Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nucleic Acids Res ; 48(15): 8601-8616, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32687187

RESUMEN

The CRISPR-Cas9 system is widely used for target-specific genome engineering. CRISPR-Cas12a (Cpf1) is one of the CRISPR effectors that controls target genes by recognizing thymine-rich protospacer adjacent motif (PAM) sequences. Cas12a has a higher sensitivity to mismatches in the guide RNA than does Cas9; therefore, off-target sequence recognition and cleavage are lower. However, it tolerates mismatches in regions distant from the PAM sequence (TTTN or TTN) in the protospacer, and off-target cleavage issues may become more problematic when Cas12a activity is improved for therapeutic purposes. Therefore, we investigated off-target cleavage by Cas12a and modified the Cas12a (cr)RNA to address the off-target cleavage issue. We developed a CRISPR-Cas12a that can induce mutations in target DNA sequences in a highly specific and effective manner by partially substituting the (cr)RNA with DNA to change the energy potential of base pairing to the target DNA. A model to explain how chimeric (cr)RNA guided CRISPR-Cas12a and SpCas9 nickase effectively work in the intracellular genome is suggested. Chimeric guide-based CRISPR- Cas12a genome editing with reduced off-target cleavage, and the resultant, increased safety has potential for therapeutic applications in incurable diseases caused by genetic mutations.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Asociadas a CRISPR/genética , Sistemas CRISPR-Cas/genética , ADN/genética , Endodesoxirribonucleasas/genética , ARN Guía de Kinetoplastida/genética , Disparidad de Par Base/genética , División del ADN , Edición Génica , Humanos , Modelos Moleculares , Mutación/genética , Conformación de Ácido Nucleico , ARN/genética , ARN Circular/genética
2.
Angiogenesis ; 22(2): 281-293, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30471052

RESUMEN

Testis-specific protein, Y-encoded like (TSPYL) family proteins (TSPYL1-6), which are members of the nucleosome assembly protein superfamily, have been determined to be involved in the regulation of various cellular functions. However, the potential role of TSPYL family proteins in endothelial cells (ECs) has not been determined. Here, we demonstrated that the expression of TSPYL5 is highly enriched in human ECs such as human umbilical vein endothelial cells (HUVECs) and human pluripotent stem cell-differentiated ECs (hPSC-ECs). Importantly, TSPYL5 overexpression was shown to promote EC proliferation and functions, such as migration and tube formation, by downregulating p53 expression. Adriamycin-induced senescence was markedly blocked by TSPYL5 overexpression. In addition, the TSPYL5 depletion-mediated loss of EC functions was blocked by p53 inhibition. Significantly, TSPYL5 overexpression promoted angiogenesis in Matrigel plug and wound repair in a mouse skin wound healing model in vivo. Our results suggest that TSPYL5, a novel angiogenic regulator, plays a key role in maintaining endothelial integrity and function. These findings extend the understanding of TSPYL5-dependent mechanisms underlying the regulation of p53-related functions in ECs.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/fisiología , Neovascularización Fisiológica/genética , Proteínas Nucleares/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Regulación hacia Abajo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Proteína p53 Supresora de Tumor/metabolismo
3.
Stem Cells ; 36(10): 1525-1534, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29873154

RESUMEN

The importance of alternative splicing (AS) events in pluripotency regulation has been highlighted by the determination of different roles and contributions of different splice isoforms of pluripotency-related genes and by the identification of distinct pluripotency-related splicing factors. In particular, epithelial splicing regulatory protein 1 (ESRP1) has been characterized as an essential splicing factor required for the regulation of human pluripotency and differentiation. Nevertheless, a detailed molecular characterization of ESRP1 (mRNA splice variants 1-6) in human pluripotency is lacking. In this study, we determined that ESRP1 splice variants are differentially expressed in undifferentiated and differentiated human pluripotent stem cells (PSCs). Undifferentiated human PSCs predominantly expressed the ESRP1 v1, v4, and v5, and their expression was downregulated upon differentiation. Ectopic expression of ESRP1 v1, v4, or v5 enhanced the pluripotent reprogramming of human fibroblasts and restored the ESRP1 knockdown-mediated reduction of reprogramming efficiency. Notably, undifferentiated human PSCs expressed the cell surface protein CD44 variant 3 (CD44 v3), and isoform switching from CD44 v3 to CD44 variant 6 (CD44 v6) occurred upon differentiation. Importantly, the human PSC-specific ESRP1 variants influenced CD44 v3 expression. CD44 knockdown or inhibition of binding of CD44 with its major ligand, hyaluronan, significantly induced the loss of human PSC pluripotency and the reduction of reprogramming efficiency. Our results demonstrate that the effect of ESRP1 and CD44 on human PSC pluripotency is isoform-dependent and that ESRP1-induced CD44 v3 is functionally associated with human PSC pluripotency control. Stem Cells 2018;36:1525-1534.


Asunto(s)
Receptores de Hialuranos/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas de Unión al ARN/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Humanos
4.
Molecules ; 25(1)2019 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-31877894

RESUMEN

In CRISPR genome editing, CRISPR proteins form ribonucleoprotein complexes with guide RNAs to bind and cleave the target DNAs with complete sequence complementarity. CRISPR genome editing has a high potential for use in precision gene therapy for various diseases, including cancer and genetic disorders, which are caused by DNA mutations within the genome. However, several studies have shown that targeting the DNA via sequence complementarity is imperfect and subject to unintended genome editing of other genomic loci with similar sequences. These off-target problems pose critical safety issues in the therapeutic applications of CRISPR technology, with particular concerns in terms of the genome editing of pathogenic point mutations, where non-mutant alleles can become an off-target with only a one-base difference. In this study, we sought to assess a novel CRISPR genome editing technique that has been proposed to achieve a high specificity by positioning the mismatches within the protospacer adjacent motif (PAM) sequence. To this end, we compared the genome editing specificities of the PAM-based and conventional methods on an oncogenic single-base mutation in the endothelial growth factor receptor (EGFR). The results indicated that the PAM-based method provided a significantly increased genome editing specificity for pathogenic mutant alleles with single-base precision.


Asunto(s)
Edición Génica/métodos , Mutación Puntual , Sistemas CRISPR-Cas , ADN Complementario/genética , ADN Intergénico/genética , Receptores ErbB/genética , Humanos
5.
Genome Biol ; 23(1): 92, 2022 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-35410288

RESUMEN

Prime editing can induce a desired base substitution, insertion, or deletion in a target gene using reverse transcriptase after nick formation by CRISPR nickase. In this study, we develop a technology that can be used to insert or replace external bases in the target DNA sequence by linking reverse transcriptase to the Francisella novicida Cas9, which is a CRISPR-Cas9 ortholog. Using FnCas9(H969A) nickase, the targeting limitation of existing Streptococcus pyogenes Cas9 nickase [SpCas9(H840A)]-based prime editing is dramatically extended, and accurate prime editing is induced specifically for the target genes in human cell lines.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Desoxirribonucleasa I/metabolismo , Francisella , Humanos , ADN Polimerasa Dirigida por ARN
6.
Mol Ther Nucleic Acids ; 28: 353-362, 2022 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-35505967

RESUMEN

The clustered regularly interspaced short palindromic repeats (CRISPR)-Cas12a system is composed of a Cas12a effector that acts as a DNA-cleaving endonuclease and a crispr RNA (crRNA) that guides the effector to the target DNA. It is considered a key molecule for inducing target-specific gene editing in various living systems. Here, we improved the efficiency and specificity of the CRISPR-Cas12a system through protein and crRNA engineering. In particular, to optimize the CRISPR-Cas12a system at the molecular level, we used a chimeric DNA-RNA guide chemically similar to crRNA to maximize target sequence specificity. Compared with the wild-type (wt)-Cas12a system, when using enhanced Cas12a system (en-Cas12a), the efficiency and target specificity improved on average by 2.58 and 2.77 times, respectively. In our study, when the chimeric DNA-RNA-guided en-Cas12a effector was used, the gene-editing efficiency and accuracy were simultaneously increased. These findings could contribute to highly accurate genome editing, such as human gene therapy, in the near future.

7.
Phytother Res ; 25(8): 1141-7, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21287651

RESUMEN

Individual pharmacokinetic differences for herb-drug interaction have been associated with genetic variations of the multidrug resistance (MDR) gene. A high level expression of MDR protein increases cellular efflux and might decrease drug sensitivity. This study investigated the drug efflux activity difference of human MDR1 triallelic variant 2677G/T/A (rs2032582), as a nonsynonymous 893Ala/Ser/Thr, using Xenopus laevis oocytes and MDR1 overexpressing LLC-PK1 cells. Two MDR1 variants (2667T/893Ser and 2667A/893Thr) were generated using human MDR1 cDNA (2677G/893Ala). No significant difference in the expression of MDR1 893Ala/Ser/Thr was found in X. laevis oocytes. However, the MDR1 2667A/893Thr variant interestingly showed a significant decrease of efflux activity for both digoxin and daunorubicin compared with those of 893Ala and 893Ser variants. In further investigation assessing the inhibitory effects of three herbal extracts on MDR1, 893Ala and 893Ser showed significant decreases of efflux activities in treatments with P. cocos (p = 0.005 for 893Ser) and D. dasycarpus (p = 0.0009 for 893Ala; p = 0.002 for 893Ser) in X. laevis oocytes. The results in this study suggest that herbal medicines could interact with other drugs and change the therapeutic effects depending on the genetic polymorphisms of individuals.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Interacciones de Hierba-Droga , Extractos Vegetales/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Animales , Transporte Biológico/efectos de los fármacos , Daunorrubicina/farmacocinética , Dictamnus/química , Digoxina/antagonistas & inhibidores , Digoxina/farmacocinética , Genes MDR/efectos de los fármacos , Variación Genética , Humanos , Células LLC-PK1 , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Extractos Vegetales/farmacocinética , Poria/química , Rhus/química , Porcinos , Xenopus laevis
8.
Genes (Basel) ; 12(5)2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-34068253

RESUMEN

Although comparative genome-wide transcriptomic analysis has provided insight into the biology of human induced pluripotent stem cell-derived mesenchymal stem cells (iMSCs), the distinct alternative splicing (AS) signatures of iMSCs remain elusive. Here, we performed Illumina RNA sequencing analysis to characterize AS events in iMSCs compared with tissue-derived MSCs. A total of 4586 differentially expressed genes (|FC| > 2) were identified between iMSCs and umbilical cord blood-derived MSCs (UCB-MSCs), including 2169 upregulated and 2417 downregulated genes. Of these, 164 differentially spliced events (BF > 20) in 112 genes were identified between iMSCs and UCB-MSCs. The predominant type of AS found in iMSCs was skipped exons (43.3%), followed by retained introns (19.5%), alternative 3' (15.2%) and 5' (12.8%) splice sites, and mutually exclusive exons (9.1%). Functional enrichment analysis showed that the differentially spliced genes (|FC| > 2 and BF > 20) were mainly enriched in functions associated with focal adhesion, extracellular exosomes, extracellular matrix organization, cell adhesion, and actin binding. Splice isoforms of selected genes including TRPT1, CNN2, and AP1G2, identified in sashimi plots, were further validated by RT-PCR analysis. This study provides valuable insight into the biology of iMSCs and the translation of mechanistic understanding of iMSCs into therapeutic applications.


Asunto(s)
Empalme Alternativo/genética , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Mesenquimatosas/fisiología , Células Cultivadas , Regulación hacia Abajo/genética , Exosomas/genética , Matriz Extracelular/genética , Adhesiones Focales/genética , Expresión Génica/genética , Humanos , Transcriptoma/genética , Regulación hacia Arriba/genética
9.
J Biol Res (Thessalon) ; 28(1): 22, 2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34814951

RESUMEN

BACKGROUND: Peroxiredoxins (Prxs) are antioxidant enzymes that protect cells from oxidative stress induced by several factors. They regulate several signaling pathways, such as metabolism, immune response, and intracellular reactive oxygen species (ROS) homeostasis. Epithelial-mesenchymal transition (EMT) is a transforming process that induces the loss of epithelial features of cancer cells and the gain of the mesenchymal phenotype. The EMT promotes metastasis and cancer cell progression mediated by several pathways, such as mitogen-activated protein kinases (MAPKs) and epigenetic regulators. METHODS: We used Prx6 overexpressed and downregulated HCT116 cells to study the mechanism between Prx6 and colon cancer. The expression of Prx6, GAPDH, Snail, Twist1, E-cadherin, Vimentin, N-cadherin, ERK, p-ERK, p38, p-p38, JNK, and p-JNK were detected by Western blotting. Additionally, an animal study for xenograft assay was conducted to explore the function of Prx6 on tumorigenesis. Cell proliferation and migration were determined by IncuCyte Cell Proliferation and colony formation assays. RESULTS: We confirmed that the expression of Prx6 and EMT signaling highly occurs in HCT116 compared with that in other colon cancer cell lines. Prx6 regulates the EMT signaling pathway by modulating EMT-related transcriptional repressors and mesenchymal genes in HCT116 colon cancer cells. Under the Prx6-overexpressed condition, HCT116 cells proliferation increased significantly. Moreover, the HCT116 cells proliferation decreased in the siPrx6-treated cells. Eleven days after HCT116 cell injection, Prx6 was overexpressed in the HCT116-injected mice, and the tumor volume increased significantly compared with that of the control mice. Furthermore, Prx6 regulates EMT signaling through p38 phosphorylation in colon cancer cells. CONCLUSION: We suggested that Prx6 regulates EMT signaling pathway through p38 phosphorylation modulation in HCT116 colon cancer cells.

10.
Nat Biomed Eng ; 5(11): 1360-1376, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34341536

RESUMEN

Efficacious and accessible sources of natural killer (NK) cells would widen their use as immunotherapeutics, particularly for solid cancers. Here, we show that human somatic cells can be directly reprogrammed into NK cells with a CD56brightCD16bright phenotype using pluripotency transcription factors and an optimized reprogramming medium. The directly reprogrammed NK cells have strong innate-adaptive immunomodulatory activity and are highly potent against a wide range of cancer cells, including difficult-to-treat solid cancers and cancer stem cells. Both directly reprogrammed NK cells bearing a cancer-specific chimeric antigen receptor and reprogrammed NK cells in combination with antibodies competent for antibody-dependent cell-mediated cytotoxicity led to selective anticancer effects with augmented potency. The direct reprogramming of human somatic cells into NK cells is amenable to the production of autologous and allogeneic NK cells, and will facilitate the design and testing of cancer immunotherapies and combination therapies.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Inmunoterapia , Células Asesinas Naturales , Neoplasias/terapia
11.
J Neurochem ; 113(6): 1565-76, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20367752

RESUMEN

Previously, we reported the phosphorylation of moesin induced by electroconvulsive shock in rat brain and by glutamate in immortalized rat hippocampal cells. However, the function of phosphorylated moesin in differentiated neurons is not well understood. In this study, we observed that glutamate induces phosphorylation of ezrin/radixin/moesin proteins (ERM) in cultured hippocampal cells and that phosphorylated ERM localizes at the newly formed filopodia of neurites. The glutamate-induced phosphorylation of ERM is calcium-dependent, and inhibition of protein kinase C abolishes ERM phosphorylation as well as RhoA activation. The inhibitions of RhoA and RhoA kinase also diminishes the glutamate-induced ERM phosphorylation in cultured hippocampal cells. The knock-down of moesin or the inhibition of ERM phosphorylation results in the reduction of glutamate-induced filopodia protrusion and diminishes the increase in active synaptic boutons induced by glutamate treatment. These results indicate that glutamate-induced phosphorylation of ERM proteins in primary cultured differentiated hippocampal neurons is mediated by calcium-dependent protein kinase C, RhoA and RhoA kinase, and the phosphorylated ERM protein is necessary for the formation of filopodial protrusion and may be involved in pre-synaptic trafficking.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Hipocampo/citología , Neuronas/ultraestructura , Seudópodos/fisiología , Receptores de Glutamato/fisiología , Animales , Células Cultivadas , Quelantes/farmacología , Homólogo 4 de la Proteína Discs Large , Relación Dosis-Respuesta a Droga , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Ácido Glutámico/farmacología , Proteínas Fluorescentes Verdes/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Fosforilación/efectos de los fármacos , Seudópodos/efectos de los fármacos , Compuestos de Piridinio/metabolismo , Compuestos de Amonio Cuaternario/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo , Factores de Tiempo , Transfección/métodos , Proteína de Unión al GTP rhoA/metabolismo
12.
Stem Cell Res Ther ; 11(1): 257, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32586386

RESUMEN

BACKGROUND: Schwann cells (SCs) are primarily responsible for regeneration and repair of the peripheral nervous system (PNS). Renewable and lineage-restricted SC precursors (SCPs) are considered highly desirable and promising cell sources for the production of SCs and for studies of SC lineage development, but SCPs are extremely limited. Here, we present a novel direct conversion strategy for the generation of human SCPs, capable of differentiating into functional SCs. METHODS: Easily accessible human skin fibroblast cells were directly induced into integration-free SCPs using episomal vectors (Oct3/4, Klf4, Sox2, L-Myc, Lin28 and p53 shRNA) under SCP lineage-specific chemically defined medium conditions. Induced SCPs (iSCPs) were further examined for their ability to differentiate into SCs. The identification and functionality of iSCPs and iSCP-differentiated SCs (iSCs) were confirmed according to morphology, lineage-specific markers, neurotropic factor secretion, and/or standard functional assays. RESULTS: Highly pure, Sox 10-positive of iSCPs (more than 95% purity) were generated from human skin fibroblasts within 3 weeks. Established iSCPs could be propagated in vitro while maintaining their SCP identity. Within 1 week, iSCPs could efficiently differentiate into SCs (more than 95% purity). The iSCs were capable of secreting various neurotrophic factors such as GDNF, NGF, BDNF, and NT-3. The in vitro myelinogenic potential of iSCs was assessed by myelinating cocultures using mouse dorsal root ganglion (DRG) neurons or human induced pluripotent stem cell (iPSC)-derived sensory neurons (HSNs). Furthermore, iSC transplantation promoted sciatic nerve repair and improved behavioral recovery in a mouse model of sciatic nerve crush injury in vivo. CONCLUSIONS: We report a robust method for the generation of human iSCPs/iSCs that might serve as a promising cellular source for various regenerative biomedical research and applications, such as cell therapy and drug discovery, especially for the treatment of PNS injury and disorders.


Asunto(s)
Células Madre Pluripotentes Inducidas , Traumatismos de los Nervios Periféricos , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Factor 4 Similar a Kruppel , Ratones , Regeneración Nerviosa , Células de Schwann , Nervio Ciático
13.
ACS Appl Mater Interfaces ; 12(19): 21424-21432, 2020 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-32319751

RESUMEN

Personal accessories such as glasses and watches that we usually carry in our daily life can yield useful information from the human body, yet most of them are limited to exercise-related parameters or simple heart rates. Since these restricted characteristics might arise from interfaces between the body and items as one of the main reasons, an interface design considering such a factor can provide us with biologically meaningful data. Here, we describe three-dimensional-printed, personalized, multifunctional electronic eyeglasses (E-glasses), not only to monitor various biological phenomena but also to propose a strategy to coordinate the recorded data for active commands and game operations for human-machine interaction (HMI) applications. Soft, highly conductive composite electrodes embedded in the E-glasses enable us to achieve reliable, continuous recordings of physiological activities. UV-responsive, color-tunable lenses using an electrochromic ionic gel offer the functionality of both eyeglass and sunglass modes, and accelerometers provide the capability of tracking precise human postures and behaviors. Detailed studies of electrophysiological signals including electroencephalogram and electrooculogram demonstrate the feasibility of smart electronic glasses for practical use as a platform for future HMI systems.


Asunto(s)
Interfaces Cerebro-Computador , Anteojos , Monitoreo Fisiológico/instrumentación , Dispositivos Electrónicos Vestibles , Humanos , Monitoreo Fisiológico/métodos , Impresión Tridimensional , Juegos de Video
14.
Nat Commun ; 11(1): 3596, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32681048

RESUMEN

CRISPR effectors, which comprise a CRISPR-Cas protein and a guide (g)RNA derived from the bacterial immune system, are widely used for target-specific genome editing. When the gRNA recognizes genomic loci with sequences that are similar to the target, deleterious mutations can occur. Off-target mutations with a frequency below 0.5% remain mostly undetected by current genome-wide off-target detection techniques. Here we report a method to effectively detect extremely small amounts of mutated DNA based on predicted off-target-specific amplification. In this study, we used various genome editors to induce intracellular genome mutations, and the CRISPR amplification method detected off-target mutations at a significantly higher rate (1.6~984 fold increase) than an existing targeted amplicon sequencing method. In the near future, CRISPR amplification in combination with genome-wide off-target detection methods will allow detection of genome editor-induced off-target mutations with high sensitivity and in a non-biased manner.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , ADN/genética , Sistemas CRISPR-Cas , Edición Génica , Humanos , Mutación , ARN Guía de Kinetoplastida/genética
15.
J Korean Med Sci ; 24(1): 146-51, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19270828

RESUMEN

This study was designed to determine whether early gabapentin treatment has a protective analgesic effect on neuropathic pain and compared its effect to the late treatment in a rat neuropathic model, and as the potential mechanism of protective action, the alpha(2)delta(1)-subunit of the voltage-dependent calcium channel (alpha(2)delta(1)-subunit) was evaluated in both sides of the L5 dorsal root ganglia (DRG). Neuropathic pain was induced in male Sprague-Dawley rats by a surgical ligation of left L5 nerve. For the early treatment group, rats were injected with gabapentin (100 mg/kg) intraperitoneally 15 min prior to surgery and then every 24 hr during postoperative day (POD) 1-4. For the late treatment group, the same dose of gabapentin was injected every 24 hr during POD 8-12. For the control group, L5 nerve was ligated but no gabapentin was administered. In the early treatment group, the development of allodynia was delayed up to POD 10, whereas allodynia was developed on POD 2 in the control and the late treatment group (p<0.05). The alpha(2)delta(1)-subunit was up-regulated in all groups, however, there was no difference in the level of the alpha(2)delta(1)-subunit among the three groups. These results suggest that early treatment with gabapentin offers some protection against neuropathic pain but it is unlikely that this action is mediated through modulation of the alpha(2)delta(1)-subunit in DRG.


Asunto(s)
Aminas/uso terapéutico , Analgésicos/uso terapéutico , Canales de Calcio/metabolismo , Ácidos Ciclohexanocarboxílicos/uso terapéutico , Neuralgia/tratamiento farmacológico , Ácido gamma-Aminobutírico/uso terapéutico , Aminas/administración & dosificación , Analgésicos/administración & dosificación , Animales , Canales de Calcio/genética , Canales de Calcio Tipo L , Ácidos Ciclohexanocarboxílicos/administración & dosificación , Modelos Animales de Enfermedad , Gabapentina , Inyecciones Intraperitoneales , Ligadura , Masculino , Neuralgia/metabolismo , Dimensión del Dolor , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Nervios Espinales/cirugía , Regulación hacia Arriba , Ácido gamma-Aminobutírico/administración & dosificación
16.
Stem Cell Reports ; 8(6): 1714-1726, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28506533

RESUMEN

Schwann cells play a crucial role in successful nerve repair and regeneration by supporting both axonal growth and myelination. However, the sources of human Schwann cells are limited both for studies of Schwann cell development and biology and for the development of treatments for Schwann cell-associated diseases. Here, we provide a rapid and scalable method to produce self-renewing Schwann cell precursors (SCPs) from human pluripotent stem cells (hPSCs), using combined sequential treatment with inhibitors of the TGF-ß and GSK-3 signaling pathways, and with neuregulin-1 for 18 days under chemically defined conditions. Within 1 week, hPSC-derived SCPs could be differentiated into immature Schwann cells that were functionally confirmed by their secretion of neurotrophic factors and their myelination capacity in vitro and in vivo. We propose that hPSC-derived SCPs are a promising, unlimited source of functional Schwann cells for treating demyelination disorders and injuries to the peripheral nervous system.


Asunto(s)
Enfermedades Desmielinizantes/terapia , Células Madre Pluripotentes/metabolismo , Células de Schwann/trasplante , Axones/fisiología , Diferenciación Celular/efectos de los fármacos , Autorrenovación de las Células , Células Cultivadas , Reprogramación Celular , Proteína GAP-43/metabolismo , Humanos , Neurregulina-1/farmacología , Células Madre Pluripotentes/citología , Receptor de Factor de Crecimiento Nervioso/metabolismo , Regeneración , Células de Schwann/citología , Células de Schwann/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Stem Cell Res ; 22: 43-53, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28595116

RESUMEN

Spliceosomes are the core host of pre-mRNA splicing, allowing multiple protein isoforms to be produced from a single gene. Herein, we reveal that spliceosomes are more abundant in human pluripotent stem cells (hPSs), including human embryonic stem cells (hESs) and human induced pluripotent stem cells (hiPSs), than non-hPSs, and their presence is associated with high transcriptional activity. Supportively, spliceosomal components involved in the catalytically active pre-mRNA splicing step were mainly co-localized with hPS spliceosomes. By profiling the gene expression of 342 selected splicing factors, we found that 71 genes were significantly altered during the reprogramming of human somatic cells into hiPSs. Among them, SNRPA1, SNRPD1, and PNN were significantly up-regulated during the early stage of reprogramming, identified as hub genes by interaction network and cluster analysis. SNRPA1, SNRPD1, or PNN depletion led to a pronounced loss of pluripotency and significantly blocked hiPS generation. SNRPA1, SNRPD1, and PNN co-localized with the hPS spliceosomes, physically interacted with each other, and positively influenced the appearance of hPS spliceosomes. Our data suggest that SNRPA1, SNRPD1, and PNN are key players in the regulation of pluripotency-specific spliceosome assembly and the acquisition and maintenance of pluripotency.


Asunto(s)
Moléculas de Adhesión Celular/genética , Proteínas Nucleares/genética , Células Madre Pluripotentes/fisiología , Empalme del ARN/genética , Ribonucleoproteína Nuclear Pequeña U1/genética , Empalmosomas/genética , Proteínas Nucleares snRNP/genética , Línea Celular , Humanos , ARN Nuclear Pequeño/genética , Empalmosomas/metabolismo , Transcriptoma
18.
Lab Chip ; 16(6): 959-76, 2016 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-26891410

RESUMEN

Implantable devices have provided various potential diagnostic options and therapeutic methods in diverse medical fields. A variety of hard-material-based implantable electrodes have been developed. However, several limitations for their chronic implantation remain, including mechanical mismatches at the interface between the electrode and the soft tissue, and biocompatibility. Soft-material-based implantable devices are suitable candidates for complementing the limitations of hard electrodes. Advances in microtechnology and materials science have largely solved many challenges, such as optimization of shape, minimization of infection, enhancement of biocompatibility and integration with components for diverse functions. Significant strides have also been made in mechanical matching of electrodes to soft tissue. In this review, we provide an overview of recent advances in soft-material-based implantable electrodes for medical applications, categorized according to their implantation site and material composition. We then review specific applications in three categories: neuroprosthetics, neural signal recording, and neuromodulation. Finally, we describe various strategies for the future development and application of implantable, soft-material-based devices.


Asunto(s)
Investigación Biomédica , Electrodos Implantados , Vías Nerviosas , Neurotransmisores/análisis , Materiales Biocompatibles , Humanos , Microelectrodos , Microtecnología
20.
Stem Cell Res ; 12(1): 60-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24145188

RESUMEN

The direct lineage reprogramming of somatic cells to other lineages by defined factors has led to innovative cell-fate-change approaches for providing patient-specific cells. Recent reports have demonstrated that four pluripotency factors (Oct4, Sox2, Klf4, and c-Myc) are sufficient to directly reprogram fibroblasts to other specific cells, including induced neural stem cells (iNSCs). Here, we show that mouse fibroblasts can be directly reprogrammed into midbrain dopaminergic neuronal progenitors (DPs) by temporal expression of the pluripotency factors and environment containing sonic hedgehog and fibroblast growth factor 8. Within thirteen days, self-renewing and functional induced DPs (iDPs) were generated. Interestingly, the inhibition of both Jak and Gsk3ß notably enhanced the iDP reprogramming efficiency. We confirmed the functionality of the iDPs by showing that the dopaminergic neurons generated from iDPs express midbrain markers, release dopamine, and show typical electrophysiological profiles. Our results demonstrate that the pluripotency factors-mediated direct reprogramming is an invaluable strategy for supplying functional and proliferating iDPs and may be useful for other neural progenitors required for disease modeling and cell therapies for neurodegenerative disorders.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Neuronas Dopaminérgicas/citología , Fibroblastos/citología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula , Reprogramación Celular/efectos de los fármacos , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Factor 8 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas Hedgehog/farmacología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Factor 4 Similar a Kruppel , Mesencéfalo/citología , Ratones , Piridinas/farmacología , Pirimidinas/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA