Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(6): e2209967120, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36719921

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe muscle wasting disease caused by the lack of dystrophin. Heart failure, driven by cardiomyocyte death, fibrosis, and the development of dilated cardiomyopathy, is the leading cause of death in DMD patients. Current treatments decrease the mechanical load on the heart but do not address the root cause of dilated cardiomyopathy: cardiomyocyte death. Previously, we showed that telomere shortening is a hallmark of DMD cardiomyocytes. Here, we test whether prevention of telomere attrition is possible in cardiomyocytes differentiated from patient-derived induced pluripotent stem cells (iPSC-CMs) and if preventing telomere shortening impacts cardiomyocyte function. We observe reduced cell size, nuclear size, and sarcomere density in DMD iPSC-CMs compared with healthy isogenic controls. We find that expression of just one telomere-binding protein, telomeric repeat-binding factor 2 (TRF2), a core component of the shelterin complex, prevents telomere attrition and rescues deficiencies in cell size as well as sarcomere density. We employ a bioengineered platform to micropattern cardiomyocytes for calcium imaging and perform Southern blots of telomere restriction fragments, the gold standard for telomere length assessments. Importantly, preservation of telomere lengths in DMD cardiomyocytes improves their viability. These data provide evidence that preventing telomere attrition ameliorates deficits in cell morphology, activation of the DNA damage response, and premature cell death, suggesting that TRF2 is a key player in DMD-associated cardiac failure.


Asunto(s)
Cardiomiopatía Dilatada , Insuficiencia Cardíaca , Células Madre Pluripotentes Inducidas , Distrofia Muscular de Duchenne , Humanos , Cardiomiopatía Dilatada/genética , Supervivencia Celular , Distrofina/genética , Insuficiencia Cardíaca/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Miocitos Cardíacos/metabolismo , Telómero/genética , Telómero/metabolismo
2.
NPJ Regen Med ; 7(1): 19, 2022 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-35304486

RESUMEN

Duchenne muscular dystrophy (DMD) is a progressive genetic myopathy that leads to heart failure from dilated cardiomyopathy by early adulthood. Recent evidence suggests that tamoxifen, a selective estrogen receptor modulator widely used to treat breast cancer, ameliorates DMD cardiomyopathy. However, the mechanism of action of 4-hydroxytamoxifen, the active metabolite of tamoxifen, on cardiomyocyte function remains unclear. To examine the effects of chronic 4-hydroxytamoxifen treatment, we used state-of-the-art human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and a bioengineered platform to model DMD. We assessed the beating rate and beating velocity of iPSC-CMs in monolayers and as single cells on micropatterns that promote a physiological cardiomyocyte morphology. We found that 4-hydroxytamoxifen treatment of DMD iPSC-CMs decreased beating rate, increased beating velocity, and ameliorated calcium-handling deficits, leading to prolonged viability. Our study highlights the utility of a bioengineered iPSC-CM platform for drug testing and underscores the potential of repurposing tamoxifen as a therapy for DMD cardiomyopathy.

3.
Stem Cell Reports ; 16(9): 2169-2181, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34019816

RESUMEN

Duchenne muscular dystrophy (DMD) is a rare X-linked recessive disease that is associated with severe progressive muscle degeneration culminating in death due to cardiorespiratory failure. We previously observed an unexpected proliferation-independent telomere shortening in cardiomyocytes of a DMD mouse model. Here, we provide mechanistic insights using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Using traction force microscopy, we show that DMD hiPSC-CMs exhibit deficits in force generation on fibrotic-like bioengineered hydrogels, aberrant calcium handling, and increased reactive oxygen species levels. Furthermore, we observed a progressive post-mitotic telomere shortening in DMD hiPSC-CMs coincident with downregulation of shelterin complex, telomere capping proteins, and activation of the p53 DNA damage response. This telomere shortening is blocked by blebbistatin, which inhibits contraction in DMD cardiomyocytes. Our studies underscore the role of fibrotic stiffening in the etiology of DMD cardiomyopathy. In addition, our data indicate that telomere shortening is progressive, contraction dependent, and mechanosensitive, and suggest points of therapeutic intervention.


Asunto(s)
Distrofias Musculares/genética , Distrofias Musculares/fisiopatología , Contracción Miocárdica/genética , Miocitos Cardíacos/metabolismo , Acortamiento del Telómero/genética , Biomarcadores , Cardiomiopatías/etiología , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Diferenciación Celular , Células Cultivadas , Microambiente Celular/efectos de los fármacos , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Fibrosis , Técnica del Anticuerpo Fluorescente , Expresión Génica , Humanos , Inmunofenotipificación , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Fenómenos Mecánicos , Distrofias Musculares/patología , Distrofia Muscular de Duchenne/etiología , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Contracción Miocárdica/efectos de los fármacos
4.
PLoS One ; 15(12): e0243905, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33351840

RESUMEN

Pyrrole-imidazole (Py-Im) polyamides are synthetic molecules that can be rationally designed to target specific DNA sequences to both disrupt and recruit transcriptional machinery. While in vitro binding has been extensively studied, in vivo effects are often difficult to predict using current models of DNA binding. Determining the impact of genomic architecture and the local chromatin landscape on polyamide-DNA sequence specificity remains an unresolved question that impedes their effective deployment in vivo. In this report we identified polyamide-DNA interaction sites across the entire genome, by covalently crosslinking and capturing these events in the nuclei of human LNCaP cells. This technique confirms the ability of two eight ring hairpin-polyamides, with similar architectures but differing at a single ring position (Py to Im), to retain in vitro specificities and display distinct genome-wide binding profiles.


Asunto(s)
Proteínas de Unión al ADN/genética , Genoma Humano/efectos de los fármacos , Conformación de Ácido Nucleico/efectos de los fármacos , Nylons/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Humanos , Imidazoles/farmacología , Pirroles/farmacología
5.
FEBS Lett ; 592(6): 888-900, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29389011

RESUMEN

Transcription factors (TFs) reprogram cell states by exerting control over gene regulatory networks and the epigenetic landscape of a cell. Artificial transcription factors (ATFs) are designer regulatory proteins comprised of modular units that can be customized to overcome challenges faced by natural TFs in establishing and maintaining desired cell states. Decades of research on DNA-binding proteins and synthetic molecules has provided a molecular toolkit for ATF design and the construction of genome-scale libraries of ATFs capable of phenotypic manipulation and reprogramming of cell states. Here, we compare the unique strengths and limitations of different ATF platforms, highlight the advantages of cooperative assembly, and present the potential of ATF libraries in revealing gene regulatory networks that govern cell fate choices.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Reprogramación Celular , Epigénesis Genética , Redes Reguladoras de Genes , Factores de Transcripción , Animales , Humanos , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
6.
Science ; 358(6370): 1617-1622, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29192133

RESUMEN

The release of paused RNA polymerase II into productive elongation is highly regulated, especially at genes that affect human development and disease. To exert control over this rate-limiting step, we designed sequence-specific synthetic transcription elongation factors (Syn-TEFs). These molecules are composed of programmable DNA-binding ligands flexibly tethered to a small molecule that engages the transcription elongation machinery. By limiting activity to targeted loci, Syn-TEFs convert constituent modules from broad-spectrum inhibitors of transcription into gene-specific stimulators. Here we present Syn-TEF1, a molecule that actively enables transcription across repressive GAA repeats that silence frataxin expression in Friedreich's ataxia, a terminal neurodegenerative disease with no effective therapy. The modular design of Syn-TEF1 defines a general framework for developing a class of molecules that license transcription elongation at targeted genomic loci.


Asunto(s)
Cromatina/metabolismo , Ataxia de Friedreich/genética , Proteínas de Unión a Hierro/genética , Activación Transcripcional , Factores de Elongación Transcripcional/síntesis química , Factores de Elongación Transcripcional/metabolismo , Silenciador del Gen , Humanos , ARN Polimerasa II/metabolismo , Transcripción Genética , Frataxina
7.
Proc Natl Acad Sci U S A ; 113(51): E8257-E8266, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27930301

RESUMEN

Artificial transcription factors (ATFs) are precision-tailored molecules designed to bind DNA and regulate transcription in a preprogrammed manner. Libraries of ATFs enable the high-throughput screening of gene networks that trigger cell fate decisions or phenotypic changes. We developed a genome-scale library of ATFs that display an engineered interaction domain (ID) to enable cooperative assembly and synergistic gene expression at targeted sites. We used this ATF library to screen for key regulators of the pluripotency network and discovered three combinations of ATFs capable of inducing pluripotency without exogenous expression of Oct4 (POU domain, class 5, TF 1). Cognate site identification, global transcriptional profiling, and identification of ATF binding sites reveal that the ATFs do not directly target Oct4; instead, they target distinct nodes that converge to stimulate the endogenous pluripotency network. This forward genetic approach enables cell type conversions without a priori knowledge of potential key regulators and reveals unanticipated gene network dynamics that drive cell fate choices.


Asunto(s)
Linaje de la Célula , Reprogramación Celular , Factores de Transcripción/metabolismo , Animales , Sitios de Unión/genética , Chaperonina con TCP-1/metabolismo , Epigénesis Genética , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Biblioteca Genómica , Células HEK293 , Humanos , Ratones , Dominios Proteicos , Ingeniería de Proteínas , Análisis de Secuencia de ARN , Factores de Transcripción/genética , Transcripción Genética , Dedos de Zinc/genética
8.
J Vis Exp ; (107): e53510, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26863565

RESUMEN

The genome is the target of some of the most effective chemotherapeutics, but most of these drugs lack DNA sequence specificity, which leads to dose-limiting toxicity and many adverse side effects. Targeting the genome with sequence-specific small molecules may enable molecules with increased therapeutic index and fewer off-target effects. N-methylpyrrole/N-methylimidazole polyamides are molecules that can be rationally designed to target specific DNA sequences with exquisite precision. And unlike most natural transcription factors, polyamides can bind to methylated and chromatinized DNA without a loss in affinity. The sequence specificity of polyamides has been extensively studied in vitro with cognate site identification (CSI) and with traditional biochemical and biophysical approaches, but the study of polyamide binding to genomic targets in cells remains elusive. Here we report a method, the crosslinking of small molecules to isolate chromatin (COSMIC), that identifies polyamide binding sites across the genome. COSMIC is similar to chromatin immunoprecipitation (ChIP), but differs in two important ways: (1) a photocrosslinker is employed to enable selective, temporally-controlled capture of polyamide binding events, and (2) the biotin affinity handle is used to purify polyamide-DNA conjugates under semi-denaturing conditions to decrease DNA that is non-covalently bound. COSMIC is a general strategy that can be used to reveal the genome-wide binding events of polyamides and other genome-targeting chemotherapeutic agents.


Asunto(s)
Cromatina/aislamiento & purificación , ADN/química , Estudio de Asociación del Genoma Completo/métodos , Nylons/química , Sitios de Unión , Cromatina/química , ADN/genética , ADN/metabolismo , Humanos , Imidazoles/química , Nylons/metabolismo , Pirroles/química , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo
9.
Biochem J ; 462(3): 397-413, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25145439

RESUMEN

Transcription factors control the fate of a cell by regulating the expression of genes and regulatory networks. Recent successes in inducing pluripotency in terminally differentiated cells as well as directing differentiation with natural transcription factors has lent credence to the efforts that aim to direct cell fate with rationally designed transcription factors. Because DNA-binding factors are modular in design, they can be engineered to target specific genomic sequences and perform pre-programmed regulatory functions upon binding. Such precision-tailored factors can serve as molecular tools to reprogramme or differentiate cells in a targeted manner. Using different types of engineered DNA binders, both regulatory transcriptional controls of gene networks, as well as permanent alteration of genomic content, can be implemented to study cell fate decisions. In the present review, we describe the current state of the art in artificial transcription factor design and the exciting prospect of employing artificial DNA-binding factors to manipulate the transcriptional networks as well as epigenetic landscapes that govern cell fate.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Redes Reguladoras de Genes/fisiología , Factores de Transcripción/metabolismo , Proteínas Asociadas a CRISPR/fisiología , Regulación de la Expresión Génica , Nylons/química , Nylons/metabolismo , Ingeniería de Proteínas , Estructura Terciaria de Proteína , Dedos de Zinc/fisiología
10.
Angew Chem Int Ed Engl ; 53(38): 10124-8, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25066383

RESUMEN

Targeting the genome with sequence-specific synthetic molecules is a major goal at the interface of chemistry, biology, and personalized medicine. Pyrrole/imidazole-based polyamides can be rationally designed to target specific DNA sequences with exquisite precision in vitro; yet, the biological outcomes are often difficult to interpret using current models of binding energetics. To directly identify the binding sites of polyamides across the genome, we designed, synthesized, and tested polyamide derivatives that enabled covalent crosslinking and localization of polyamide-DNA interaction sites in live human cells. Bioinformatic analysis of the data reveals that clustered binding sites, spanning a broad range of affinities, best predict occupancy in cells. In contrast to the prevailing paradigm of targeting single high-affinity sites, our results point to a new design principle to deploy polyamides and perhaps other synthetic molecules to effectively target desired genomic sites in vivo.


Asunto(s)
ADN/química , ADN/efectos de los fármacos , Diseño de Fármacos , Genoma/genética , Nylons/química , Nylons/farmacología , Sitios de Unión/efectos de los fármacos , Biología Computacional , ADN/genética , Genómica , Células HEK293 , Humanos , Modelos Moleculares , Estructura Molecular , Nylons/síntesis química
11.
J Neurochem ; 109(1): 74-84, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19183270

RESUMEN

During CNS injury and diseases, nitric oxide (NO) is released at a high flux rate leading to formation of peroxynitrite (ONOO(*)) and other reactive nitrogenous species, which nitrate tyrosines of proteins to form 3-nitrotyrosine (3NY), leading to cell death. Previously, we have found that motor neurons exposed to low levels of NO become resistant to subsequent cytotoxic NO challenge; an effect dubbed induced adaptive resistance (IAR). Here, we report IAR mitigates, not only cell death, but 3NY formation in response to cytotoxic NO. Addition of an NO scavenger before NO challenge duplicates IAR, implicating reactive nitrogenous species in cell death. Addition of uric acid (a peroxynitrite scavenger) before cytotoxic NO challenge, duplicates IAR, implicating peroxynitrite, with subsequent 3NY formation, in cell death, and abrogation of this pathway as a mechanism of IAR. IAR is dependent on the heme-metabolizing enzyme, heme oxygenase-1 (HO1), as indicated by the elimination of IAR by a specific HO1 inhibitor, and by the finding that neurons isolated from HO1 null mice have increased NO sensitivity with concomitant increased 3NY formation. This data indicate that IAR is an HO1-dependent mechanism that prevents peroxynitrite-mediated NO toxicity in motor neurons, thereby elucidating therapeutic targets for the mitigation of CNS disease and injury.


Asunto(s)
Adaptación Fisiológica/fisiología , Sistema Nervioso Central/patología , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/toxicidad , Ácido Peroxinitroso/metabolismo , Adaptación Fisiológica/efectos de los fármacos , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Enfermedades del Sistema Nervioso Central/inducido químicamente , Enfermedades del Sistema Nervioso Central/metabolismo , Enfermedades del Sistema Nervioso Central/patología , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Óxido Nítrico/metabolismo , Embarazo , Especies de Nitrógeno Reactivo/metabolismo
12.
J Neurochem ; 109(1): 93-104, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19226373

RESUMEN

Depending on its concentration, nitric oxide (NO) has beneficial or toxic effects. In pathological conditions, NO reacts with superoxide to form peroxynitrite, which nitrates proteins forming nitrotyrosine residues (3NY), leading to loss of protein function, perturbation of signal transduction, and cell death. 3NY immunoreactivity is present in many CNS diseases, particularly multiple sclerosis. Here, using the high flux NO donor, spermine-NONOate, we report that oligodendrocytes are resistant to NO, while motor neurons are NO sensitive. Motor neuron sensitivity correlates with the NO-dependent formation of 3NY, which is significantly more pronounced in motor neurons when compared with oligodendrocytes, suggesting peroxynitrite as the toxic molecule. The heme-metabolizing enzyme, heme-oxygenase-1 (HO1), is necessary for oligodendrocyte NO resistance, as demonstrated by loss of resistance after HO1 inhibition. Resistance is reinstated by peroxynitrite scavenging with uric acid further implicating peroxynitrite as responsible for NO sensitivity. Most importantly, differential sensitivity to NO is also present in cultures of primary oligodendrocytes and motor neurons. Finally, motor neurons cocultured with oligodendrocytes, or oligodendrocyte-conditioned media, become resistant to NO toxicity. Preliminary studies suggest oligodendrocytes release a soluble factor that protects motor neurons. Our findings challenge the current paradigm that oligodendrocytes are the exclusive target of multiple sclerosis pathology.


Asunto(s)
Neuronas Motoras/metabolismo , Esclerosis Múltiple/metabolismo , Oligodendroglía/metabolismo , Especies de Nitrógeno Reactivo/metabolismo , Animales , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Femenino , Hemo-Oxigenasa 1/metabolismo , Humanos , Ratones , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/patología , Esclerosis Múltiple/patología , Óxido Nítrico/metabolismo , Óxido Nítrico/toxicidad , Donantes de Óxido Nítrico/farmacología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/patología , Ácido Peroxinitroso/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...