Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Biol ; 21(6): e3002121, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37315073

RESUMEN

Pluripotency defines the unlimited potential of individual cells of vertebrate embryos, from which all adult somatic cells and germ cells are derived. Understanding how the programming of pluripotency evolved has been obscured in part by a lack of data from lower vertebrates; in model systems such as frogs and zebrafish, the function of the pluripotency genes NANOG and POU5F1 have diverged. Here, we investigated how the axolotl ortholog of NANOG programs pluripotency during development. Axolotl NANOG is absolutely required for gastrulation and germ-layer commitment. We show that in axolotl primitive ectoderm (animal caps; ACs) NANOG and NODAL activity, as well as the epigenetic modifying enzyme DPY30, are required for the mass deposition of H3K4me3 in pluripotent chromatin. We also demonstrate that all 3 protein activities are required for ACs to establish the competency to differentiate toward mesoderm. Our results suggest the ancient function of NANOG may be establishing the competence for lineage differentiation in early cells. These observations provide insights into embryonic development in the tetrapod ancestor from which terrestrial vertebrates evolved.


Asunto(s)
Proteínas de Homeodominio , Células Madre Pluripotentes , Animales , Proteínas de Homeodominio/metabolismo , Ambystoma mexicanum/genética , Ambystoma mexicanum/metabolismo , Pez Cebra/genética , Diferenciación Celular , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Regulación del Desarrollo de la Expresión Génica
2.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-35163379

RESUMEN

Non-viral gene delivery has become a popular approach in tissue engineering, as it permits the transient delivery of a therapeutic gene, in order to stimulate tissue repair. However, the efficacy of non-viral delivery vectors remains an issue. Our lab has created gene-activated scaffolds by incorporating various non-viral delivery vectors, including the glycosaminoglycan-binding enhanced transduction (GET) peptide into collagen-based scaffolds with proven osteogenic potential. A modification to the GET peptide (FLR) by substitution of arginine residues with histidine (FLH) has been designed to enhance plasmid DNA (pDNA) delivery. In this study, we complexed pDNA with combinations of FLR and FLH peptides, termed GET* nanoparticles. We sought to enhance our gene-activated scaffold platform by incorporating GET* nanoparticles into collagen-nanohydroxyapatite scaffolds with proven osteogenic capacity. GET* N/P 8 was shown to be the most effective formulation for delivery to MSCs in 2D. Furthermore, GET* N/P 8 nanoparticles incorporated into collagen-nanohydroxyapatite (coll-nHA) scaffolds at a 1:1 ratio of collagen:nanohydroxyapatite was shown to be the optimal gene-activated scaffold. pDNA encoding stromal-derived factor 1α (pSDF-1α), an angiogenic chemokine which plays a role in BMP mediated differentiation of MSCs, was then delivered to MSCs using our optimised gene-activated scaffold platform, with the aim of significantly increasing angiogenesis as an important precursor to bone repair. The GET* N/P 8 coll-nHA scaffolds successfully delivered pSDF-1α to MSCs, resulting in a significant, sustained increase in SDF-1α protein production and an enhanced angiogenic effect, a key precursor in the early stages of bone repair.


Asunto(s)
Péptidos de Penetración Celular/farmacología , Quimiocina CXCL12/administración & dosificación , Sistemas de Liberación de Medicamentos , Neovascularización Fisiológica , Ingeniería de Tejidos , Andamios del Tejido/química , Activación Transcripcional , Animales , Materiales Biocompatibles/farmacología , Quimiocina CXCL12/farmacología , Colágeno/química , ADN/química , Durapatita/química , Células Progenitoras Endoteliales/metabolismo , Glicosaminoglicanos/química , Nanopartículas , Neovascularización Fisiológica/efectos de los fármacos , Plásmidos/química , Ratas Sprague-Dawley , Activación Transcripcional/efectos de los fármacos , Transfección
3.
J Biol Chem ; 294(2): 424-436, 2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30373771

RESUMEN

Ubiquitin-specific proteases (USPs) reverse ubiquitination and regulate virtually all cellular processes. Defined noncatalytic domains in USP4 and USP15 are known to interact with E3 ligases and substrate recruitment factors. No such interactions have been reported for these domains in the paralog USP11, a key regulator of DNA double-strand break repair by homologous recombination. We hypothesized that USP11 domains adjacent to its protease domain harbor unique peptide-binding sites. Here, using a next-generation phage display (NGPD) strategy, combining phage display library screening with next-generation sequencing, we discovered unique USP11-interacting peptide motifs. Isothermal titration calorimetry disclosed that the highest affinity peptides (KD of ∼10 µm) exhibit exclusive selectivity for USP11 over USP4 and USP15 in vitro Furthermore, a crystal structure of a USP11-peptide complex revealed a previously unknown binding site in USP11's noncatalytic ubiquitin-like (UBL) region. This site interacted with a helical motif and is absent in USP4 and USP15. Reporter assays using USP11-WT versus a binding pocket-deficient double mutant disclosed that this binding site modulates USP11's function in homologous recombination-mediated DNA repair. The highest affinity USP11 peptide binder fused to a cellular delivery sequence induced significant nuclear localization and cell cycle arrest in S phase, affecting the viability of different mammalian cell lines. The USP11 peptide ligands and the paralog-specific functional site in USP11 identified here provide a framework for the development of new biochemical tools and therapeutic agents. We propose that an NGPD-based strategy for identifying interacting peptides may be applied also to other cellular targets.


Asunto(s)
Péptidos/química , Péptidos/metabolismo , Tioléster Hidrolasas/química , Tioléster Hidrolasas/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Cristalografía por Rayos X , Reparación del ADN , Recombinación Homóloga , Humanos , Cinética , Ligandos , Ratones , Datos de Secuencia Molecular , Péptidos/genética , Dominios Proteicos , Tioléster Hidrolasas/genética , Ubiquitina/química , Ubiquitina/metabolismo , Ubiquitinación
4.
Proc Natl Acad Sci U S A ; 113(3): E291-9, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26733682

RESUMEN

Protein transduction domains (PTDs) are powerful nongenetic tools that allow intracellular delivery of conjugated cargoes to modify cell behavior. Their use in biomedicine has been hampered by inefficient delivery to nuclear and cytoplasmic targets. Here we overcame this deficiency by developing a series of novel fusion proteins that couple a membrane-docking peptide to heparan sulfate glycosaminoglycans (GAGs) with a PTD. We showed that this GET (GAG-binding enhanced transduction) system could deliver enzymes (Cre, neomycin phosphotransferase), transcription factors (NANOG, MYOD), antibodies, native proteins (cytochrome C), magnetic nanoparticles (MNPs), and nucleic acids [plasmid (p)DNA, modified (mod)RNA, and small inhibitory RNA] at efficiencies of up to two orders of magnitude higher than previously reported in cell types considered hard to transduce, such as mouse embryonic stem cells (mESCs), human ESCs (hESCs), and induced pluripotent stem cells (hiPSCs). This technology represents an efficient strategy for controlling cell labeling and directing cell fate or behavior that has broad applicability for basic research, disease modeling, and clinical application.


Asunto(s)
Péptidos de Penetración Celular/metabolismo , Sistemas de Liberación de Medicamentos , Glicosaminoglicanos/metabolismo , Secuencias de Aminoácidos , Animales , Diferenciación Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Péptidos de Penetración Celular/química , Detergentes/farmacología , Endocitosis/efectos de los fármacos , Genoma , Proteínas de Homeodominio/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Integrasas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Desarrollo de Músculos/efectos de los fármacos , Proteína MioD/metabolismo , Células 3T3 NIH , Proteína Homeótica Nanog , Nanopartículas , Ácidos Nucleicos/metabolismo , Estructura Terciaria de Proteína , Solubilidad , Tripsina/metabolismo
5.
Proc Natl Acad Sci U S A ; 111(15): 5580-5, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24706900

RESUMEN

The ability of materials to define the architecture and microenvironment experienced by cells provides new opportunities to direct the fate of human pluripotent stem cells (HPSCs) [Robinton DA, Daley GQ (2012) Nature 481(7381):295-305]. However, the conditions required for self-renewal vs. differentiation of HPSCs are different, and a single system that efficiently achieves both outcomes is not available [Giobbe GG, et al. (2012) Biotechnol Bioeng 109(12):3119-3132]. We have addressed this dual need by developing a hydrogel-based material that uses ionic de-cross-linking to remove a self-renewal permissive hydrogel (alginate) and switch to a differentiation-permissive microenvironment (collagen). Adjusting the timing of this switch can preferentially steer the HPSC differentiation to mimic lineage commitment during gastrulation to ectoderm (early switch) or mesoderm/endoderm (late switch). As an exemplar differentiated cell type, we showed that directing early lineage specification using this single system can promote cardiogenesis with increased gene expression in high-density cell populations. This work will facilitate regenerative medicine by allowing in situ HPSC expansion to be coupled with early lineage specification within defined tissue geometries.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Hidrogeles/farmacología , Células Madre Pluripotentes/fisiología , Nicho de Células Madre/efectos de los fármacos , Alginatos , Carbocianinas , Colágeno , Ácido Glucurónico , Ácidos Hexurónicos , Humanos , Células Madre Pluripotentes/efectos de los fármacos , Medicina Regenerativa/métodos , Análisis Espectral
6.
PLoS Biol ; 11(4): e1001538, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23610558

RESUMEN

Little is known about extrinsic signals required for the advancement of motor neuron (MN) axons, which extend over long distances in the periphery to form precise connections with target muscles. Here we present that Rnf165 (Arkadia-like; Arkadia2; Ark2C) is expressed specifically in the nervous system and that its loss in mice causes motor innervation defects that originate during development and lead to wasting and death before weaning. The defects range from severe reduction of motor axon extension as observed in the dorsal forelimb to shortening of presynaptic branches of the phrenic nerve, as observed in the diaphragm. Molecular functional analysis showed that in the context of the spinal cord Ark2C enhances transcriptional responses of the Smad1/5/8 effectors, which are activated (phosphorylated) downstream of Bone Morphogenetic Protein (BMP) signals. Consistent with Ark2C-modulated BMP signaling influencing motor axons, motor pools in the spinal cord were found to harbor phosphorylated Smad1/5/8 (pSmad) and treatment of primary MN with BMP inhibitor diminished axon length. In addition, genetic reduction of BMP-Smad signaling in Ark2C (+/-) mice caused the emergence of Ark2C (-/-) -like dorsal forelimb innervation deficits confirming that enhancement of BMP-Smad responses by Ark2C mediates efficient innervation. Together the above data reveal an involvement of BMP-Smad signaling in motor axon advancement.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Neuronas Motoras/fisiología , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Proteínas Smad/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Axones/fisiología , Tipificación del Cuerpo , Aumento de la Célula , Miembro Anterior/inervación , Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Músculo Esquelético/inervación , Atrofia Muscular/genética , Mutagénesis Insercional , Proteínas del Tejido Nervioso/genética , Fenotipo , Nervio Frénico/patología , Proteolisis , Médula Espinal/metabolismo , Activación Transcripcional , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
7.
Eur Heart J ; 35(16): 1078-87, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23470493

RESUMEN

AIMS: Long-QT syndromes (LQTS) are mostly autosomal-dominant congenital disorders associated with a 1:1000 mutation frequency, cardiac arrest, and sudden death. We sought to use cardiomyocytes derived from human-induced pluripotency stem cells (hiPSCs) as an in vitro model to develop and evaluate gene-based therapeutics for the treatment of LQTS. METHODS AND RESULTS: We produced LQTS-type 2 (LQT2) hiPSC cardiomyocytes carrying a KCNH2 c.G1681A mutation in a IKr ion-channel pore, which caused impaired glycosylation and channel transport to cell surface. Allele-specific RNA interference (RNAi) directed towards the mutated KCNH2 mRNA caused knockdown, while leaving the wild-type mRNA unaffected. Electrophysiological analysis of patient-derived LQT2 hiPSC cardiomyocytes treated with mutation-specific siRNAs showed normalized action potential durations (APDs) and K(+) currents with the concurrent rescue of spontaneous and drug-induced arrhythmias (presented as early-afterdepolarizations). CONCLUSIONS: These findings provide in vitro evidence that allele-specific RNAi can rescue diseased phenotype in LQTS cardiomyocytes. This is a potentially novel route for the treatment of many autosomal-dominant-negative disorders, including those of the heart.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/genética , Síndrome de QT Prolongado/genética , Miocitos Cardíacos/fisiología , Células Madre Pluripotentes/fisiología , Interferencia de ARN/fisiología , Canal de Potasio ERG1 , Fenómenos Electrofisiológicos/genética , Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Terapia Genética , Humanos , Síndrome de QT Prolongado/fisiopatología , Síndrome de QT Prolongado/terapia , Mutación Missense/genética , Fenotipo , Transfección
8.
Nano Lett ; 14(12): 6997-7002, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25381734

RESUMEN

Resonantly driven quantum emitters offer a very promising route to obtain highly coherent sources of single photons required for applications in quantum information processing (QIP). Realizing this for on-chip scalable devices would be important for scientific advances and practical applications in the field of integrated quantum optics. Here we report on-chip quantum dot (QD) resonance fluorescence (RF) efficiently coupled into a single-mode waveguide, a key component of a photonic integrated circuit, with a negligible resonant laser background and show that the QD coherence is enhanced by more than a factor of 4 compared to off-resonant excitation. Single-photon behavior is confirmed under resonant excitation, and fast fluctuating charge dynamics are revealed in autocorrelation g((2)) measurements. The potential for triggered operation is verified in pulsed RF. These results pave the way to a novel class of integrated quantum-optical devices for on-chip quantum information processing with embedded resonantly driven quantum emitters.

9.
ACS Omega ; 9(4): 5014-5023, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38313497

RESUMEN

Transfection, defined as functional delivery of cell-internalized nucleic acids, is dependent on many factors linked to formulation, vector, cell type, and microenvironmental culture conditions. We previously developed a technology termed glycosaminoglycan (GAG)-binding enhanced transduction (GET) to efficiently deliver a variety of cargoes intracellularly, using GAG-binding peptides and cell penetrating peptides (CPPs) in the form of nanoparticles, using conventional cell culture. Herein, we demonstrate that the most simple GET transfection formulation (employing the FLR peptide) is relatively poor at transfecting cells at increasingly lower dosages. However, with an endosomally escaping version (FLR:FLH peptide formulations) we demonstrate more effective transfection of cells with lower quantities of plasmid (p)DNA in vitro. We assessed the ability of single and serial delivery of our formulations to readily transfect cells and determined that temperature, pH, and atmospheric pressure can significantly affect transfected cell number and expression levels. Cytocompatible temperatures that maintain high cell metabolism (20-37 °C) were the optimal for transfection. Interestingly, serial delivery can maintain and enhance expression without viability being compromised, and alkaline pH conditions can aid overall efficiencies. Positive atmospheric pressures can also improve the transgene expression levels generated by GET transfection on a single-cell level. Novel nanotechnologies and gene therapeutics such as GET could be transformative for future regenerative medicine strategies. It will be important to understand how such approaches can be optimized at the formulation and application levels in order to achieve efficacy that will be competitive with viral strategies.

10.
J Control Release ; 367: 209-222, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38244841

RESUMEN

Physical-based gene delivery via biolistic methods (such as the Helios gene gun) involve precipitation of nucleic acids onto microparticles and direct transfection through cell membranes of exposed tissue (e.g. skin) by high velocity acceleration. The glycosaminoglycan (GAG)-binding enhanced transduction (GET) system exploits novel fusion peptides consisting of cell-binding, nucleic acid condensing, and cell-penetrating domains, which enable enhanced transfection across multiple cell types. In this study, we combined chemical (GET) and physical (gene gun) DNA delivery systems, and hypothesized the combination would generate enhanced distribution and effective uptake in cells not initially transfected by biolistic penetration. Physicochemical characterization, optimization of bullet contents and transfection experiments in vitro in cell monolayers and engineered tissue demonstrated these formulations transfected efficiently, including DC2.4 dendritic cells. We incorporated these formulations into a biolistic format for gene gun by forming fireable dry bullets obtained via lyophilization (freeze drying). This system is simple and with enhanced scalability compared to conventional methods to generate bullets. Flushed GET bullet contents retained their ability to mediate transfection (17-fold greater and 13-fold greater reporter gene expression than standard spermidine bullets in the absence and presence of serum, respectively). Fired GET bullets in vitro (in cells and collagen gels) and in vivo (mice) showed increased reporter gene transfection compared to untreated controls, whilst maintaining cell viability in vitro and having no obvious toxicity in vivo. Lastly, a SARS-CoV-2 plasmid DNA vaccine with spike (S) protein-receptor binding domain (S-RBD) was delivered by gene gun using GET bullets. Specific T cell and antibody responses comparable to the conventional system were generated. The non-physical and physical combination of GET­gold-DNA carriers using gene gun shows potential as an alternative DNA delivery method that is scalable for mass deployable vaccination and intradermal gene delivery.


Asunto(s)
Péptidos de Penetración Celular , Ácidos Nucleicos , Vacunas de ADN , Ratones , Animales , Biolística/métodos , Péptidos de Penetración Celular/química , Transfección , ADN/química
11.
Development ; 137(18): 2973-80, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20736286

RESUMEN

Cells in the pluripotent ground state can give rise to somatic cells and germ cells, and the acquisition of pluripotency is dependent on the expression of Nanog. Pluripotency is conserved in the primitive ectoderm of embryos from mammals and urodele amphibians, and here we report the isolation of a Nanog ortholog from axolotls (axNanog). axNanog does not contain a tryptophan repeat domain and is expressed as a monomer in the axolotl animal cap. The monomeric form is sufficient to regulate pluripotency in mouse embryonic stem cells, but axNanog dimers are required to rescue LIF-independent self-renewal. Our results show that protein interactions mediated by Nanog dimerization promote proliferation. More importantly, they demonstrate that the mechanisms governing pluripotency are conserved from urodele amphibians to mammals.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes/metabolismo , Ambystoma mexicanum , Anfibios , Animales , Línea Celular , Proliferación Celular , Proteínas de Homeodominio/genética , Humanos , Mamíferos , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Unión Proteica
12.
J Control Release ; 360: 93-109, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37315695

RESUMEN

Insulin regulates blood glucose levels, and is the mainstay for the treatment of type-1 diabetes and type-2 when other drugs provide inadequate control. Therefore, effective oral Insulin delivery would be a significant advance in drug delivery. Herein, we report the use of the modified cell penetrating peptide (CPP) platform, Glycosaminoglycan-(GAG)-binding-enhanced-transduction (GET), as an efficacious transepithelial delivery vector in vitro and to mediate oral Insulin activity in diabetic animals. Insulin can be conjugated with GET via electrostatic interaction to form nanocomplexes (Insulin GET-NCs). These NCs (size and charge; 140 nm, +27.10 mV) greatly enhanced Insulin transport in differentiated in vitro intestinal epithelium models (Caco2 assays; >22-fold increased translocation) with progressive and significant apical and basal release of up-taken Insulin. Delivery resulted in intracellular accumulation of NCs, enabling cells to act as depots for subsequent sustained release without affecting viability and barrier integrity. Importantly Insulin GET-NCs have enhanced proteolytic stability, and retained significant Insulin biological activity (exploiting Insulin-responsive reporter assays). Our study culminates in demonstrating oral delivery of Insulin GET-NCs which can control elevated blood-glucose levels in streptozotocin (STZ)-induced diabetic mice over several days with serial dosing. As GET promotes Insulin absorption, transcytosis and intracellular release, along with in vivo function, our simplistic complexation platform could allow effective bioavailability of other oral peptide therapeutics and help transform the treatment of diabetes.


Asunto(s)
Péptidos de Penetración Celular , Diabetes Mellitus Experimental , Humanos , Ratones , Animales , Insulina/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Control Glucémico , Células CACO-2 , Péptidos de Penetración Celular/química , Transcitosis , Administración Oral , Glucemia
13.
Biotechnol Bioeng ; 109(10): 2630-41, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22511037

RESUMEN

Tissue function during development and in regenerative medicine completely relies on correct cell organization and patterning at micro and macro scales. We describe a rapid method for patterning mammalian cells including human embryonic stem cells (HESCs) and induced pluripotent stem cells (iPSCs) on elastomeric membranes such that micron-scale control of cell position can be achieved over centimeter-length scales. Our method employs surface engineering of hydrophobic polydimethylsiloxane (PDMS) membranes by plasma polymerization of allylamine. Deposition of plasma polymerized allylamine (ppAAm) using our methods may be spatially restricted using a micro-stencil leaving faithful hydrophilic ppAAm patterns. We employed airbrushing to create aerosols which deposit extracellular matrix (ECM) proteins (such as fibronectin and Matrigel™) onto the same patterned ppAAm rich regions. Cell patterns were created with a variety of well characterized cell lines (e.g., NIH-3T3, C2C12, HL1, BJ6, HESC line HUES7, and HiPSC line IPS2). Individual and multiple cell line patterning were also achieved. Patterning remains faithful for several days and cells are viable and proliferate. To demonstrate the utility of our technique we have patterned cells in a variety of configurations. The ability to rapidly pattern cells at high resolution over macro scales should aid future tissue engineering efforts for regenerative medicine applications and in creating in vitro stem cell niches.


Asunto(s)
Membranas/química , Polímeros , Ingeniería de Tejidos/métodos , Alilamina/química , Animales , Línea Celular , Dimetilpolisiloxanos/química , Elastómeros , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Células Madre Pluripotentes , Propiedades de Superficie
14.
Mol Ther ; 19(9): 1695-703, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21694703

RESUMEN

The limited ability of the heart to regenerate has prompted development of new systems to produce cardiomyocytes for therapeutics. While differentiation of human embryonic stem cells (hESCs) into cardiomyocytes has been well documented, the process remains inefficient and/or expensive, and progress would be facilitated by better understanding the early genetic events that cause cardiac specification. By maintaining a transgenic cardiac-specific MYH6-monomeric red fluorescent protein (mRFP) reporter hESC line in conditions that promote pluripotency, we tested the ability of combinations of 15 genes to induce cardiac specification. Screening identified GATA4 plus TBX5 as the minimum requirement to activate the cardiac gene regulatory network and produce mRFP(+) cells, while a combination of GATA4, TBX5, NKX2.5, and BAF60c (GTNB) was necessary to generate beating cardiomyocytes positive for cTnI and α-actinin. Including the chemotherapeutic agent, Ara-C, from day 10 of induced differentiation enriched for cTnI/α-actinin double positive cells to 45%. Transient expression of GTNB for 5-7 days was necessary to activate the cardiogenesis through progenitor intermediates in a manner consistent with normal heart development. This system provides a route to test the effect of different factors on human cardiac differentiation and will be useful in understanding the network failures that underlie disease phenotypes.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Redes Reguladoras de Genes , Actinina/metabolismo , Línea Celular , Proteínas Cromosómicas no Histona , Clonación Molecular , Citarabina/farmacología , Células Madre Embrionarias/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Vectores Genéticos , Corazón/embriología , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Lentivirus/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Miocitos Cardíacos/citología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Fluorescente Roja
15.
Biomater Adv ; 143: 213177, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36371970

RESUMEN

Nanotechnologies are being increasingly applied as systems for peptide and nucleic acid macromolecule drug delivery. However systemic targeting of these, or efficient topical and localized delivery remains an issue. A controlled release system that can be patterned and locally administered such as topically to accessible tissue (skin, eye, intestine) would therefore be transformative in realizing the potential of such strategies. We previously developed a technology termed GAG-binding enhanced transduction (GET) to efficiently deliver a variety of cargoes intracellularly, using GAG-binding peptides to mediate cell targeting, and cell penetrating peptides (CPPs) to promote uptake. Herein we demonstrate that the GET transfection system can be used with the moisturizing thermo-reversible hydrogel Pluronic-F127 (PF127) and methyl cellulose (MC) to mediate site specific and effective intracellular transduction and gene delivery through GET nanoparticles (NPs). We investigated hydrogel formulation and the temperature dependence of delivery, optimizing the delivery system. GET-NPs retain their activity to enhance gene transfer within our formulations, with uptake transferred to cells in direct contact with the therapy-laden hydrogel. By using Azowipe™ material in a bandage approach, we were able to show for the first-time localized gene transfer in vitro on cell monolayers. The ability to simply control localization of gene delivery on millimetre scales using contact-mediated transfer from moisture-providing thermo-reversible hydrogels will facilitate new drug delivery methods. Importantly our technology to site-specifically deliver the activity of novel nanotechnologies and gene therapeutics could be transformative for future regenerative medicine.


Asunto(s)
Péptidos de Penetración Celular , Hidrogeles , Hidrogeles/química , Poloxámero/química , Vendajes , Terapia Genética
16.
Mol Cancer ; 10(1): 7, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-21232089

RESUMEN

BACKGROUND: Breast cancer is a disease characterised by both genetic and epigenetic alterations. Epigenetic silencing of tumour suppressor genes is an early event in breast carcinogenesis and reversion of gene silencing by epigenetic reprogramming can provide clues to the mechanisms responsible for tumour initiation and progression. In this study we apply the reprogramming capacity of oocytes to cancer cells in order to study breast oncogenesis. RESULTS: We show that breast cancer cells can be directly reprogrammed by amphibian oocyte extracts. The reprogramming effect, after six hours of treatment, in the absence of DNA replication, includes DNA demethylation and removal of repressive histone marks at the promoters of tumour suppressor genes; also, expression of the silenced genes is re-activated in response to treatment. This activity is specific to oocytes as it is not elicited by extracts from ovulated eggs, and is present at very limited levels in extracts from mouse embryonic stem cells. Epigenetic reprogramming in oocyte extracts results in reduction of cancer cell growth under anchorage independent conditions and a reduction in tumour growth in mouse xenografts. CONCLUSIONS: This study presents a new method to investigate tumour reversion by epigenetic reprogramming. After testing extracts from different sources, we found that axolotl oocyte extracts possess superior reprogramming ability, which reverses epigenetic silencing of tumour suppressor genes and tumorigenicity of breast cancer cells in a mouse xenograft model. Therefore this system can be extremely valuable for dissecting the mechanisms involved in tumour suppressor gene silencing and identifying molecular activities capable of arresting tumour growth. These applications can ultimately shed light on the contribution of epigenetic alterations in breast cancer and advance the development of epigenetic therapies.


Asunto(s)
Ambystoma mexicanum , Neoplasias de la Mama/genética , Extractos Celulares/farmacología , Oocitos/química , Xenopus , Animales , Neoplasias de la Mama/patología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina , Digitonina/farmacología , Células Madre Embrionarias/química , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Histonas/metabolismo , Humanos , Metilación , Ratones , Proteínas de Microfilamentos/genética , Trasplante de Neoplasias , Regiones Promotoras Genéticas , Activación Transcripcional , Trasplante Heterólogo , Células Tumorales Cultivadas
17.
Mem Cognit ; 39(7): 1317-31, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21547605

RESUMEN

People create counterfactual alternatives that change an exceptional action to be like a usual one (e.g., "if he had placed his usual small bet he would have lost less"), as shown in Experiment 1. Experiments 2 and 3 eliminated and reversed this well-known effect: An exceptional action is instead changed to an exceptional alternative when it leads to a better outcome. Experiments 4 and 5 show that the reversal occurs whether or not the exceptional alternative is a justified action. The results indicate that the tendency to change an exceptional action to be like a usual one is guided by the optimality of the counterfactual outcome more than the exceptionality or justifiability of the action. The implications for theories of the counterfactual imagination are discussed.


Asunto(s)
Toma de Decisiones/fisiología , Pensamiento/fisiología , Adolescente , Adulto , Anciano , Femenino , Juego de Azar/psicología , Humanos , Juicio/fisiología , Masculino , Persona de Mediana Edad , Adulto Joven
18.
ACS Appl Nano Mater ; 4(1): 167-181, 2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33763629

RESUMEN

Non-viral delivery systems are generally of low efficiency, which limits their use in gene therapy and editing applications. We previously developed a technology termed glycosaminoglycan (GAG)-binding enhanced transduction (GET) to efficiently deliver a variety of cargos intracellularly; our system employs GAG-binding peptides, which promote cell targeting, and cell penetrating peptides (CPPs), which enhance endocytotic cell internalization. Herein, we describe a further modification by combining gene delivery and magnetic targeting with the GET technology. We associated GET peptides, plasmid (p)DNA, and iron oxide superparamagnetic nanoparticles (MNPs), allowing rapid and targeted GET-mediated uptake by application of static magnetic fields in NIH3T3 cells. This produced effective transfection levels (significantly higher than the control) with seconds to minutes of exposure and localized gene delivery two orders of magnitude higher in targeted over non-targeted cell monolayers using magnetic fields (in 15 min exposure delivering GFP reporter pDNA). More importantly, high cell membrane targeting by GET-DNA and MNP co-complexes and magnetic fields allowed further enhancement to endocytotic uptake, meaning that the nucleic acid cargo was rapidly internalized beyond that of GET complexes alone (GET-DNA). Magnetofection by MNPs combined with GET-mediated delivery allows magnetic field-guided local transfection in vitro and could facilitate focused gene delivery for future regenerative and disease-targeted therapies in vivo.

19.
PLoS Biol ; 5(3): e67, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17341133

RESUMEN

Regulation of transforming growth factor-beta (TGF-beta) signaling is critical in vertebrate development, as several members of the TGF-beta family have been shown to act as morphogens, controlling a variety of cell fate decisions depending on concentration. Little is known about the role of intracellular regulation of the TGF-beta pathway in development. E3 ubiquitin ligases target specific protein substrates for proteasome-mediated degradation, and several are implicated in signaling. We have shown that Arkadia, a nuclear RING-domain E3 ubiquitin ligase, is essential for a subset of Nodal functions in the embryo, but the molecular mechanism of its action in embryonic cells had not been addressed. Here, we find that Arkadia facilitates Nodal signaling broadly in the embryo, and that it is indispensable for cell fates that depend on maximum signaling. Loss of Arkadia in embryonic cells causes nuclear accumulation of phospho-Smad2/3 (P-Smad2/3), the effectors of Nodal signaling; however, these must be repressed or hypoactive as the expression of their direct target genes is reduced or lost. Molecular and functional analysis shows that Arkadia interacts with and ubiquitinates P-Smad2/3 causing their degradation, and that this is via the same domains required for enhancing their activity. Consistent with this dual function, introduction of Arkadia in homozygous null (-/-) embryonic stem cells activates the accumulated and hypoactive P-Smad2/3 at the expense of their abundance. Arkadia-/- cells, like Smad2-/- cells, cannot form foregut and prechordal plate in chimeras, confirming this functional interaction in vivo. As Arkadia overexpression never represses, and in some cells enhances signaling, the degradation of P-Smad2/3 by Arkadia cannot occur prior to their activation in the nucleus. Therefore, Arkadia provides a mechanism for signaling termination at the end of the cascade by coupling degradation of P-Smad2/3 with the activation of target gene transcription. This mechanism can account for achieving efficient and maximum Nodal signaling during embryogenesis and for rapid resetting of target gene promoters allowing cells to respond to dynamic changes in extracellular signals.


Asunto(s)
Transducción de Señal , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitina/fisiología , Animales , Secuencia de Bases , Quimera , Cartilla de ADN , Ratones , Ubiquitina-Proteína Ligasas
20.
Artículo en Inglés | MEDLINE | ID: mdl-32850720

RESUMEN

Developing non-viral gene therapy vectors that both protect and functionally deliver nucleic acid cargoes will be vital if gene augmentation and editing strategies are to be effectively combined with advanced regenerative medicine approaches. Currently such methodologies utilize high concentrations of recombinant growth factors, which result in toxicity and off-target effects. Herein we demonstrate the use of modified cell penetrating peptides (CPPs), termed Glycosaminoglycan (GAG)-binding Enhanced Transduction (GET) peptides with plasmid DNA (pDNA) encapsulated poly (lactic-co-glycolic acid) PLGA nanoparticles (pDNA-encapsulated PLGA NPs). In order to encapsulate the pDNA, it was first condensed with a cationic low molecular weight Poly L-Lysine (PLL) into 30-60 nm NPs followed by encapsulation in PLGA NPs by double emulsion; yielding encapsulation efficiencies (EE) of ∼30%. PLGA NPs complexed with GET peptides show enhanced intracellular delivery (up to sevenfold) and transfection efficiencies (up to five orders of magnitude). Moreover, the pDNA cargo has enhanced protection from nucleases (such as DNase I) promoting their translatability. As an example, we show these NPs efficiently deliver pBMP2 which can promote osteogenic differentiation in vitro. Gene delivery to human Mesenchymal Stromal Cells (hMSCs) inducing their osteogenic programming was confirmed by Alizarin red calcium staining and bone lineage specific gene expression (Q RT-PCR). By combining simplistic and FDA-approved PLGA polymer nanotechnology with the GET delivery system, therapeutic non-viral vectors could have significant impact in future cellular therapy and regenerative medicine applications.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA