Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Total Environ ; 926: 171873, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38521275

RESUMEN

Research on High Spatial-Resolved Source-Specific Exposure and Risk (HSRSSER) was conducted based on multiple-year, multiple-site synchronous measurement of PM2.5-bound (particulate matter with aerodynamic diameter<2.5 µm) toxic components in a Chinese megacity. The developed HSRSSER model combined the Positive Matrix Factorization (PMF) and Land Use Regression (LUR) to predict high spatial-resolved source contributions, and estimated the source-specific exposure and risk by personal activity time- and population-weighting. A total of 287 PM2.5 samples were collected at ten sites in 2018-2020, and toxic species including heavy metals (HMs), polycyclic aromatic hydrocarbons (PAHs) and organophosphate esters (OPEs) were analyzed. The percentage non-cancer risk were in the order of traffic emission (48 %) > industrial emission (22 %) > coal combustion (12 %) > waste incineration (11 %) > resuspend dust (7 %) > OPE-related products (0 %) ≈ secondary particles (0 %). Similar orders were observed in cancer risk. For traffic emission, due to its higher source contributions and large population in central area, non-cancer and cancer risk fraction increased from 23 % to 48 % and 20 % to 46 % after exposure estimation; while for industrial emission, higher source contributions but small population in suburb area decreased the percentage non-cancer and cancer risk from 38 % to 22 % and 39 % to 24 %, respectively.


Asunto(s)
Contaminantes Atmosféricos , Hidrocarburos Policíclicos Aromáticos , Contaminantes Atmosféricos/análisis , Emisiones de Vehículos/análisis , Monitoreo del Ambiente , Material Particulado/análisis , Ciudades , Hidrocarburos Policíclicos Aromáticos/análisis , China/epidemiología
2.
Cell Biochem Funct ; 42(2): e3968, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38439590

RESUMEN

Over the past decade, the prevalence of diabetes has increased significantly worldwide, leading to an increase in vascular complications of diabetes (VCD), such as diabetic cardiomyopathy (DCM), diabetic nephropathy (DN), and diabetic retinopathy (DR). Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs), long Noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), play a key role in cellular processes, including the pathophysiology of diabetes and VCD via pyroptosis. ncRNAs (e.g., miR-17, lnc-MEG3, and lnc-KCNQ1OT1) can regulate pyroptosis in pancreatic ß cells. Some ncRNAs are involved in VCD progression. For example, miR-21, lnc-KCNQ1OT1, lnc-GAS5, and lnc-MALAT1 were reported in DN and DCM, and lnc-MIAT was identified in DCM and DR. Herein, this review aimed to summarize recent research findings related to ncRNAs-mediated pyroptosis at the onset and progression of diabetes and VCD.


Asunto(s)
Diabetes Mellitus , Cardiomiopatías Diabéticas , Nefropatías Diabéticas , MicroARNs , Humanos , Piroptosis , Cardiomiopatías Diabéticas/genética , Nefropatías Diabéticas/genética , ARN no Traducido/genética , MicroARNs/genética , Diabetes Mellitus/genética
3.
Environ Res ; 246: 118110, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38184066

RESUMEN

The aim of this survey was to evaluate the residue levels, distribution and exposure risk of the 38 most commonly used pesticides in rapeseed samples collected from the main production areas in China over a two-year period. The sampling area covered 12 provinces, including Guizhou, Shaanxi, Yunnan, Hunan, Jiangxi, Sichuan, Chongqing, Anhui, Henan, Hubei, Zhejiang, and Jiangsu provinces. The pesticide residues were determined using a QuEChERS (Quick Easy Cheap Effective Rugged and Safe) method coupled with gas chromatography-tandem mass spectrometry and liquid chromatography-tandem mass spectrometry. 8.4% of the rapeseed samples contained pesticides with a residue level ranging from 0.001 to 0.634 mg/kg. The detected analytes were imidacloprid, quizalofop-P-ethyl, thiamethoxam, paclobutrazol, prochloraz, tebuconazole, difenoconazole, s-metolachlor, carbofuran, and carbendazim. The concentrations of four analytes, including thiamethoxam, difenoconazole, carbendazim and prochloraz, exceeded the maximum residue level set by the Chinese government for rapeseed, with exceedance rates of 0.1%, 0.1%, 0.1%, and 1.1%, respectively. Based on the index of quality for residues (IqR) values, 91.6% of the total rapeseed samples had an IqR category of Excellent (IqR = 0). Only 1.5% of the tested samples were of inadequate quality. Furthermore, the assessment of chronic and acute exposure, as well as health risks associated with pesticide residues in rapeseed, was conducted for different age groups within the Chinese population, including adults (6-14 years), children (15-49 years), and the elderly (50-74 years). The results of this assessment indicated that pesticide residues in rapeseed cultivated in China are not expected to be of short- or long-term risks to the Chinese customers.


Asunto(s)
Bencimidazoles , Brassica napus , Carbamatos , Residuos de Plaguicidas , Plaguicidas , Niño , Humanos , Anciano , Adolescente , Residuos de Plaguicidas/análisis , Tiametoxam/análisis , China/epidemiología , Plaguicidas/análisis , Medición de Riesgo , Contaminación de Alimentos/análisis
5.
Nano Lett ; 23(23): 11120-11128, 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38032110

RESUMEN

An oncolytic virus is a promising strategy for glioblastoma (GBM) therapy. However, there are still some challenges such as the blood-brain barrier (BBB) and preexisting immunity for targeted treatment of GBM with an oncolytic virus. In this study, two kinds of cell membrane-coated oncolytic adenoviruses (NCM-Ad and GCM-Ad) were prepared using neural stem cells (NSCs) and GBM cells as sources of membranes, respectively, and were shown to improve the targeted infectivity on GBM cells and avoid the immune clearance of preexisting neutralizing antibodies in vitro and in vivo. Specifically, NCM-Ad showed a strong ability to cross the BBB and target tumor cells in vivo. To improve the cytotoxicity to GBM, a capsid dual-modified oncolytic adenovirus (A4/k37) was also encapsulated, and NCM-A4/k37 showed outstanding tumor targeting and inhibition capacity in an orthotopic xenograft tumor model of GBM upon intravenous administration. This study provides a promising oncolytic virus-based targeted therapeutic strategy for glioma.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Glioblastoma/terapia , Glioblastoma/patología , Adenoviridae/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Virus Oncolíticos/genética , Membrana Celular/metabolismo
6.
Genome Biol ; 24(1): 187, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37582787

RESUMEN

BACKGROUND: The international Dog10K project aims to sequence and analyze several thousand canine genomes. Incorporating 20 × data from 1987 individuals, including 1611 dogs (321 breeds), 309 village dogs, 63 wolves, and four coyotes, we identify genomic variation across the canid family, setting the stage for detailed studies of domestication, behavior, morphology, disease susceptibility, and genome architecture and function. RESULTS: We report the analysis of > 48 M single-nucleotide, indel, and structural variants spanning the autosomes, X chromosome, and mitochondria. We discover more than 75% of variation for 239 sampled breeds. Allele sharing analysis indicates that 94.9% of breeds form monophyletic clusters and 25 major clades. German Shepherd Dogs and related breeds show the highest allele sharing with independent breeds from multiple clades. On average, each breed dog differs from the UU_Cfam_GSD_1.0 reference at 26,960 deletions and 14,034 insertions greater than 50 bp, with wolves having 14% more variants. Discovered variants include retrogene insertions from 926 parent genes. To aid functional prioritization, single-nucleotide variants were annotated with SnpEff and Zoonomia phyloP constraint scores. Constrained positions were negatively correlated with allele frequency. Finally, the utility of the Dog10K data as an imputation reference panel is assessed, generating high-confidence calls across varied genotyping platform densities including for breeds not included in the Dog10K collection. CONCLUSIONS: We have developed a dense dataset of 1987 sequenced canids that reveals patterns of allele sharing, identifies likely functional variants, informs breed structure, and enables accurate imputation. Dog10K data are publicly available.


Asunto(s)
Lobos , Perros , Animales , Lobos/genética , Mapeo Cromosómico , Alelos , Polimorfismo de Nucleótido Simple , Nucleótidos , Demografía
7.
ACS Nano ; 17(16): 15424-15440, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37552584

RESUMEN

Infection diseases such as AIDS and COVID-19 remain challenging in regard to protective vaccine design, while adjuvants are critical for subunit vaccines to induce strong, broad, and durable immune responses against variable pathogens. Here, we demonstrate that periodic mesoporous organosilica (PMO) acts as a multifunctional nanoadjuvant by adsorbing recombinant protein antigens. It can effectively deliver antigens to lymph nodes (LNs), prolong antigen exposure, and rapidly elicit germinal center (GC) responses by directly activating naive B cells via the C-type lectin receptor signaling pathway. In mice, both the gp120 trimer (HIV-1 antigen) and the receptor-binding domain (SARS-CoV-2 antigen) with the PMO nanoadjuvant elicit potent and durable antibodies that neutralize heterologous virus strains. LN immune cells analysis shows that PMO helps to effectively activate the T-follicular helper cells, GC B cells, and memory B cells and eventually develop broad and durable humoral responses. Moreover, the PMO nanoadjuvant elicits a strong cellular immune response and shapes this immune response by eliciting high levels of effector T helper cell cytokines. This study identifies a promising nanoadjuvant for subunit vaccines against multiple pathogens.


Asunto(s)
COVID-19 , Animales , Ratones , SARS-CoV-2 , Centro Germinal , Linfocitos B , Antígenos , Vacunas de Subunidad
8.
J Exp Clin Cancer Res ; 42(1): 216, 2023 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-37605148

RESUMEN

BACKGROUND: In addition to specifically inducing tumor cell apoptosis, recombinant tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has also been reported to influence the cancer immune microenvironment; however, its underlying effects and mechanisms remain unclear. Investigating the immunomodulatory effects and mechanisms of recombinant TRAIL in the tumor microenvironment (TME) may provide an important perspective and facilitate the exploration of novel TRAIL strategies for tumor therapy. METHODS: Immunocompetent mice with different tumors were treated with three doses of recombinant TRAIL, and then the tumors were collected for immunological detection and mechanistic investigation. Methodological approaches include flow cytometry analysis and single-cell sequencing. RESULTS: In an immunocompetent mouse model, recombinant soluble mouse TRAIL (smTRAIL) had dose-related immunomodulatory effects. The optimal dose of smTRAIL (2 mg/kg) activated innate immune cells and CD8+ T cells, whereas higher doses of smTRAIL (8 mg/kg) promoted the formation of a tumor-promoting immune microenvironment to counteract the apoptotic effects on tumor cells. The higher doses of smTRAIL treatment promoted M2-like macrophage recruitment and polarization and increased the production of protumor inflammatory cytokines, such as IL-10, which deepened the suppression of natural killer (NK) cells and CD8+ T cells in the tumor microenvironment. By constructing an HU-HSC-NPG.GM3 humanized immune system mouse model, we further verified the immunomodulatory effects induced by recombinant soluble human TRAIL (shTRAIL) and found that combinational administration of shTRAIL and trabectedin, a macrophage-targeting drug, could remodel the tumor immune microenvironment, further enhance antitumor immunity, and strikingly improve antitumor effects. CONCLUSION: Our results highlight the immunomodulatory role of recombinant TRAIL and suggest promising therapeutic strategies for clinical application.


Asunto(s)
Linfocitos T CD8-positivos , Microambiente Tumoral , Humanos , Animales , Ratones , Apoptosis , Citocinas , Modelos Animales de Enfermedad
9.
Sensors (Basel) ; 23(4)2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36850771

RESUMEN

This paper defines a smart home use case to automatically adjust home temperature and/or hot water. The main objective is to reduce the energy consumption of cooling, heating and hot water systems in smart homes. To this end, the residents set a temperature (i.e., X degree Celsius) for home and/or hot water. When the residents leave homes (e.g., for work), they turn off the cooling or heating devices. A few minutes before arriving at their residences, the cooling or heating devices start working automatically to adjust the home or water temperature according to the residents' preference (i.e., X degree Celsius). This can help reduce the energy consumption of these devices. To estimate the arrival time of the residents (i.e., drivers), this paper uses a machine learning-based street traffic prediction system. Unlike many related works that use machine learning for tracking and predicting residents' behaviors inside their homes, this paper focuses on predicting resident behavior outside their home (i.e., arrival time as a context) to reduce the energy consumption of smart homes. One main objective of this paper is to find the most appropriate machine learning and neural network-based (MLNN) algorithm that can be integrated into the street traffic prediction system. To evaluate the performance of several MLNN algorithms, we utilize an Uber's dataset for the city of San Francisco and complete the missing values by applying an imputation algorithm. The prediction system can also be used as a route recommender to offer the quickest route for drivers.

10.
Protein Pept Lett ; 29(12): 1072-1081, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36098412

RESUMEN

BACKGROUND: Accumulating evidence has demonstrated the immunomodulatory effects of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in rheumatoid arthritis and the tumor microenvironment, besides its known capacity of specifically inducing the apoptosis of cancer cells. Mice are common available animal models for studying the roles of TRAIL. However, mice express only a single TRAIL receptor (mTRAILR) with an intracellular death domain, in contrast to the two TRAIL receptors (TRAILR1 and TRAILR2) in humans. Moreover, human TRAIL binds weakly to mTRAILR, whereas mouse TRAIL has a high affinity for human TRAIL-Rs. Therefore, we considered that murine TRAIL would be more suitable than human TRAIL for exploring the immunoregulatory effect of TRAIL in immunocompetent mice or when using mouse cells as the target. To our knowledge, the detailed method for the production of recombinant murine TRAIL has not been reported. OBJECTIVE: In this study, we aimed to design and express two soluble forms of murine TRAIL and verify the properties of the protein. METHODS: Recombinant murine TRAILs were expressed in Escherichia coli BL21 (DE3, and Nichelating affinity chromatography was used for protein purification. SDS-PAGE, GDS-PAGE and HPLC were applied to analyze the protein structure. The cytotoxicity of our purified murine TRAILs was evaluated in the TRAIL-sensitive human breast cancer ZR-75-30 cells and murine breast cancer 4T1 cells. Finally, validation of the tumor-killing ability of the murine protein in vivo. RESULTS: Two soluble forms of murine TRAILs (mT_N99 and mT_N188) were purified and demonstrated with high purity and trimeric structure. In addition, Zn2+ supplement was essential to produce soluble murine TRAILs in E.coli BL21 (DE3). The two purified soluble mTRAILs showed similar cytotoxicity to cancer cells, moreover, mT_N99 also showed a good anti-tumor effect in vivo and is more suitable for the treatment of murine tumor models. CONCLUSION: A production approach for recombinant murine TRAIL was determined, which covered the design of shortened forms, expression, purification and characterization.


Asunto(s)
Ligando Inductor de Apoptosis Relacionado con TNF , Animales , Femenino , Humanos , Ratones , Apoptosis , Línea Celular Tumoral , Suplementos Dietéticos , Escherichia coli/genética , Escherichia coli/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Microambiente Tumoral , Zinc/farmacología
11.
Med Oncol ; 39(5): 70, 2022 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-35568793

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand is a potential therapeutic anti-cancer drug with selective cytotoxicity in cancer cells. However, in multiple clinical trials, the therapeutic effect of TRAIL is limited owing to tumor resistance. The combination of small molecules or other drugs may represent a suitable strategy to overcome TRAIL resistance. This study found that 20(s)-ginsenoside Rh2 sensitized non-sensitive human hepatocellular carcinoma cells to TRAIL-induced apoptosis. The combination of TRAIL and Rh2 decreased cell viability and increased caspase cascade-induced apoptosis in several liver cancer cell lines. Moreover, we found that Rh2 reduced the apoptosis-related protein XIAP and Survivin, a negative regulator of the apoptosis pathway. At the same time, Rh2 can further enhance TRAIL-induced apoptosis by upregulating the death receptor 5, thereby significantly enhancing its anti-tumor effect. Furthermore, Rh2 enhanced the therapeutic efficacy of TRAIL in mouse xenograft models, suggesting that Rh2 also sensitizes TRAIL in vivo. Taken together, our study indicates that Rh2 may act as a sensitizer in combination with TRAIL to increase the efficacy of its anti-tumor activity.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Apoptosis , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Ginsenósidos , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Ratones , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Regulación hacia Arriba
12.
Acta Biomater ; 143: 392-405, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35259519

RESUMEN

Traditional anticancer treatments directly target tumor cells. In contrast, cancer immunotherapy fortifies host immunity. Nanoparticles that incorporate both immunomodulatory and chemotherapeutic agents regulate the tumor microenvironment by activating immune cells and enhancing antitumor immunity. Nanoparticle-based cancer immunotherapy has received considerable attention and has been extensively studied in recent years. In this study, we developed a targeted drug delivery system to enhance immunotherapeutic efficacy and overcome drug resistance by inducing tumor apoptosis and immunogenic cell death (ICD), and activating immune cells. Periodic mesoporous organosilica nanoparticles (PMOs) bore tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on their surfaces, and their inner cores were loaded with doxorubicin (DOX). TRAIL enhanced the nanoparticle-targeting capacity and worked synergistically with DOX against breast cancer cells in vitro and in vivo. Furthermore, we revealed for the first time the ability of PMOs to activate dendritic cells (DCs) and elevate ICD levels of DOX in vitro, and TRAIL further enhances the immunomodulatory function of PMOs. Systemic exposure to DOX@PMO-hT induced an immune response, activated DCs and CD4+ and CD8+ T cells, and significantly suppressed tumor growth in a 4T1-bearing immunocompetent mouse model. Overall, our study demonstrates that TRAIL-modified, DOX-embedded PMO nanoparticles represent a good candidate for tumor-targeted immunotherapy, which has relatively superior therapeutic efficacy and highly promising future application prospects. STATEMENT OF SIGNIFICANCE: This study revealed for the first time the ability of PMOs to elevate ICD levels and activate DCs in vitro. The results explained the immunomodulatory function of PMOs and demonstrated the synergistic effects of TRAIL and DOX in triple-negative breast cancer. In addition, immunomodulatory effects of the drug delivery vectors constructed in this study were verified in vivo.


Asunto(s)
Nanopartículas , Neoplasias de la Mama Triple Negativas , Animales , Linfocitos T CD8-positivos , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Sistemas de Liberación de Medicamentos , Humanos , Inmunoterapia , Ratones , Ratones Endogámicos BALB C , Nanopartículas/uso terapéutico , Microambiente Tumoral
13.
Cancer Lett ; 535: 215661, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35325845

RESUMEN

The immune status of the tumor microenvironment is a key indicator determining the antitumor effect of immunotherapy. Oncolytic viruses directly target tumor cells or indirectly modulate the tumor microenvironment (TME) especially when properly armed. It was previously demonstrated that conditionally replicating adenovirus serotype 5 (CRAd5) encoding tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) had outstanding antitumor effects in different human cancer cells xenograft models; however, its antitumor immune mechanism has not been evaluated in immunocompetent preclinical mouse models. We first explored the antitumor activity of CRAd5-TRAIL in several murine tumor models and found that the expression of TRAIL induced increases or activation in tumor-infiltrating T cells. To further improve the antitumor effects, mouse CD40 ligand (mCD40L) as an immune activator expressed by recombinant Ad5 vector was firstly used in combination with CRAd5-TRAIL for tumor immunotherapy. Both in vitro and in vivo studies demonstrated that mCD40L effectively activated dendritic cells (DCs), B cells, and tumor-infiltrating T cells, and also promoted tumor cell apoptosis by increasing the expression of TRAIL receptors, thereby significantly enhancing the antitumor activity of oncolytic adenoviruses in CT26 and B16 tumor-bearing models. Although affected by the restriction of oncolytic adenovirus replication in mouse cells, the combination treatment failed to completely eliminate tumor cells, our research still provided a promising strategy for oncolytic adenovirus-mediated solid tumor immunotherapy.


Asunto(s)
Adenoviridae , Ligando de CD40 , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Ligando de CD40/genética , Ligando de CD40/metabolismo , Humanos , Inmunoterapia , Ratones , Ratones Desnudos , Microambiente Tumoral , Replicación Viral
14.
J Cancer ; 11(19): 5713-5726, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32913465

RESUMEN

Glioblastoma, the most common human brain tumor, is highly invasive and difficult to cure using conventional cancer therapies. As an alternative, adenovirus-mediated virotherapies represent a popular and maturing technology. However, the cell surface coxsackievirus and adenovirus receptor (CAR)-dependent infection mechanism limits the infectivity and oncolytic effects of Adenovirus type 5. To address this limitation, in this study we aimed to develop a novel oncolytic adenovirus for enhanced infectivity and therapeutic efficacy toward glioblastoma. We developed a novel genetically modified oncolytic adenovirus vector with dual capsid modifications to facilitate infection and specific cytotoxicity toward glioma cells. Modification of the adenoviral capsid proteins involved the incorporation of a synthetic leucine zipper-like dimerization domain into the capsid protein IX (pIX) of human adenovirus serotype 5 (Ad5) and the exchange of the fiber knob from Ad37. The virus infection mechanism and anti-tumor efficacy of modified vectors were evaluated in both in vitro (cell) and in vivo (mouse) models. Ad37-knob exchange efficiently promoted the virus infection and replication-induced glioma cell lysis by oncolytic Ad5. We also found that gene therapy mediated by the dual-modified oncolytic Ad5 vector coupled with the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exhibited significantly enhanced anti-tumor efficacy in vitro and in vivo. This genetically modified oncolytic adenovirus provides a promising vector for future use in glioblastoma gene-viral-based therapies.

15.
PLoS One ; 14(9): e0222340, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31509580

RESUMEN

Growth hormone (GH) is an important hormone released by the pituitary gland that plays a key role in the growth and development of organisms. In our study, TargetScan analysis and the dual luciferase reporter assays were used to predict and screen for miRNAs that might act on the rat Gh1 gene, and we identified miR-543-5p. Then, the GH3 cell line and the primary rat pituitary cells were transfected with miRNA mimic, inhibitor, and siRNA. We detected the Gh1 gene expression and the GH secretion by real-time PCR and ELISAs, respectively, to verify the regulatory effect of miR-543-5p on GH secretion. The results showed that miR-543-5p can inhibit Gh1 mRNA expression and reduce GH secretion. MiR-543-5p inhibitor upregulated Gh1 mRNA expression and increased GH secretion compared with the negative control. In summary, miR-543-5p downregulates Gh1 expression, resulting in a decrease in GH synthesis and secretion, which demonstrates the important role of miRNAs in regulating GH and animal growth and development.


Asunto(s)
Hormona del Crecimiento/genética , MicroARNs/genética , Hormonas Adenohipofisarias/genética , Regiones no Traducidas 3'/genética , Animales , Línea Celular , Expresión Génica , Regulación de la Expresión Génica/genética , Hormona del Crecimiento/metabolismo , Masculino , Hipófisis/metabolismo , Adenohipófisis/metabolismo , Hormonas Adenohipofisarias/metabolismo , Cultivo Primario de Células , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Transfección
16.
PLoS One ; 12(6): e0179476, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28622392

RESUMEN

Parkinson's disease (PD) is a progressive and age-associated neurodegenerative disorder. Patients at different stages of the disease course have distinguished features, mainly in the number of dopaminergic neurons. Cerebral dopamine neurotrophic factor (CDNF) is a recently discovered neurotrophic factor, being deemed as a hopeful candidate for PD treatment. Here, we evaluated the efficacy of CDNF in protecting dopaminergic function using the 6-OHDA-induced PD rat model suffering from different levels of neuronal loss and the recombinant adeno-associated virus 8 (AAV8) as a carrier for the CDNF gene. The results showed that AAV8-CDNF administration significantly improved the motor function and increased the tyrosine hydroxylase (TH) levels in PD rats with mild lesions (2 weeks post lesion), but it had limited therapeutic effects in rats with severe lesions (5 weeks post lesion). To better improve the recovery of motor function in severely lesioned PD rats, we employed a strategy using the CDNF gene along with the aromatic amino acid decarboxylase (AADC) gene. This combination therapeutic strategy indeed showed an enhanced benefit in restoring the motor function of severely lesioned PD rats by providing the neuroprotective effect of CDNF and dopamine enhancing effect of AADC as expected. This study may provide a basis for future clinical application of CDNF in PD patients at different stages and offer a new alternative strategy of joint use of CDNF and AADC for advanced PD patients in clinical trials.


Asunto(s)
Cuerpo Estriado , Dependovirus , Terapia Genética/métodos , Actividad Motora , Factores de Crecimiento Nervioso , Oxidopamina/efectos adversos , Enfermedad de Parkinson Secundaria , Recuperación de la Función , Transducción Genética/métodos , Animales , Descarboxilasas de Aminoácido-L-Aromático/biosíntesis , Descarboxilasas de Aminoácido-L-Aromático/genética , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiopatología , Humanos , Masculino , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Oxidopamina/farmacología , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/genética , Enfermedad de Parkinson Secundaria/metabolismo , Enfermedad de Parkinson Secundaria/terapia , Ratas , Ratas Wistar
17.
Int J Med Sci ; 14(4): 340-347, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28553166

RESUMEN

Methamphetamine (METH) exerts significant neurotoxicity in experimental animals and humans when taken at high doses or abused chronically. Long-term abusers have decreased dopamine levels, and they are more likely to develop Parkinson's disease (PD). To date, few medications are available to treat the METH-induced damage of neurons. Glial cell line-derived neurotrophic factor (GDNF) has been previously shown to reduce the dopamine-depleting effects of neurotoxic doses of METH. However, the effect of cerebral dopamine neurotrophic factor (CDNF), which has been reported to be more specific and efficient than GDNF in protecting dopaminergic neurons against 6-OHDA toxicity, in attenuating METH neurotoxicity has not been determined. Thus, the present study aimed to evaluate the neuroprotective effect of CDNF against METH-induced damage to the dopaminergic system in vitro and in vivo. In vitro, CDNF protein increased the survival rate and reduced the tyrosine hydroxylase (TH) loss of METH-treated PC12 cells. In vivo, METH was administered to rats following human CDNF overexpression mediated by the recombinant adeno-associated virus. Results demonstrated that CDNF overexpression in the brain could attenuate the METH-induced dopamine and TH loss in the striatum but could not lower METH-induced hyperthermia.


Asunto(s)
Técnicas de Transferencia de Gen , Metanfetamina/toxicidad , Factores de Crecimiento Nervioso/genética , Síndromes de Neurotoxicidad/terapia , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Dopamina/metabolismo , Fiebre/inducido químicamente , Fiebre/tratamiento farmacológico , Fiebre/genética , Terapia Genética , Humanos , Factores de Crecimiento Nervioso/uso terapéutico , Neuronas/efectos de los fármacos , Neuronas/patología , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/patología , Células PC12 , Ratas , Tirosina 3-Monooxigenasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...