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










Base de datos
Intervalo de año de publicación
1.
Exp Neurol ; 379: 114870, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38897539

RESUMEN

BACKGROUND AND PURPOSE: The pathophysiological features of acute ischemic stroke (AIS) often involve dysfunction of the blood-brain barrier (BBB), characterized by the degradation of tight junction proteins (Tjs) leading to increased permeability. This dysfunction can exacerbate cerebral injury and contribute to severe complications. The permeability of the BBB fluctuates during different stages of AIS and is influenced by various factors. Developing effective therapies to restore BBB function remains a significant challenge in AIS treatment. High levels of vascular endothelial growth factor (VEGF) in the early stages of AIS have been shown to worsen BBB breakdown and stroke progression. Our study aimed to investigate the protective effects of the VEGF receptor inhibitor Axitinib on BBB dysfunction and cerebral ischemia/reperfusion-induced injury. METHODS: BEnd3 cell exposed to oxygen-glucose deprivation (OGD) model was constructed to estimate pharmacological activity of Axitinib (400 ng/ml) on anti-apoptosis and pathological barrier function recovery. In vivo, rats were subjected to a 1 h transient middle cerebral artery occlusion and 23 h reperfusion (tMCAO/R) to investigate the permeability of BBB and cerebral tissue damage. Axitinib was administered through the tail vein at the beginning of reperfusion. BBB integrity was assessed by Evans blue leakage and the expression levels of Tjs claudin-5 and occludin. RESULTS: Our research revealed that co-incubation with Axitinib enhanced the cell viability of OGD-insulted bEnd3 cells, decreased LDH leakage rate, and suppressed the expression of apoptosis-related proteins cytochrome C and Bax. Axitinib also mitigated the damage to Tjs and facilitated the restoration of transepithelial electrical resistance in OGD-insulted bEnd.3 cells. In vivo, Axitinib administration reduced intracerebral Evans blue leakage and up-regulated the expression of Tjs in the penumbra brain tissue in tMCAO/R rats. Notably, 10 mg/kg Axitinib exerted a significant anti-ischemic effect by decreasing cerebral infarct volume and brain edema volume, improving neurological function, and reducing pro-inflammatory cytokines IL-6 and TNF-α in the brain. CONCLUSIONS: Our study highlights Axitinib as a potent protectant of blood-brain barrier function, capable of promoting pathological blood-brain barrier recovery through VEGF inhibition and increased expression of tight junction proteins in AIS. This suggests that VEGF antagonism within the first 24 h post-stroke could be a novel therapeutic approach to enhance blood-brain barrier function and mitigate ischemia-reperfusion injury.

2.
Adv Healthc Mater ; 13(8): e2302939, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38117094

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative illness characterized by intracellular tau-phosphorylation, ß-amyloid (Aß) plaques accumulation, neuroinflammation, and impaired behavioral ability. Owing to the lack of effective brain delivery approaches and the presence of the blood-brain barrier (BBB), current AD therapeutic endeavors are severely limited. Herein, a multifunctional delivery system (RVG-DDQ/PDP@siBACE1) is elaborately combined with a protein kinase B (AKT) agonist (SC79) for facilitating RVG-DDQ/PDP@siBACE1 to target and penetrate BBB, enter brain parenchyma, and further accumulate in AD brain lesion. Moreover, compared with the unitary dose of RVG-DDQ/PDP@siBACE1, this collaborative therapy strategy exhibits a distinctive synergistic function including scavenging reactive oxygen species (ROS), decreasing of Aß production, alleviating neuroinflammation by promoting the polarized microglia into the anti-inflammatory M2-like phenotype and significantly enhancing the cognitive functions of AD mice. More strikingly, according to these results, an innovative signaling pathway "lncRNA MALAT1/miR-181c/BCL2L11" is found that can mediate the neuronal apoptosis of AD. Taken together, combining the brain targeted delivery system with noninvasive BBB opening can provide a promising strategy and platform for targeting treatment of AD and other neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Animales , Enfermedad de Alzheimer/terapia , Barrera Hematoencefálica/patología , Enfermedades Neuroinflamatorias , Péptidos beta-Amiloides/metabolismo , Especies Reactivas de Oxígeno/metabolismo
3.
J Nanobiotechnology ; 21(1): 422, 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37957722

RESUMEN

BACKGROUND: Despite being a common malignant tumor, the molecular mechanism underlying the initiation and progression of triple-negative breast cancers (TNBCs) remain unclear. Tumor-associated macrophages (TAMs) are often polarized into a pro-tumor phenotype and are associated with a poor prognosis of TNBCs. Exosomes, important mediators of cell-cell communication, can be actively secreted by donor cells to reprogram recipient cells. The functions and molecular mechanisms of tumor cell-derived exosomes in TNBCs progression and TAMs reprogramming urgently need to be further explored. RESULTS: We demonstrated that tumor cell-derived exosomes enriched with miR-184-3p were taken up by macrophages to inhibit JNK signaling pathway by targeting EGR1, thereby inducing M2 polarization of macrophages and synergistically promoting tumor progression. Nanoparticles loaded with oncogene c-Myc inhibitor JQ1 could suppress the polarization process by reducing Rac1-related exosome uptake by macrophage. More importantly, it was found for the first time that tumor-suppressive miR-184-3p was actively sorted into exosomes by binding to RNA-binding protein heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1), thus facilitating tumor cell proliferation and metastasis by relieving the inhibitory effect of miR-184-3p on Mastermind-like 1 (MAML1). Overexpressing miR-184-3p in tumor cells and simultaneously knocking down hnRNPA2B1 to block its secretion through exosomes could effectively inhibit tumor growth and metastasis. CONCLUSIONS: Our study revealed that hnRNPA2B1-mediated exosomal transfer of tumor-suppressive miR-184-3p from breast cancer cells to macrophages was an important mediator of TNBCs progression, providing new insights into TNBCs pathogenesis and therapeutic strategies.


Asunto(s)
MicroARNs , Neoplasias , MicroARNs/genética , MicroARNs/metabolismo , Línea Celular Tumoral , Microambiente Tumoral , Proliferación Celular
4.
Int J Oncol ; 63(6)2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37888615

RESUMEN

Acidosis is a hallmark of the tumor microenvironment caused by the metabolic switch from glucose oxidative phosphorylation to glycolysis. It has been associated with tumor growth and progression; however, the precise mechanism governing how acidosis promotes metastatic dissemination has yet to be elucidated. In the present study, a long­term acidosis model was established using patient­derived lung cancer cells, to identify critical components of metastatic colonization via transcriptome profiling combined with both in vitro and in vivo functional assays, and association analysis using clinical samples. Xenograft inoculates of 1 or 10 acidotic cells mimicking circulating tumor cell clusters were shown to exhibit increased tumor incidence compared with their physiological pH counterparts. Transcriptomics revealed that profound remodeling of the extracellular matrix (ECM) occurred in the acidotic cells, including upregulation of the integrin subunit α­4 (ITGA4) gene. In clinical lung cancer, ITGA4 expression was found to be upregulated in primary tumors with metastatic capability, and this trait was retained in the corresponding secondary tumors. Expression of ITGA4 was markedly upregulated around the vasculogenic mimicry structures of the acidotic tumors, while acidotic cells exhibited a higher ability of vasculogenic mimicry in vitro. Acidosis was also found to induce the enrichment of side population cells, suggesting an enhanced resistance to noxious attacks of the tumor microenvironment. Taken together, these results demonstrated that acidosis actively contributed to tumor metastatic colonization, and novel mechanistic insights into the therapeutic management and prognosis of lung cancer were discussed.


Asunto(s)
Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/genética , Neovascularización Patológica/tratamiento farmacológico , Pronóstico , Pulmón/patología , Matriz Extracelular/metabolismo , Línea Celular Tumoral , Microambiente Tumoral
5.
Nanoscale ; 15(27): 11625-11646, 2023 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-37377137

RESUMEN

Ischemic stroke is characterized by high morbidity, disability, and mortality. Unfortunately, the only FDA-approved pharmacological thrombolytic, alteplase, has a narrow therapeutic window of only 4.5 h. Other drugs like neuroprotective agents have not been clinically used because of their low efficacy. To improve the efficacy of neuroprotective agents and the effectiveness of rescue therapies for hyperacute ischemic stroke, we investigated and verified the variation trends of the blood-brain barrier (BBB) permeability and regional cerebral blood flow over 24 h in rats that had ischemic strokes. Hypoperfusion and the biphasic increase of BBB permeability are still the main limiting factors for lesion-specific drug distribution and drug brain penetration. Herein, the nitric oxide donor hydroxyurea (HYD) was reported to downregulate the expression of tight junction proteins and upregulate intracellular nitric oxide content in the brain microvascular endothelial cells subjected to oxygen-glucose deprivation, which was shown to facilitate the transport of liposomes across  brain endothelial monolayer in an in vitro model. HYD also increased the BBB permeability and promoted microcirculation in the hyperacute phase of stroke. The neutrophil-like cell-membrane-fusogenic hypoxia-sensitive liposomes exhibited excellent performance in targeting the inflamed brain microvascular endothelial cells, enhancing cell association, and promoting rapid hypoxic-responsive release in the hypoxic microenvironment. Overall, the combined HYD and hypoxia-sensitive liposome dosing regimen effectively decreased the cerebral infarction volume and relieved neurological dysfunction in rats that had ischemic strokes; these therapies were involved in the anti-oxidative stress effect and the neurotrophic effect mediated by macrophage migration inhibitory factor.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Fármacos Neuroprotectores , Accidente Cerebrovascular , Ratas , Animales , Liposomas/metabolismo , Hidroxiurea/farmacología , Hidroxiurea/metabolismo , Hidroxiurea/uso terapéutico , Accidente Cerebrovascular Isquémico/metabolismo , Fármacos Neuroprotectores/farmacología , Células Endoteliales , Encéfalo/metabolismo , Barrera Hematoencefálica/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Hipoxia , Isquemia Encefálica/tratamiento farmacológico
6.
iScience ; 26(3): 106267, 2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36936782

RESUMEN

Microglia play an important role in neuroinflammation and neurodegeneration. Here, we report an approach for generating microglia-containing cerebral organoids derived from human pluripotent stem cells involving the supplementation of growth factors (FGF, EGF, heparin) and 10% CO2 culture conditions. Using this platform, Western Pacific Amyotrophic Lateral Sclerosis and Parkinsonism-Dementia Complex (ALS-PDC) cerebral organoids were generated from patient-derived induced pluripotent stem cells (iPSCs). These ALS-PDC-affected organoids had more reactive astrocytes and M1 microglia, and had fewer M2 microglia than their unaffected counterparts, leading to impaired microglia-mediated phagocytosis. RNA-seq analysis of ALS-PDC and control organoids indicated that the most significant changes were microglia- and astrocyte-related genes (IFITM1/2, TGF-ß, and GFAP). The most significantly downregulated pathway was type I interferon signaling. Interferon-gamma supplementation increased IFITM expression, enhanced microglia-mediated phagocytosis, and reduced beta-amyloid accumulation in ALS-PDC-affected network. The results demonstrated the feasibility of using microglia-containing organoids for the study of neurodegenerative diseases.

7.
Foods ; 11(2)2022 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-35053904

RESUMEN

Pre-cooked adzuki beans (Vigna angularis), which looks like dried adzuki bean, is easily cooked and preserved. This study aimed to optimize the microwave pre-cooked conditions on adzuki beans by applying the response surface methodology. The results showed that soaking time has a significant effect on the gelatinization degree of adzuki beans according to microwave time. The most suitable gelatinization and the sensory scores were obtained with a soaking time of 7.8 h, a microwave power of 830 W, and microwave time of 92 s. The pre-cooked treatment had no significant effect (p > 0.05) on the protein, free amino acid, fat and starch content of adzuki bean products. The results of SEM and polarized light microscopy showed that the surface and center of starch were damaged after microwave treatment. XRD showed that microwave pre-cooking did not change the crystal structure of starch and maintained the original order of type A structure while reducing the relative starch crystallinity. FT-IR showed that the pre-cooked treatment did not produce new structure in adzuki bean starch, but the ratio of 1047/1022 cm-1 was slightly decreased, indicating that the starch crystallization area decreased relative to the amorphous area and the relative crystallinity decreased. The results of FTIR were consistent with X-ray diffraction results. Therefore, microwaves improved the gelatinization of adzuki beans and made the pre-cooked adzuki beans more suitable.

8.
Toxicology ; 461: 152898, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34403730

RESUMEN

Silver nanoparticles (AgNPs) are widely used in medical and commercial products for their unique antibacterial functions. However, the impact of AgNPs on human neural development is not well understood. To investigate the effect of AgNPs on human neural development, various doses of 20 nm citrate-coated AgNP (AgSC) were administered to human embryonic stem cell derived neural progenitors during the neuronal differentiation. Immunofluorescence staining with neuronal progenitor markers SOX2 (sex determining region Y-box 2) and Nestin (VI intermediate filament protein) showed that AgSC inhibited rosette formation, neuronal progenitor proliferation, and neurite outgrowth. Furthermore, AgSC promoted astrocyte activation and neuronal apoptosis. These adverse effects can be partially recovered with ascorbic acid. A genome-wide transcriptome analysis of both AgSC treated and untreated samples indicated that the most up-graduated genes were a group of Metallothionein (1F, 1E, 2A) proteins, a metal-binding protein that plays an essential role in metal homeostasis, heavy metal detoxification, and cellular anti-oxidative defence. The most significantly down-regulated genes were neuronal differentiation 6 (NEUROD6) and fork head box G1 (FOXG1). GO analyse indicated that the regulation of cholesterol biosynthetic process, neuron differentiation, synapse organization and pattern specification, oliogenesis, and neuronal apoptosis were the most impacted biological processes. KEGG pathway analyse showed that the most significantly impacted pathways were C5 isoprenoid, axon guidance, Notch, WNT, RAS-MAPK signalling pathways, lysosome, and apoptosis. Our data suggests that AgSCs interfered with metal homeostasis and cholesterol biosynthesis which induced oxidative stress, inhibited neurogenesis, axon guidance, and promoted apoptosis. Supplementation with ascorbic acid could act as an antioxidant to prevent AgSC-mediated neurotoxicity.


Asunto(s)
Ácido Cítrico/química , Células Madre Embrionarias Humanas/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Plata , Apoptosis/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Colesterol/biosíntesis , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Embrionarias Humanas/citología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/genética , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
9.
Biomaterials ; 275: 120958, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34130142

RESUMEN

Tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) play an important role in the development of tumors by secreting a variety of cytokines or directly communicating with tumor cells, making TAMs-targeted therapeutic strategies very attractive. It has been reported that oncogene c-Myc is related to every aspect of the oncogenic process of tumor cells and the alternative activation of macrophages. Hence, we constructed a glycolipid nanocarrier containing ROS-responsive peroxalate linkages (CSOPOSA) for ROS-triggered release of drugs and further modified it with Ex 26 (Ex 26-CSOPOSA), a selective sphingosine 1-phosphate receptor 1 (S1PR1) antagonist, to achieve the dual-targeted delivery of the c-Myc inhibitor JQ1 via S1PR1, which is overexpressed on both tumor cells and TAMs, thereby inducing apoptosis of tumor cells, and blocking M2 polarization of macrophages. More strikingly, our studies found that JQ1 could effectively inhibit the migration of tumor cells induced by M2 macrophages-derived exosomes via blocking Caveolin-1 related intercellular exosome exchange through lncRNA H19 and miR-107. The in vivo results revealed that this dual-targeted delivery strategy effectively inhibited tumor growth and metastasis with less systemic toxicity, providing a potential method for effective tumor treatment.


Asunto(s)
Caveolina 1 , Exosomas , Neoplasias Experimentales/tratamiento farmacológico , Especies Reactivas de Oxígeno , Animales , Línea Celular Tumoral , Portadores de Fármacos , Liberación de Fármacos , Femenino , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-myc , Células RAW 264.7 , Microambiente Tumoral
10.
Mater Sci Eng C Mater Biol Appl ; 118: 111385, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33254992

RESUMEN

Diabetic foot ulcer, one of the most common diabetic complications, is a progressive wound occurred on the skin with irregularly delayed wound healing rate due to impaired metabolism and weak immune responses. Such chronic wound remains a serious healthcare burden to the diabetics since it is often associated with high risk of limb loss due to amputation and leads to a reduced survival consequently. To improve the efficiency of diabetic wound healing, a synthetic chitosan-based composite hydrogel named SNPECHG incorporating silver ions (Ag+) and nanoparticle-encapsulated epidermal growth factor (EGF) was developed in this study. The optimal effective dosages of 24-mM Ag+ and 60-µg mL-1 EGF for the SNPECHG manufacture were first determined based on the results of antibacterial, cytotoxicity, and cell growth examinations. We then characterized the optimized SNPECHG and found that the composite hydrogel was able to provide sustained release of Ag+ and EGF, and exhibited a significantly higher hydration capacities, including the swelling degree and equilibrium water content, in PBS than those in deionized water, showing that the developed SNPECHG is highly applicable in the ion-rich environment such as chronic wound site. According to the results of in vivo study using diabetic rats, the one with SNPECHG exhibited a markedly enhanced wound healing effect compared with the other settings since day 3, and may reach a degree of wound closure of 97% at day 14 that was 7.4% (P < 0.05) and 18.9% (P < 0.05) higher than the values gained from the groups with the commercial dressing HeraDerm and gauze, respectively. Moreover, the wound treated with the SNPECHG exhibited thorough re-epithelization, sufficient collagen deposition, and accelerated collagen maturation confirmed by the histological analysis. Taken all together, we anticipate that the SNPECHG is highly advantageous for use in the clinical diabetic/chronic wound treatment.


Asunto(s)
Quitosano , Diabetes Mellitus Experimental , Nanopartículas , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Hidrogeles , Ratas , Plata , Cicatrización de Heridas
11.
Front Plant Sci ; 11: 1255, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32922425

RESUMEN

Poly(A) tail length (PAL) has been implicated in the regulation of mRNA translation activities. However, the extent of such regulation at the transcriptome level is less understood in plants. Herein, we report the development and optimization of a large-scale sequencing technique called the Assay for PAL-sequencing (APAL-seq). To explore the role of PAL on post-transcriptional modification and translation, we performed PAL profiling of Arabidopsis transcriptome in response to heat shock. Transcripts of 2,477 genes were found to have variable PAL upon heat treatments. Further study of the transcripts of 14 potential heat-responsive genes identified two distinct groups of genes. In one group, PAL was heat stress-independent, and in the other, PAL was heat stress-sensitive. Meanwhile, the protein expression of HSP70 and HSP17.6C was determined to test the impact of PAL on translational activity. In the absence of heat stress, neither gene demonstrated protein expression; however, under gradual or abrupt heat stress, both transcripts showed enhanced protein expression with elongated PAL. Interestingly, HSP17.6C protein levels were positively correlated with the severity of heat treatment and peaked when treated with abrupt heat. Our results suggest that plant genes have a high variability of PALs and that PAL contributes to swift posttranslational stress responses.

12.
Environ Int ; 127: 324-332, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30953815

RESUMEN

Bisphenol-A (BPA) is a lipophilic, organic, synthetic compound that has been used as an additive in polycarbonate plastics manufacturing since 1957. Studies have shown that BPA interferes with the development and functions of the brain, but little is known about the effects of BPA on human glutamatergic neurons (hGNs) at the molecular and cellular levels. We investigated the impact of chronic exposure to BPA to hGNs derived from human embryonic stem cells (hESCs). The results showed that chronic exposure of different concentrations of BPA (0, 0.1, 1.0 and 10 µM) to hGNs for 14 days reduced neurite outgrowth in a concentration-dependent manner. Using presynaptic protein synaptophysin and postsynaptic protein PSD-95 antibodies, immunofluorescence staining and western blotting results indicated that BPA exposure altered the morphology of dendritic spines and increased synaptophysin and PSD-95 expression. Furthermore, BPA exposure at concentrations higher than 1.0 µM resulted in the increase of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) expression and deterioration of dendritic spines. In addition, our results suggested that these BPA mediated neurotoxicity effects were due to an increased production of reactive nitrogen species (RNS) and reactive oxygen species (ROS) via increased nitric oxide synthase (iNOS), neuronal nitric oxide synthase (nNOS), 3-nitrotyrosine expression and Ca2+ influx. These results imply that hESC-based neuronal differentiation is an excellent cellular model to examine BPA-induced neurotoxicity on human neurons at the cellular and molecular level.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Células Madre Embrionarias Humanas/efectos de los fármacos , Fenoles/farmacología , Humanos , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad , Óxido Nítrico Sintasa de Tipo I/metabolismo , Especies Reactivas de Oxígeno/metabolismo
13.
RNA Biol ; 16(6): 785-797, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30810468

RESUMEN

Alternative polyadenylation (APA) has been discovered to play regulatory roles in the development of many cancer cells through preferential addition of a poly(A) tail at specific sites of pre-mRNA. A recent study found that APA was involved in the mediation of acute myeloid leukaemia (AML). However, unlike gene expression heterogeneity, little attention has been directed toward variations in single-cell APA for different cell types during AML development. Here, we used single-cell RNA-seq data of a massive population of 16,843 bone marrow mononuclear cells (BMMCs) from healthy and AML patient samples to investigate dynamic APA usage in different cell types. Abnormalities of APA dynamics in the BMMCs from AML patient samples were uncovered compared to the stable APA dynamics in samples from healthy individuals, as well as lower APA diversity between eight cell types in AML patients. Genes with APA dynamics specific to the AML samples were significantly enriched in cellular signal transduction pathways that contribute to AML development. Moreover, many leukaemic cell marker genes such as NF-κB, GATA2 and IAP-Family genes exhibited APA dynamics that specifically affected abnormal proliferation and differentiation of leukemic BMMCs. Additionally, mature erythroid cells displayed greater APA dynamics and global 3' UTR shortening compared with other cell types. Our results revealed extensive involvement of APA regulation in leukemia development and erythropoiesis at the single-cell level, providing a high-resolution atlas to navigate cellular mRNA processing landscapes of differentiated cells in AML.


Asunto(s)
Leucemia Mieloide Aguda/genética , Poliadenilación , Células de la Médula Ósea/metabolismo , Humanos , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual
14.
Methods Mol Biol ; 1894: 57-72, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30547455

RESUMEN

Stem cells are undifferentiated biological cells that can differentiate into all lineages under defined control condition. Stem cell neuronal differentiation can faithfully recapitulate stages of neural development and generate neuronal progenitors, mature neurons, and glial cells. Stem cell technology will largely allow for the replacement of animal studies and reduce costs, and will provide a new paradigm for in toxic genomics, bioinformatics, systems biology, and epigenetics studies. Here, we describe a nonadherent neuronal differentiation methodology developed in our laboratory, which can rapidly derive neurons and astrocytes from human embryonic stem cells (hESCs) and induced pluripotent stem cell (hiPSC) and use of this platform for nanoparticle neurotoxicity study.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Nanopartículas del Metal/toxicidad , Neurogénesis , Animales , Astrocitos/fisiología , Línea Celular , Células Madre Embrionarias Humanas , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Neuronas/fisiología , Plata/toxicidad , Pruebas de Toxicidad/métodos
15.
J Food Sci Technol ; 55(9): 3353-3361, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30150793

RESUMEN

The aims of this study were to observe the antimicrobial effect and mechanism of cinnamon oil combined with gamma radiation on Shewanella putrefaciens. Gamma radiation increased the antimicrobial activity of cinnamon oil, and the relative radiation sensitivity of gamma radiation on S. putrefaciens was increased by cinnamon oil. Gamma radiation significantly increased the changes of bacterial morphology, intra-adenosine 5'-triphosphate (intra-ATP) and extra-ATP concentrations and pHin value of S. putrefaciens treated cinnamon oil. Although, gamma radiation used alone didn't damage the bacterial morphology and ATP concentrations significantly. Gamma radiation assisted cinnamon oil to damage the cell permeability and integrity of S. putrefaciens, thus the combination of cinnamon oil and gamma radiation showed a better antimicrobial activity than used alone.

16.
Nanotoxicology ; 12(2): 104-116, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29334833

RESUMEN

Silver nanoparticles (AgNPs) are among the most extensively used nanoparticles and are found in a variety of products. This ubiquity leads to inevitable exposure to these particles in everyday life. However, the effects of AgNPs on neuron and astrocyte networks are still largely unknown. In this study, we used neurons and astrocytes derived from human embryonic stem cells as a cellular model to study the neurotoxicity that is induced by citrate-coated AgNPs (AgSCs). Immunostaining with the astrocyte and neuron markers, glial fibrillary acidic protein and microtubule-associated protein-2 (MAP2), respectively, showed that exposure to AgSCs at the concentration of 0.1 µg/mL increased the astrocyte/neuron ratio. In contrast, a higher concentration of AgSCs (5.0 µg/ml) significantly changed the morphology of astrocytes. These results suggest that astrocytes are sensitive to AgSC exposure and that low concentrations of AgSCs promote astrogenesis. Furthermore, our results showed that AgSCs reduced neurite outgrowth, decreased the expression of postsynaptic density protein 95 and synaptophysin, and induced neurodegeneration in a concentration-dependent manner. Our findings additionally suggest that the expression and phosphorylation status of MAP2 isoforms, as modulated by the activation of the Akt/glycogen synthase kinase-3/caspase-3 signaling pathway, may play an important role in AgSC-mediated neurotoxicity. We also found that AgNO3 exposure only slightly reduced neurite outgrowth and had little effect on MAP2 expression, suggesting that AgSCs and AgNO3 have different neuronal toxicity mechanisms. In addition, most of these effects were reduced when the cell culture was co-treated with AgSCs and the antioxidant ascorbic acid, which implies that oxidative stress is the major cause of AgSC-mediated astrocytic/neuronal toxicity and that antioxidants may have a neuroprotective effect.


Asunto(s)
Astrocitos/efectos de los fármacos , Células Madre Embrionarias Humanas/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Neuronas/efectos de los fármacos , Plata/toxicidad , Relación Dosis-Respuesta a Droga , Humanos , Nanopartículas del Metal/química , Transducción de Señal/efectos de los fármacos , Plata/química , Nitrato de Plata/toxicidad
17.
Environ Res ; 158: 194-202, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28647514

RESUMEN

BACKGROUND: Cigarette butts are the most common form of litter in the world, and approximately 4.5 trillion smoked cigarettes are discarded every year worldwide. Cigarette butts contain over 4000 chemicals, many of which are known to have neurotoxic effects. Stem cell neuronal differentiation provides an excellent cellular model with which to examine the impact of aqueous cigarette tar extracts (ACTEs) on neurodevelopment. METHODS: We have developed a neurosphere-based stem cell neuronal differentiation protocol that can recapitulate corticogenesis and produce cell types that are similar to upper and lower layer cortical projection neurons found in the germinal zone of the developing human cortex. In this study, ACTEs were generated from smoked cigarette butts and then applied at different concentrations to neuronal progenitors and cortical neurons derived from human embryonic stem cells. RESULTS: ACTEs reduced the expression of the cortical neuronal progenitor markers pax6, tbr2, and neuroD and decreased the number of cortical layer neurons (tbr1, satb2, foxp2, and brn2) after exposure to as low as 1.87% of the extract from one smoked cigarette butt. Furthermore, our results showed that ACTEs increased reactive oxygen species (ROS) production in cortical neurons, which caused a substantial loss of the synaptic proteins PSD95, synaptophysin, vesicular glutamate transporter1 (vGlut1), and the extracellular matrix molecule reelin; all of those molecules are important for the maintenance of cortical neuron identity and activity. CONCLUSION: ACTEs from smoked cigarettes have significant effects on cortical neuron development and neurodegeneration. The stem cell neuronal differentiation model holds great promise as a potentially powerful tool for the assessment of ACTEs on neurotoxicity.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Células Madre Embrionarias Humanas/efectos de los fármacos , Neuronas/efectos de los fármacos , Breas/toxicidad , Corteza Cerebral/fisiología , Células Madre Embrionarias Humanas/fisiología , Humanos , Neuronas/fisiología , Proteína Reelina
18.
J Stem Cell Res Ther ; 1(1): 1-3, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-33409004

RESUMEN

Stem cell-based neuronal differentiation has provided a unique opportunity for disease modeling and regenerative medicine. We have reported a novel culture condition and method for generating neuronal progenitors and neural networks from human embryonic and induced pluripotent stem cells without any genetic manipulation. Neurospheres generated under 10% CO2 with Supplemented Knockout Serum Replacement Medium (SKSRM) had doubled the expression of NESTIN, PAX6 and FOXG1 genes compared to the neurospheres generated under 5% CO2. Furthermore, an additional step (AdStep) was introduced to fragment the neurospheres, which increased the expression of neuronal progenitor genes NEUROD1, NEUROG2, TBR1, TBR2, and NOTCH1 and the formation of the neuroepithelial-type cells. With the supplements, neuronal progenitors further differentiated into different layers of cortical, pyramidal, GABAergic, glutamatergic, cholinergic, dopaminergic and purkinje neurons within 27-40 days, which is faster than traditional neurodifferentiation protocols (42-60 days). Furthermore, our in vivo studies indicated that neuronal progenitors derived under our culture conditions with "AdStep" showed significantly increased neurogenesis in Severe Combined Immunodeficiency (SCID) mouse brains. This neurosphere-based neurodifferentiation protocol is a valuable tool for studies neurogenesis, neuronal transplantation and high throughput screening assays.

19.
Neurotoxicology ; 57: 45-53, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27593553

RESUMEN

Silver nanoparticles (AgNPs) are used extensively as anti-microbial agents in various products, but little is known about their potential neurotoxic effects. In this study, we used glutamatergic neurons derived from human embryonic stem cells as a cellular model to study 20nm citrate-coated AgNPs (AgSCs) and Polyvinylpyrrolidone-coated AgNPs (AgSPs) induced neurotoxicity. AgSCs significantly damaged neurite outgrowths; increased the production of reactive oxygen species and Ca2+ influxes; reduced the expression of MAP2, PSD95, vGlut1 and NMDA receptor proteins at concentrations as low as 0.1µg/ml. In contrast, AgSPs exhibited neurotoxicity only at higher concentration. Furthermore, our results showed that AgSCs induced glutamate excitotoxicity by the activation of calmodulin and the induction of nitric oxide synthase; increased the phosphorylation of glycogen synthase kinase-3 α/ß at Tyr216 and Tau at Ser396 and reduced the expression of Tau46, which are typically observed in Alzheimer's disease. This study indicated that stem cells can provide an excellent platform for studying nanoparticle induced neurotoxicity.


Asunto(s)
Glutamatos/metabolismo , Células Madre Embrionarias Humanas/efectos de los fármacos , Nanopartículas del Metal , Neuronas/efectos de los fármacos , Plata/farmacología , Calcio , Diferenciación Celular/efectos de los fármacos , Línea Celular Transformada , Tamaño de la Célula/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , L-Lactato Deshidrogenasa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuritas/efectos de los fármacos , Neuronas/citología , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Glutamato/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Tirosina/análogos & derivados , Tirosina/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
20.
Stem Cell Res ; 15(3): 731-741, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26613348

RESUMEN

Stem cell-based neuronal differentiation has provided a unique opportunity for disease modeling and regenerative medicine. Neurospheres are the most commonly used neuroprogenitors for neuronal differentiation, but they often clump in culture, which has always represented a challenge for neurodifferentiation. In this study, we report a novel method and defined culture conditions for generating sub-type or region-specific neurons from human embryonic and induced pluripotent stem cells derived neurosphere without any genetic manipulation. Round and bright-edged neurospheres were generated in a supplemented knockout serum replacement medium (SKSRM) with 10% CO2, which doubled the expression of the NESTIN, PAX6 and FOXG1 genes compared with those cultured with 5% CO2. Furthermore, an additional step (AdSTEP) was introduced to fragment the neurospheres and facilitate the formation of a neuroepithelial-type monolayer that we termed the "neurosphederm". The large neural tube-type rosette (NTTR) structure formed from the neurosphederm, and the NTTR expressed higher levels of the PAX6, SOX2 and NESTIN genes compared with the neuroectoderm-derived neuroprogenitors. Different layers of cortical, pyramidal, GABAergic, glutamatergic, cholinergic neurons appeared within 27 days using the neurosphederm, which is a shorter period than in traditional neurodifferentiation-protocols (42-60 days). With additional supplements and timeline dopaminergic and Purkinje neurons were also generated in culture too. Furthermore, our in vivo results indicated that the fragmented neurospheres facilitated significantly better neurogenesis in severe combined immunodeficiency (SCID) mouse brains compared with the non-fragmented neurospheres. Therefore, this neurosphere-based neurodifferentiation protocol is a valuable tool for studies of neurodifferentiation, neuronal transplantation and high throughput screening assays.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Neurogénesis/inmunología , Neuronas/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Neuronas/citología , Medicina Regenerativa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...