Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Life (Basel) ; 13(12)2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38137903

RESUMEN

The gastrointestinal tract microbiome is essential for regulating nutrient absorption, gut immune function, and host growth and development. In the present study, we characterized the development of ileum and cecum microbiota in pullets throughout the rearing period, encompassing a period from the day of hatching to 18 weeks of age. The growth performance and intestinal microbiome (ileum and cecum) of pullets were analyzed at 1, 5, 11, and 18 weeks of age. The richness of the ileum and cecum bacterial communities (alpha diversity) was higher in pullets at 18 weeks of age than in those at 1 and 5 weeks of age. Microbiota from weeks 1, 5, 11, and 18 were distinctly grouped in a NMDS plot, representing beta diversity within the ileum. However, the results for cecum microbiota did not reveal evident separation among the different age groups in the weighted UniFrac. In conclusion, our findings demonstrate variations and diversification in ileum and cecum microbiota across different rearing stages in pullets. These insights have the potential to inform the development of nutritional strategies that promote gut health and contribute to the improved development of pullets.

2.
Front Neurosci ; 14: 558204, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33071737

RESUMEN

Huntington's disease (HD) is a devastating, autosomal-dominant inheritance disorder with the progressive loss of medium spiny neurons (MSNs) and corticostriatal connections in the brain. Cell replacement therapy has been proposed as a potential therapeutic strategy to treat HD. Among various types of stem cells, human-induced pluripotent stem cells (iPSCs) have received special attention to develop disease modeling and cell therapy for HD. In the present study, the therapeutic effects of neural precursor cells (NPCs) derived from a human iPSC line (1231A3-NPCs) were investigated in the quinolinic acid (QA)-lesioned rat model of HD. 1231A3-NPCs were transplanted into the ipsilateral striatum 1 week after QA lesioning, and the transplanted animals showed significant behavioral improvements for up to 12 weeks based on the staircase, rotarod, stepping, apomorphine-induced rotation, and cylinder tests. Transplanted 1231A3-NPCs also partially replaced the lost neurons, enhanced endogenous neurogenesis, reduced inflammatory responses, and reconstituted the damaged neuronal connections. Taken together, these results strongly indicate that NPCs derived from iPSCs can potentially be useful to treat HD in the future.

3.
Stem Cells ; 38(8): 936-947, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32374064

RESUMEN

Huntington's disease (HD) is a devastating, autosomal-dominant neurodegenerative disease, for which there are currently no disease-modifying therapies. Clinical trials to replace the damaged striatal medium spiny neurons (MSNs) have been attempted in the past two decades but have met with only limited success. In this study, we investigated whether a clonal, conditionally immortalized neural stem cell line (CTX0E03), which has already shown safety and signals of efficacy in chronic ischemic stroke patients, could rescue deficits seen in an animal model of HD. After CTX0E03 transplantation into the quinolinic acid-lesioned rat model of HD, behavioral changes were measured using the rotarod, stepping, and staircase tests. In vivo differentiation and neuronal connections of the transplanted CTX0E03 cells were evaluated with immunohistochemical staining and retrograde tracing with Fluoro-Gold. We found that transplantation of CTX0E03 gave rise to a significant behavioral improvement compared with the sham- or fibroblast-transplanted group. Transplanted CTX0E03 formed MSNs (DARPP-32) and GABAergic neurons (GABA, GAD65/67) with BDNF expression in the striatum, while cortically transplanted cells formed Tbr1-positive neurons. Using a retrograde label, we also found stable engraftment and connection of the transplanted cells with host brain tissues. CTX0E03 transplantation also reduced glial scar formation and inflammation, as well as increasing endogenous neurogenesis and angiogenesis. Overall, our results demonstrate that CTX0E03, a clinical-grade neural stem cell line, is effective for preclinical test in HD, and, therefore, will be useful for clinical development in the treatment of HD patients.


Asunto(s)
Enfermedad de Huntington/metabolismo , Células-Madre Neurales/metabolismo , Ácido Quinolínico/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Ratones , Clasificación del Tumor
4.
Exp Neurobiol ; 29(2): 130-137, 2020 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-32408403

RESUMEN

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by abnormally expanded CAG repeats in the huntingtin gene. The huntingtin gene mutation leads to the progressive degeneration of striatal GABAergic medium spiny neurons (MSN) and reduces the level of brain-derived neurotrophic factor (BDNF) in HD patient's brain. BDNF is an essential neurotrophic factor for the cortico-striatal synaptic activity and the survival of GABAergic neurons. In this study, we transplanted BDNF-overexpressing human neural stem cells (HB1.F3.BDNF) into the contra-lateral side of unilateral quinolinic acid (QA)-lesioned striatum of HD rat model. The results of in vivo transplantation were monitored using various behavioral tests, 4.7 T animal magnetic resonance imaging (MRI) and immunohistochemical staining. We observed that the QA-lesioned rats receiving HB1.F3.BDNF cells exhibited significant behavioral improvements in the stepping, rotarod and apomorphine-induced rotation tests. Interestingly, contralaterally transplanted cells were migrated to the QA-lesioned striatum and the size of lateral ventricle was reduced. Histological analyses further revealed that the transplanted cells, which had migrated to the QA lesion site, were differentiated into the cells of GABAergic, MSN-type neurons expressing DARPP-32, and neural networks were established between the transplanted cells and the host brain, as revealed by retrograde tracing. Finally, there was a significant reduction of inflammatory response in HB1.F3.BDNF-transplanted HD animal model, compared with vehicle-transplanted group. Taken together, these results suggest that HB1.F3.BDNF can be an effective therapeutic strategy to treat HD patients in the future.

5.
Acta Vet Hung ; 67(4): 610-618, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31842597

RESUMEN

The 3D8 single-chain variable fragment (scFv) is a mini-antibody sequence with independent nuclease activity that shows antiviral effects against all types of viruses in chickens and mice. In this study, chickens were treated daily with an oral dose of 109 CFU Lactobacillus paracasei (L. paracasei) expressing either a secreted or anchored 3D8 scFv for three weeks. After L. paracasei administration, the chickens were challenged with avian influenza virus (AIV). From each experimental group, three chickens were directly infected with 100 µL of 107.5 EID50/mL H9N2 AIV and seven chickens were indirectly challenged through contact transmission. oropharyngeal and cloacal swab samples were collected at 3, 5, 7, and 9 days post-inoculation (dpi) from AIV-challenged chickens, AIV Shedding titres were measured by quantitative real-time PCR. Contact transmission in the chickens that were fed 3D8 scFv-secreting L. paracasei showed a significant reduction in viral shedding when compared with other groups. These results suggest that L. paracasei secreting 3D8 provides a basis for the development of ingestible antiviral probiotics with activity against AIV.


Asunto(s)
Pollos , Gripe Aviar/tratamiento farmacológico , Lacticaseibacillus paracasei/química , Enfermedades de las Aves de Corral/tratamiento farmacológico , Probióticos/administración & dosificación , Animales , Subtipo H9N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H9N2 del Virus de la Influenza A/fisiología , Gripe Aviar/virología , Lacticaseibacillus paracasei/genética , Enfermedades de las Aves de Corral/virología , Esparcimiento de Virus/efectos de los fármacos
6.
Exp Mol Med ; 50(4): 1-12, 2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29650950

RESUMEN

The human umbilical cord is a promising source of mesenchymal stromal cells (MSCs). Intravenous administration of human umbilical cord-derived MSCs (IV-hUMSCs) showed a favorable effect in a rodent stroke model by a paracrine mechanism. However, its underlying therapeutic mechanisms must be determined for clinical application. We investigated the therapeutic effects and mechanisms of our good manufacturing practice (GMP)-manufactured hUMSCs using various cell doses and delivery time points in a rodent model of stroke. IV-hUMSCs at a dose of 1 × 106 cells at 24 h after stroke improved functional deficits and reduced neuronal damage by attenuation of post-ischemic inflammation. Transcriptome and immunohistochemical analyses showed that interleukin-1 receptor antagonist (IL-1ra) was highly upregulated in ED-1-positive inflammatory cells in rats treated with IV-hUMSCs. Treatment with conditioned medium of hUMSCs increased the expression of IL-1ra in a macrophage cell line via activation of cAMP-response element-binding protein (CREB). These results strongly suggest that the attenuation of neuroinflammation mediated by endogenous IL-1ra is an important therapeutic mechanism of IV-hUMSCs for the treatment of stroke.


Asunto(s)
Proteína Antagonista del Receptor de Interleucina 1/genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Cordón Umbilical/citología , Animales , Encéfalo/metabolismo , Isquemia Encefálica/etiología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Neuronas/metabolismo , Neuroprotección , Ratas , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/terapia
7.
Acta Neuropathol ; 132(4): 577-92, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27221146

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder of the central nervous system (CNS) that is defined by a CAG expansion in exon 1 of the huntingtin gene leading to the production of mutant huntingtin (mHtt). To date, the disease pathophysiology has been thought to be primarily driven by cell-autonomous mechanisms, but, here, we demonstrate that fibroblasts derived from HD patients carrying either 72, 143 and 180 CAG repeats as well as induced pluripotent stem cells (iPSCs) also characterized by 143 CAG repeats can transmit protein aggregates to genetically unrelated and healthy host tissue following implantation into the cerebral ventricles of neonatal mice in a non-cell-autonomous fashion. Transmitted mHtt aggregates gave rise to both motor and cognitive impairments, loss of striatal medium spiny neurons, increased inflammation and gliosis in associated brain regions, thereby recapitulating the behavioural and pathological phenotypes which characterizes HD. In addition, both in vitro work using co-cultures of mouse neural stem cells with 143 CAG fibroblasts and the SH-SY5Y human neuroblastoma cell line as well as in vivo experiments conducted in newborn wild-type mice suggest that exosomes can cargo mHtt between cells triggering the manifestation of HD-related behaviour and pathology. This is the first evidence of human-to-mouse prion-like propagation of mHtt in the mammalian brain; a finding which will help unravel the molecular bases of HD pathology as well as to lead to the development of a whole new range of therapies for neurodegenerative diseases of the CNS.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Células Madre Pluripotentes Inducidas/citología , Proteínas Mutantes/metabolismo , Neuronas/citología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Niño , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/terapia , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones , Neuronas/patología
8.
Poult Sci ; 95(4): 912-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26908883

RESUMEN

Cathelicidins form a family of vertebrate-specific immune molecules with an evolutionarily conserved gene structure. We analyzed the expression patterns of cathelicidin genes (CAMP, CATH3, and CATHB1) in chicken bone marrow cells (BMCs) and chicken embryonic fibroblasts (CEFs). We found that CAMP and CATHB1 were significantly up-regulated in BMCs, whereas the expression of CATH3 did not differ significantly between BMCs and CEFs. To study the mechanism underlying the up-regulation of cathelicidin genes in BMCs, we predicted the transcription factors (TFs) that bind to the 5'-flanking regions of cathelicidin genes. CEBPA, EBF1, HES1, MSX1, and ZIC3 were up-regulated in BMCs compared to CEFs. Subsequently, when a siRNA-mediated knockdown assay was performed for MSX1, the expression of CAMP and CATHB1 was decreased in BMCs. We also showed that the transcriptional activity of the CAMP promoter was decreased by mutation of the MSX1-binding sites present within the 5'-flanking region of CAMP. These results increase our understanding of the regulatory mechanisms controlling cathelicidin genes in BMCs.


Asunto(s)
Proteínas Aviares/genética , Catelicidinas/genética , Pollos/genética , Regulación de la Expresión Génica , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Proteínas Aviares/metabolismo , Células de la Médula Ósea/metabolismo , Catelicidinas/metabolismo , Embrión de Pollo , Pollos/metabolismo , Fibroblastos/metabolismo
9.
Mol Reprod Dev ; 82(12): 967-75, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26297836

RESUMEN

Early chick embryogenesis is governed by a complex mechanism involving transcriptional and post-transcriptional regulation, although how post-transcriptional processes influence the balance between pluripotency and differentiation during early chick development have not been previously investigated. Here, we characterized the microRNA (miRNA) signature associated with differentiation in the chick embryo, and found that as expression of the gga-let-7 family increases through early development, expression of their direct targets, TGFBR1 and LIN28B, decreases; indeed, gga-let-7a-5p and gga-let-7b miRNAs directly bind to TGFBR1 and LIN28B transcripts. Our data further indicate that TGFBR1 and LIN28B maintain pluripotency by regulating POUV, NANOG, and CRIPTO. Therefore, gga-let-7 miRNAs act as post-transcriptional regulators of differentiation in blastodermal cells by repressing the expression of the TGFBR1 and LIN28B, which intrinsically controls blastodermal cell differentiation in early chick development.


Asunto(s)
Proteínas Aviares/biosíntesis , Pollos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Animales , Embrión de Pollo , Receptor Tipo I de Factor de Crecimiento Transformador beta
10.
Mol Reprod Dev ; 82(12): 957-66, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26256125

RESUMEN

The mammary gland serves as a valuable bioreactor system for the production of recombinant proteins in lactating animals. Pharmaceutical-grade recombinant protein can be harvested from the milk of transgenic animals that carry a protein of interest under the control of promoter regions genes encoding milk proteins. Whey acidic protein (WAP), for example, is predominantly expressed in the mammary gland and is regulated by lactating hormones during pregnancy. We cloned the 5'-flanking region of the porcine WAP gene (pWAP) to confirm the sequence elements in its promoter that are required for gene-expression activity. In the present study, we investigated how lactogenic hormones--including prolactin, hydrocortisone, and insulin--contribute to the transcriptional activation of the pWAP promoter region in mammalian cells, finding that these hormones activate STAT5 signaling, which in turn induce gene expression via STAT5 binding sites in its 5'-flanking region. To confirm the expression and hormonal regulation of the 5'-flanking region of pWAP in vivo, we generated transgenic mice expressing human recombinant granulocyte colony stimulating factor (hCSF2) in the mammary gland under the control of the pWAP promoter. These mice secreted hCSF2 protein in their milk at levels ranging from 242 to 1,274.8 ng/ml. Collectively, our findings show that the pWAP promoter may be useful for confining the expression of foreign proteins to the mammary gland, where they can be secreted along with milk.


Asunto(s)
Glándulas Mamarias Animales/metabolismo , Proteínas de la Leche/biosíntesis , Leche/metabolismo , Regiones Promotoras Genéticas , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Animales , Femenino , Humanos , Lactancia , Ratones , Proteínas de la Leche/genética , Embarazo , Factor de Transcripción STAT5/genética , Porcinos
11.
Cytotherapy ; 17(8): 1090-103, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26031742

RESUMEN

BACKGROUND AIMS: Adipose-derived mesenchymal stromal cells (AD-MSCs) have high proliferative capacity and ability to secrete trophic factors. Although intra-arterial (IA) transplantation of stem cells induces efficient engraftment to the host brain, it is unclear whether engrafted cells exert their long-term therapeutic effects through a bystander mechanism or a cell replacement mechanism. METHODS: After induction of ischemia in rats by middle cerebral artery occlusion, we transplanted human AD-MSCs into their carotid arteries with the use of a micro-needle, and we then investigated the therapeutic effects during the early and late phases of ischemia by means of in vivo magnetic resonance imaging, functional and histological analyses. RESULTS: During the early phase of cerebral ischemia, IA transplantation of AD-MSCs attenuated inflammation and enhanced endogenous neurogenesis. Transplanted animals showed a marked improvement in functional tests during the early phase of cerebral ischemia that was less prominent but still significant during the late phase of cerebral ischemia. Although the transplanted cells effectively migrated to the infarct area, only a small number of engrafted cells survived at 8 weeks after transplantation and differentiated into neuronal, glial and endothelial cells. CONCLUSIONS: IA transplantation of human AD-MSCs provides an effective therapeutic modality in a rodent model of stroke, of which the main effects are mediated by a bystander mechanism at the early phase of ischemia.


Asunto(s)
Isquemia Encefálica/cirugía , Infarto de la Arteria Cerebral Media/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Neuroprotección/fisiología , Accidente Cerebrovascular/cirugía , Tejido Adiposo/citología , Adulto , Animales , Efecto Espectador , Diferenciación Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Células Madre Mesenquimatosas/citología , Neurogénesis/fisiología , Ratas , Ratas Sprague-Dawley , Adulto Joven
12.
In Vitro Cell Dev Biol Anim ; 51(3): 222-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25381035

RESUMEN

The laying hen is the best model for oviduct growth and development. The chicken oviduct produces the egg components, including the egg white and eggshell. However, the mechanism of egg component production during oviduct development requires further investigation. Vitelline membrane outer layer protein 1 (VMO-1) is found in the outer layer of the vitelline membrane of avian eggs. Comparison of the chicken VMO-1 protein-coding sequence and the human, mouse, rat, and bovine VMO-1 proteins via multiple sequence alignment analysis revealed high degrees of homology of 55%, 53%, 48%, and 54%, respectively. Although the avian homologue of VMO-1 is highly expressed in the magnum of the oviduct, little is known about the transcriptional and posttranscriptional regulation of VMO-1 during oviduct development. The results of this study revealed that estrogen induces VMO-1 messenger RNA (mRNA) expression in oviduct cells in vitro. The expression of genes interacting with VMO-1 by RNA interference (RNAi) functional analysis revealed that ovomucin expression was decreased by VMO-1 silencing. In addition, gga-miR-1623, 1552-3p, and 1651-3p influenced VMO-1 expression via its 3'-UTR, suggesting the posttranscriptional regulation of VMO-1 expression in chickens. Collectively, these results suggest that VMO-1 is an estrogen-induced gene that is posttranscriptionally regulated by microRNAs (miRNAs). The present study may contribute to an understanding of egg component production during chicken oviduct development.


Asunto(s)
Proteínas Aviares/genética , Pollos/crecimiento & desarrollo , Pollos/genética , Regulación de la Expresión Génica , MicroARNs/metabolismo , Oviductos/metabolismo , Membrana Vitelina/metabolismo , Envejecimiento/genética , Secuencia de Aminoácidos , Animales , Proteínas Aviares/metabolismo , Secuencia de Bases , Bases de Datos de Ácidos Nucleicos , Estradiol/farmacología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Genoma , MicroARNs/genética , Datos de Secuencia Molecular , Oviductos/efectos de los fármacos , Oviductos/crecimiento & desarrollo , Filogenia , Reproducibilidad de los Resultados , Alineación de Secuencia , Análisis de Secuencia de Proteína , Transcripción Genética/efectos de los fármacos , Membrana Vitelina/efectos de los fármacos
13.
Mol Reprod Dev ; 81(12): 1103-14, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25394196

RESUMEN

Early chick development is a systematic process governed by the concerted action of multiple mechanisms that regulate transcription and post-transcriptional processes. Post-transcriptional microRNA-mediated regulation, with regard to lineage specification and differentiation in early chick development, requires further investigation. Here, we characterize the transcriptional and post-transcriptional regulation mechanisms in undifferentiated chick blastodermal cells. Expression of the miR-302 cluster, POUV, SOX2, and STAT5B decreased in a time-dependent manner in early chick development. We found that POUV, SOX2, and STAT5B regulate the transcription of the miR-302 cluster, as its 5'-flanking region contains binding elements for each transcription factor. Additionally, POUV, SOX2, and STAT5B maintain pluripotency by regulating genes containing the miR-302 cluster target sequence. For example, microRNAs from the miR-302 cluster can bind to PBX3 and E2F7 transcripts, thus acting as a post-transcriptional regulator that maintains the undifferentiated state of blastodermal cells by balancing the expression of genes related to pluripotency and differentiation. Based on these results, we suggest that both transcriptional and post-transcriptional regulation of the miR302 cluster is critical for intrinsically controlling the undifferentiated state of chick embryonic blastodermal cells. These findings may help our understanding of the cellular and molecular mechanisms that underlie developmental decisions during early chick development.


Asunto(s)
Embrión de Pollo/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , MicroARNs/fisiología , Modelos Biológicos , Factores de Transcripción/fisiología , Animales , Embrión de Pollo/metabolismo , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Luciferasas , MicroARNs/metabolismo , Interferencia de ARN/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOX/fisiología , Factor de Transcripción STAT5/metabolismo , Factores de Transcripción/metabolismo
14.
Int J Stem Cells ; 7(1): 43-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24921027

RESUMEN

Induced pluripotent stem cells (iPSCs) generated from somatic cells of patients can provide immense opportunities to model human diseases, which may lead to develop novel therapeutics. Huntington's disease (HD) is a devastating neurodegenerative genetic disease, with no available therapeutic options at the moment. We recently reported the characteristics of a HD patient-derived iPSC carrying 72 CAG repeats (HD72-iPSC). In this study, we investigated the in vivo roles of HD72-iPSC in the YAC128 transgenic mice, a commonly used HD mouse model carrying 128 CAG repeats. To do this, we transplanted HD72-iPSC-derived neural precursors into the striatum of YAC128 mice bilaterally and observed a significant behavioral improvement in the grafted mice. Interestingly, the transplanted HD72-iPSC-derived neural precursors formed GABAeric neurons efficiently, but no EM48-positive protein aggregates were detected at 12 weeks after transplantation. Taken together, these results indicate no HD pathology was developed from the grafted cells, or no transmission of HD pathology from the host to the graft occurred at 12 weeks post-transplantation.

15.
Stem Cell Res ; 12(2): 506-16, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24463498

RESUMEN

Pluripotent stem cells can be derived from preimplantation and postimplantation mouse embryos. Embryonic stem cells (ESCs) derived from blastocysts are in a "naive" pluripotent state and meet all of the criteria for pluripotency, including the ability to generate live pups through tetraploid complementation. Epiblast stem cells (EpiSCs) derived from postimplantation epiblasts are in a "primed" pluripotent state. ESCs and EpiSCs show different phenotypes and gene expression patterns, and EpiSCs are thought to be less pluripotent than ESCs. In this study, we addressed whether EpiSCs can be differentiated into specialized cell types in vitro. To do this, we first derived EpiSCs from E5.5-6.5 mouse embryos containing the Oct4-GFP transgene. We found that EpiSCs expressed pluripotency markers and differentiated into all three germ layers in intro and in vivo. Interestingly, EpiSCs also efficiently differentiated into a homogenous population of neural stem cells (NSCs) in vitro. The EpiSC-derived NSCs (EpiSC-NSCs) expressed NSC markers (Nestin, Sox2, and Musashi), self-renewed for more than 20 passages, and differentiated into neuronal and glial neural cell subtypes in vitro. We then transplanted the EpiSC-NSCs into the neonatal mouse brains, and found that they were able to survive and differentiate into robust neurons and glial cells in the mouse brains, demonstrating that primed pluripotent EpiSCs efficiently form functional NSCs. We compared the global gene expression patterns of NSCs differentiated from EpiSC-NSCs, ESCs, and brain tissue and found that the expression patterns of most genes, including pluripotency and NSC specificity, were similarly clustered, but that the developmental process-related genes were distantly clustered. Moreover, the global gene expression pattern of brain-derived NSCs was more similar to that of ESC-derived NSCs than that of EpiSC-derived NSCs. Taken together, these results indicate that although NSCs, regardless of their origins, display very similar in vitro and in vivo differentiation properties, their global gene expression profiles may differ, depending on the pluripotency state, i.e., naive or primed.


Asunto(s)
Estratos Germinativos/citología , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Fosfatasa Alcalina/metabolismo , Animales , Diferenciación Celular/fisiología , Perfilación de la Expresión Génica , Genómica , Estratos Germinativos/metabolismo , Inmunohistoquímica , Ratones , Células-Madre Neurales/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , Análisis de Matrices Tisulares
16.
Exp Mol Med ; 45: e53, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24232252

RESUMEN

The transplantation of neural precursor cells (NPCs) is known to be a promising approach to ameliorating behavioral deficits after stroke in a rodent model of middle cerebral artery occlusion (MCAo). Previous studies have shown that transplanted NPCs migrate toward the infarct region, survive and differentiate into mature neurons to some extent. However, the spatiotemporal dynamics of NPC migration following transplantation into stroke animals have yet to be elucidated. In this study, we investigated the fates of human embryonic stem cell (hESC)-derived NPCs (ENStem-A) for 8 weeks following transplantation into the side contralateral to the infarct region using 7.0T animal magnetic resonance imaging (MRI). T2- and T2*-weighted MRI analyses indicated that the migrating cells were clearly detectable at the infarct boundary zone by 1 week, and the intensity of the MRI signals robustly increased within 4 weeks after transplantation. Afterwards, the signals were slightly increased or unchanged. At 8 weeks, we performed Prussian blue staining and immunohistochemical staining using human-specific markers, and found that high percentages of transplanted cells migrated to the infarct boundary. Most of these cells were CXCR4-positive. We also observed that the migrating cells expressed markers for various stages of neural differentiation, including Nestin, Tuj1, NeuN, TH, DARPP-32 and SV38, indicating that the transplanted cells may partially contribute to the reconstruction of the damaged neural tissues after stroke. Interestingly, we found that the extent of gliosis (glial fibrillary acidic protein-positive cells) and apoptosis (TUNEL-positive cells) were significantly decreased in the cell-transplanted group, suggesting that hESC-NPCs have a positive role in reducing glia scar formation and cell death after stroke. No tumors formed in our study. We also performed various behavioral tests, including rotarod, stepping and modified neurological severity score tests, and found that the transplanted animals exhibited significant improvements in sensorimotor functions during the 8 weeks after transplantation. Taken together, these results strongly suggest that hESC-NPCs have the capacity to migrate to the infarct region, form neural tissues efficiently and contribute to behavioral recovery in a rodent model of ischemic stroke.


Asunto(s)
Movimiento Celular , Células Madre Embrionarias/trasplante , Infarto de la Arteria Cerebral Media/cirugía , Células-Madre Neurales/trasplante , Desempeño Psicomotor , Animales , Apoptosis , Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
17.
BMB Rep ; 46(8): 404-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23977988

RESUMEN

Extracellular superoxide dismutase (EC-SOD) is a metalloprotein and functions as an antioxidant enzyme. In this study, we used lentiviral vectors to generate transgenic chickens that express the human EC-SOD gene. The recombinant lentiviruses were injected into the subgerminal cavity of freshly laid eggs. Subsequently, the embryos were incubated to hatch using phases II and III of the surrogate shell ex vivo culture system. Of 158 injected embryos, 16 chicks (G0) hatched and were screened for the hEC-SOD by PCR. Only 1 chick was identified as a transgenic bird containing the transgene in its germline. This founder (G0) bird was mated with wild-type hens to produce transgenic progeny, and 2 transgenic chicks (G1) were produced. In the generated transgenic hens (G2), the hEC-SOD protein was expressed in the egg white and showed antioxidant activity. These results highlight the potential of the chicken for production of biologically active proteins in egg white.


Asunto(s)
Superóxido Dismutasa/metabolismo , Animales , Animales Modificados Genéticamente , Pollos/metabolismo , Yema de Huevo/metabolismo , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Lentivirus/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Superóxido Dismutasa/genética
18.
Int J Oncol ; 42(3): 921-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23314408

RESUMEN

Malignant gliomas are the most common primary brain tumor in adults. A number of genes have been implicated in glioblastoma including mutation and deletion of PTEN. PTEN is a regulator of PI3K-mediated Akt signaling pathways and has been recognized as a therapeutic target in glioblastoma. To achieve potent therapeutic inhibition of the PI3K-Akt pathway in glioblastoma, it is essential to understand the interplay between the regulators of its activation. Here, ectopic expression of PTEN in the U-87MG human glioblastoma-astrocytoma cell line is shown to result in the depletion of glioblastoma stem cells (GSCs) and to cause growth retardation and senescence. These effects are likely to be associated with PTEN-mediated cooperative perturbation of Akt and Stat3 signals. Using an in vivo rat model of glioblastoma, we showed that PTEN-overexpressing U-87MG cells failed to induce tumor formation, while untreated U-87MG cells did so. Furthermore, cells expressing the phosphorylated form of Stat3 were completely absent from the brain of rats implanted with PTEN-overexpressing U-87MG cells. Based on these results, PTEN appears to function as a crucial inhibitor of GSCs and as an inducer of senescence, suggesting that functional enhancement of the PTEN pathway will be useful to provide a therapeutic strategy for targeting glioblastoma.


Asunto(s)
Glioblastoma/metabolismo , Glioblastoma/patología , Células Madre Neoplásicas/fisiología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Senescencia Celular , Humanos , Masculino , Trasplante de Neoplasias , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Mol Biotechnol ; 54(2): 504-14, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22945876

RESUMEN

Bacterial Artificial Chromosome (BAC) clones are widely used for retrieving genomic DNA sequences for gene targeting. In this study, low-copy-number plasmids pBAC-FB, pBAC-FC, and pBAC-DE, which carry the F plasmid replicon, were generated from pBACe3.6. pBAC-FB was successfully used to retrieve a sequence of a BAC that was resistant to retrieval by a high-copy-number plasmid via λ Red-mediated recombineering (gap-repair cloning). This plasmid was also used to retrieve two other genes from BAC, indicating its general usability retrieving genes from BAC. The retrieved genes were manipulated in generating targeting vectors for gene knockouts by recombineering. The functionality of the targeting vector was further validated in a targeting experiment with C57BL/6 embryonic stem cells. The low-copy-number plasmid pBAC-FB is a plasmid of choice to retrieve toxic DNA sequences from BACs and to manipulate them to generate gene-targeting constructs by recombineering.


Asunto(s)
Cromosomas Artificiales Bacterianos , Marcación de Gen/métodos , Vectores Genéticos/genética , Plásmidos/genética , Animales , Clonación Molecular/métodos , ADN/genética , Células Madre Embrionarias/metabolismo , Ratones , Ratones Endogámicos C57BL , Recombinación Genética/genética , Replicón
20.
Cell Transplant ; 22(8): 1427-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23044029

RESUMEN

Ischemic stroke mainly caused by middle cerebral artery occlusion (MCAo) is a major type of stroke, but there are currently very limited therapeutic options for its cure. Neural stem cells (NSCs) or neural precursor cells (NPCs) derived from various sources are known to survive and improve neurological functions when they are engrafted in animal models of stroke. Induced pluripotent stem cells (iPSCs) generated from somatic cells of patients are novel cells that promise the autologous cell therapy for stroke. In this study, we successfully differentiated iPSCs derived from human fibroblasts into NPCs and found their robust therapeutic potential in a rodent MCAo stroke model. We observed the significant graft-induced behavioral recovery, as well as extensive neural tissue formation. Animal MRI results indicated that the majority of contralaterally transplanted iPSC-derived NPCs migrated to the peri-infarct area, showing a pathotropism critical for tissue recovery. The transplanted animals exhibited the significant reduction of stroke-induced inflammatory response, gliosis and apoptosis, and the contribution to the endogenous neurogenesis. Our results demonstrate that iPSC-derived NPCs are effective cells for the treatment of stroke.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Trasplante de Células Madre , Accidente Cerebrovascular/terapia , Animales , Apoptosis , Conducta Animal , Diferenciación Celular , Línea Celular , Rastreo Celular , Modelos Animales de Enfermedad , Gliosis/patología , Humanos , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Inflamación/patología , Imagen por Resonancia Magnética , Masculino , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Neurogénesis , Neuronas/citología , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA