Asunto(s)
Hepatocitos/citología , Células Secretoras de Insulina/citología , Animales , Diferenciación Celular/fisiología , Línea Celular Tumoral , Células Cultivadas , Metilación de ADN , Hepatocitos/metabolismo , Proteínas de Homeodominio/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Regiones Promotoras Genéticas , Ratas , Células Madre/citología , Células Madre/metabolismo , Transactivadores/genética
12.
Microbiology (Reading) ; 160(Pt 7): 1417-1426, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24699069

RESUMEN

Pseudomonas aeruginosa is a Gram-negative opportunistic human pathogen possessing a type III secretion system (T3SS) which injects toxic effector proteins into mammalian host cells. In previous studies, P. aeruginosa strains lacking all of the known type III effectors were shown to cause cytotoxicity upon prolonged infection time. In this study, we report the identification of a new cytotoxin, nucleoside diphosphate kinase (NDK), which is injected into eukaryotic cells in a T3SS-dependent manner. Injection of NDK is inhibited by the presence of previously known effectors of the T3SS, with an effectorless strain injecting the highest amount, suggesting active competition with the known T3SS effectors. NDK is shown to cause a cytotoxic response when expressed in eukaryotic cells, and P. aeruginosa strains harbouring NDK also show a greater toxicity than strains lacking it. Interestingly, the cytotoxic effect of intracellular NDK is independent of its kinase activity. In previous studies, NDK was shown to be secreted into culture supernatants via a type I secretion system and cause cytotoxicity in a kinase-dependent manner. Therefore, the current study highlights an alternative route of NDK secretion as well as two different cytotoxic mechanisms of NDK, depending on the extra- or intra-cellular location of the protein.


Asunto(s)
Sistemas de Secreción Bacterianos , Toxinas Bacterianas/metabolismo , Nucleósido-Difosfato Quinasa/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Transporte Biológico , Supervivencia Celular , Femenino , Genes Reporteros , Células HeLa , Humanos , Nucleósido-Difosfato Quinasa/genética , Plásmidos/genética , Pseudomonas aeruginosa/genética , Proteínas Recombinantes de Fusión , Eliminación de Secuencia
13.
J Biol Chem ; 287(36): 30507-17, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22787153

RESUMEN

Nanog or Gata6-positive cells co-exist and are convertible within the inner cell mass of murine blastocysts and embryonic stem (ES) cells. Previous studies demonstrate fibroblast growth factor receptor 2 (FGFR2) triggers Nanog gene down-regulation and differentiation to primitive endoderm (PE); however, the underlying mechanisms responsible for reversible and fluctuating cell fate are poorly understood. Using an inducible FGFR2 dimerization system in ES cells, we demonstrate that FGFR2 activation rapidly down-regulated Nanog gene transcription through activation of the Mek pathway and subsequently differentiated ES cells into PE cells. FGFR2 rather selectively repressed the Nanog gene with minimal effect on other pluripotency genes, including Oct4 and Sox2. We determined the Nanog promoter region containing minimum Oct4/Sox2 binding sites was sufficient for this transcriptional down-regulation by FGFR2, when the reporter transgenes were integrated with insulators. Of interest, FGFR2-mediated Nanog transcriptional reduction occurred without dissociation of RNA polymerase II, p300, Oct4, Sox2, and Tet1 from the Nanog proximal promoter region and with no increase in repressive histone methylation marks or DNA methylation, implying the gene repression is in the early and transient phase. Furthermore, addition of a specific FGFR inhibitor readily reversed this Nanog repression status. These findings illustrate well how FGFR2 induces rapid but reversible Nanog repression within ES cells.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Multimerización de Proteína/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Transcripción Genética/fisiología , Animales , Línea Celular , Metilación de ADN/fisiología , Células Madre Embrionarias/citología , Endodermo/citología , Endodermo/embriología , Proteínas de Homeodominio/genética , Ratones , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
14.
Am J Physiol Endocrinol Metab ; 305(3): E325-35, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23736538

RESUMEN

In somatic cells, a collection of signaling pathways activated by amino acid limitation have been identified and referred to as the amino acid response (AAR). Despite the importance of possible detrimental effects of nutrient limitation during in vitro culture, the AAR has not been investigated in embryonic stem cells (ESC). AAR activation caused the expected increase in transcription factors that mediate specific AAR pathways, as well as the induction of asparagine synthetase, a terminal AAR target gene. Neither AAR activation nor stable knockdown of activating transcription factor (Atf) 4, a transcriptional mediator of the AAR, adversely affected ESC self-renewal or pluripotency. Low-level induction of the AAR over a 12-day period of embryoid body differentiation did alter lineage specification such that the primitive endodermal, visceral endodermal, and endodermal lineages were favored, whereas mesodermal and certain ectodermal lineages were suppressed. Knockdown of Atf4 further enhanced the AAR-induced increase in endodermal formation, suggesting that this phenomenon is mediated by an Atf4-independent mechanism. Collectively, the results indicate that, during differentiation of mouse embryoid bodies in culture, the availability of nutrients, such as amino acids, can influence the formation of specific cell lineages.


Asunto(s)
Aminoácidos/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Células Madre Embrionarias/metabolismo , Factor de Transcripción Activador 4/biosíntesis , Factor de Transcripción Activador 4/genética , Animales , Aspartatoamoníaco Ligasa/metabolismo , Western Blotting , Recuento de Células , Células Cultivadas , Citometría de Flujo , Ratones , Biosíntesis de Proteínas , ARN/biosíntesis , ARN/aislamiento & purificación , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcripción Genética
15.
Lab Invest ; 91(7): 972-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21555998

RESUMEN

Recent advances in DNA sequencing technologies and subsequent progress in genome-wide association study (GWAS) are rapidly changing the landscape of human diseases. Our knowledge on disease-gene linkage has been exponentially growing, and soon we will obtain complete maps of SNPs and mutations linked to nearly all major disease conditions. These studies will undoubtedly lead us to a more comprehensive understanding of how multiple genetic modifications link to human pathobiology. But what comes next after we discover these genetic linkages? To truly understand the mechanisms of how polygenic modifications identified through GWAS lead to disease conditions, we need an experimental interface to study their pathobiological effects. In this study, induced pluripotent stem cells (iPSCs), retaining all the genetic information from patients, will likely serve as a powerful resource. Indeed, pioneering studies have demonstrated that disease-specific iPSCs are useful for understanding disease mechanisms. Moreover, iPSC-derived cells, when recapitulating some disease phenotypes in vitro, can be a fast track screening tool for drug discovery. Further, with GWAS information, iPSCs will become a valuable tool to predict drug efficacy and toxicity for individuals, thus promoting personalized medicine. In this review, we will discuss how patient-specific iPSCs will become a powerful biomedical interface in clinical translational research.


Asunto(s)
Células Madre Pluripotentes/citología , Estudio de Asociación del Genoma Completo , Humanos , Modelos Biológicos , Mutación , Células Madre Pluripotentes/metabolismo , Polimorfismo de Nucleótido Simple , Medicina de Precisión
16.
Chembiochem ; 12(18): 2822-8, 2011 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-22038863

RESUMEN

Considering the essential role of chromatin remodeling in gene regulation, their directed modulation is of increasing importance. To achieve gene activation by epigenetic modification, we synthesized a series of pyrrole-imidazole polyamide conjugates (PIPs) that can bind to predetermined DNA sequences, and attached them with suberoylanilide hydroxamic acid (SAHA), a potent histone deacetylase inhibitor. As histone modification is associated with pluripotency, these new types of conjugates, termed SAHA-PIPs, were screened for their effect on the expression of induced pluripotent stem cell (iPSC) factors. We found certain SAHA-PIPs that could differentially up-regulate the endogenous expression of Oct-3/4, Nanog, Sox2, Klf4 and c-Myc. SAHA and other SAHA-PIPs did not show such induction; this implies a role for PIPs and their sequence specificity in this differential gene activation. Chromatin immunoprecipitation analysis suggested that SAHA-PIP-mediated gene induction proceeds by histone H3 Lys9 and Lys14 acetylation and Lys4 trimethylation, which are epigenetic features associated with transcriptionally active chromatin.


Asunto(s)
Embrión de Mamíferos/citología , Epigénesis Genética , Animales , Fibroblastos/citología , Factor 4 Similar a Kruppel , Ratones
17.
Front Endocrinol (Lausanne) ; 12: 737276, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858326

RESUMEN

Type 1 diabetes (T1D) is a disease that arises due to complex immunogenetic mechanisms. Key cell-cell interactions involved in the pathogenesis of T1D are activation of autoreactive T cells by dendritic cells (DC), migration of T cells across endothelial cells (EC) lining capillary walls into the islets of Langerhans, interaction of T cells with macrophages in the islets, and killing of ß-cells by autoreactive CD8+ T cells. Overall, pathogenic cell-cell interactions are likely regulated by the individual's collection of genetic T1D-risk variants. To accurately model the role of genetics, it is essential to build systems to interrogate single candidate genes in isolation during the interactions of cells that are essential for disease development. However, obtaining single-donor matched cells relevant to T1D is a challenge. Sourcing these genetic variants from human induced pluripotent stem cells (iPSC) avoids this limitation. Herein, we have differentiated iPSC from one donor into DC, macrophages, EC, and ß-cells. Additionally, we also engineered T cell avatars from the same donor to provide an in vitro platform to study genetic influences on these critical cellular interactions. This proof of concept demonstrates the ability to derive an isogenic system from a single donor to study these relevant cell-cell interactions. Our system constitutes an interdisciplinary approach with a controlled environment that provides a proof-of-concept for future studies to determine the role of disease alleles (e.g. IFIH1, PTPN22, SH2B3, TYK2) in regulating cell-cell interactions and cell-specific contributions to the pathogenesis of T1D.


Asunto(s)
Linfocitos T CD8-positivos/patología , Diabetes Mellitus Tipo 1/patología , Células Madre Pluripotentes Inducidas/patología , Diferenciación Celular/fisiología , Humanos , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología
18.
Biochim Biophys Acta ; 1789(3): 198-203, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19167530

RESUMEN

Adenine nucleotide translocase (Ant) mediates the exchange of ADP and ATP across the inner mitochondrial membrane in eukaryotes. Mice possess three distinct but highly homologous Ant isoforms, encoded by independent genes, whose transcription depends upon tissue type. Ant1 is expressed selectively in heart and skeletal muscles, Ant2 is ubiquitously expressed in most tissues but lower in skeletal muscle and testis, while Ant4 is exclusively expressed in the testis. Of interest, each of these Ant genes contains CpG islands in their proximal promoter regions. We investigated the methylation status of the three Ant genes in various tissues with active and inactive transcription. In contrast to the Ant4 gene in which CpG island methylation is essential for gene repression, the CpG islands of Ant1 and Ant2 are hypomethylated regardless of the gene expression status throughout the tissues of male mice. Despite the tissue specific expression profile of Ant1, CpG methylation is unlikely involved in the regulation of the gene. Consistent with these findings, addition of a CpG-demethylating agent, 5-aza-2'-deoxycitidine, to fibroblasts increased the expression of Ant4 but not Ant1 or Ant2 genes. This study provides insight regarding the differential regulation of Ant isoforms in mammals, whereby both the Ant1 and Ant2 genes are capable of expression, but the Ant4 gene is completely repressed throughout somatic tissues. To the best of our knowledge, this is a first example to clearly demonstrate a differential usage of CpG island methylation within a family of genes.


Asunto(s)
Islas de CpG , Metilación de ADN , Translocasas Mitocondriales de ADP y ATP/genética , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , ADN/análisis , ADN/química , ADN/genética , Metilasas de Modificación del ADN/antagonistas & inhibidores , Enzimas de Restricción del ADN , Decitabina , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Expresión Génica , Genes , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones , Translocasas Mitocondriales de ADP y ATP/metabolismo , Células 3T3 NIH , Regiones Promotoras Genéticas , Sulfitos
19.
Mol Cell Biol ; 27(12): 4347-54, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17403895

RESUMEN

The canonical Wnt/beta-catenin signaling pathway plays diverse roles in embryonic development and disease. Activation of this pathway, likely by Wnt-10b, has been shown to inhibit adipogenesis in cultured 3T3-L1 preadipocytes and in mice. Here, we report that the beta-catenin antagonist Chibby (Cby) is required for adipocyte differentiation. Cby is expressed in adipose tissue in mice, and Cby protein levels increase during adipogenic differentiation of 3T3-L1 cells. Ectopic expression of Cby induces spontaneous differentiation of these cells into mature adipocytes to an extent similar to that of dominant-negative Tcf-4. In contrast, depletion of Cby by RNA interference potently blocks adipogenesis of 3T3-L1 and mouse embryonic stem cells. In support of this, embryonic fibroblasts obtained from Cby-deficient embryos display attenuated differentiation to the adipogenic lineage. Mechanistically, Cby promotes adipocyte differentiation, in part by inhibiting beta-catenin, since gain or loss of function of Cby influences beta-catenin signaling in 3T3-L1 cells. Our results therefore establish Cby as a novel proadipogenic factor required for adipocyte differentiation.


Asunto(s)
Adipocitos/fisiología , Proteínas Portadoras/fisiología , Diferenciación Celular , Proteínas Nucleares/fisiología , beta Catenina/antagonistas & inhibidores , Células 3T3-L1 , Adipogénesis , Animales , Proteínas Portadoras/genética , Línea Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Genes Reporteros , Histocitoquímica , Humanos , Luciferasas/metabolismo , Ratones , Proteínas Nucleares/genética , Interferencia de ARN , Transducción de Señal
20.
Mol Biol Cell ; 18(9): 3375-87, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17596515

RESUMEN

Gene silencing using small interfering RNA (siRNA) is a valuable laboratory tool and a promising approach to therapeutics for a variety of human diseases. Recently, RNA interference (RNAi) has been linked to cytoplasmic GW bodies (GWB). However, the correlation between RNAi and the formation of GWB, also known as mammalian processing bodies, remains unclear. In this report, we show that transfection of functional siRNA induced larger and greater numbers of GWB. This siRNA-induced increase of GWB depended on the endogenous expression of the target mRNA. Knockdown of GW182 or Ago2 demonstrated that the siRNA-induced increase of GWB required these two proteins and correlated with RNAi. Furthermore, knockdown of rck/p54 or LSm1 did not prevent the reassembly of GWB that were induced by and correlated with siRNA-mediated RNA silencing. We propose that RNAi is a key regulatory mechanism for the assembly of GWB, and in some cases, GWB may serve as markers for RNAi in mammalian cells.


Asunto(s)
Estructuras Citoplasmáticas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Animales , Proteínas Argonautas , Autoantígenos/metabolismo , ARN Helicasas DEAD-box/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Células HeLa , Humanos , Laminas/metabolismo , Ratones , Células 3T3 NIH , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA