Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Stem Cells ; 38(2): 246-260, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31648402

RESUMEN

Physical exercise-induced enhancement of learning and memory and alleviation of age-related cognitive decline in humans have been widely acknowledged. However, the mechanistic relationship between exercise and cognitive improvement remains largely unknown. In this study, we found that exercise-elicited cognitive benefits were accompanied by adaptive hippocampal proteasome activation. Voluntary wheel running increased hippocampal proteasome activity in adult and middle-aged mice, contributing to an acceleration of neurogenesis that could be reversed by intrahippocampal injection of the proteasome inhibitor MG132. We further found that increased levels of insulin-like growth factor-1 (IGF-1) in both serum and hippocampus may be essential for exercise-induced proteasome activation. Our in vitro study demonstrated that IGF-1 stimulated proteasome activity in cultured adult neural progenitor cells (NPCs) by promoting nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2), followed by elevated expressions of proteasome subunits such as PSMB5. In contrast, pretreating adult mice with the selective IGF-1R inhibitor picropodophyllin diminished exercise-induced neurogenesis, concurrent with reduced Nrf2 nuclear translocation and proteasome activity. Likewise, lowering Nrf2 expression by RNA interference with bilateral intrahippocampal injections of recombinant adeno-associated viral particles significantly suppressed exercise-induced proteasome activation and attenuated cognitive function. Collectively, our work demonstrates that proteasome activation in hippocampus through IGF-1/Nrf2 signaling is a key adaptive mechanism underlying exercise-related neurogenesis, which may serve as a potential targetable pathway in neurodegeneration.


Asunto(s)
Ejercicio Físico/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neurogénesis/genética , Animales , Humanos , Masculino , Ratones , Transfección
2.
Int J Mol Sci ; 22(3)2021 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-33573239

RESUMEN

Sevoflurane, one of the most commonly used pediatric anesthetics, was found to cause developmental neurotoxicity. To understand specific risk groups and develop countermeasures, a better understanding of its mechanisms is needed. We hypothesize that, as in many other brain degeneration pathways, long non-coding RNAs (lncRNAs) are involved in the sevoflurane-induced neurotoxicity. Postnatal day 7 (PD7) mice were exposed to 3% sevoflurane for 6 h. To quantify neurotoxicity in these mice, we (1) detected neural apoptosis through analysis of caspase 3 expression level and activity and (2) assessed long-term learning ability via the Morris water maze at PD60. To elucidate specific mechanisms, profiles of 27,427 lncRNAs and 18,855 messenger RNAs (mRNAs) in mouse hippocampi were analyzed using microarray assays. Sevoflurane-induced abnormal lncRNA and mRNA expression-associated function pathways were predicted by bioinformatic analysis. We found that sevoflurane induced significant neurotoxicity, causing acute neuroapoptosis and abnormal expression of 148 mRNAs and 301 lncRNAs on PD7 in mouse hippocampus. Additionally, exposed mice exhibited impaired memory on PD60. Bioinformatic analysis predicted that the dysregulated mRNAs, which are highly correlated with their co-expressed dysregulated lncRNAs, might be involved in 34 neurodegenerative signaling pathways (e.g., brain cell apoptosis and intellectual developmental disorder). Our study reveals for the first time that neonatal exposure to 3% sevoflurane induces abnormal lncRNA and mRNA expression profiles. These dysregulated lncRNAs/mRNAs form wide molecular networks that might contribute to various functional neurological disease pathways in the hippocampus, resulting in the observed acute apoptosis and impaired long-term memory.


Asunto(s)
Anestésicos por Inhalación/toxicidad , Síndromes de Neurotoxicidad/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo , Sevoflurano/toxicidad , Anestésicos por Inhalación/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Desarrollo Infantil/efectos de los fármacos , Biología Computacional , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Hipocampo/patología , Humanos , Lactante , Masculino , Memoria/efectos de los fármacos , Ratones , Síndromes de Neurotoxicidad/patología , Sevoflurano/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Pruebas de Toxicidad Aguda
3.
Cell Physiol Biochem ; 49(6): 2496-2510, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30261491

RESUMEN

BACKGROUND: Propofol induces acute neurotoxicity (e.g., neuroapoptosis) followed by impairment of long-term memory and learning in animals. However, underlying mechanisms remain largely unknown. Long non-coding RNAs (lncRNAs) are found to participate in various pathological processes. We hypothesized that lncRNA profile and the associated signaling pathways were altered, and these changes might be related to the neurotoxicity observed in the neonatal mouse hippocampus following propofol exposure. METHODS: In this laboratory experiment, 7-day-old mice were exposed to a subanesthetic dose of propofol for 3 hours, with 4 animals per group. Hippocampal tissues were harvested 3 hours after propofol administration. Neuroapoptosis was analyzed based on caspase 3 activity using a colorimetric assay. A microarray was performed to investigate the profiles of 35,923 lncRNAs and 24,881 messenger RNAs (mRNAs). Representative differentially expressed lncRNAs and mRNAs were validated using reverse transcription quantitative polymerase chain reaction. All mRNAs dysregulated by propofol and the 50 top-ranked, significantly dysregulated lncRNAs were subject to bioinformatics analysis for exploring the potential mechanisms and signaling network of propofol-induced neurotoxicity. RESULTS: Propofol induced neuroapoptosis in the hippocampus, with differential expression of 159 lncRNAs and 100 mRNAs (fold change ± 2.0, P< 0.05). Bioinformatics analysis demonstrated that these lncRNAs and their associated mRNAs might participate in neurodegenerative pathways (e.g., calcium handling, apoptosis, autophagy, and synaptogenesis). CONCLUSION: This novel report emphasizes that propofol alters profiles of lncRNAs, mRNAs, and their cooperative signaling network, which provides novel insights into molecular mechanisms of anesthetic-induced developmental neurodegeneration and preventive targets against the neurotoxicity.


Asunto(s)
Anestésicos/farmacología , Apoptosis/efectos de los fármacos , Hipocampo/metabolismo , Propofol/farmacología , ARN Largo no Codificante/metabolismo , Transcriptoma/efectos de los fármacos , Animales , Biología Computacional , Hipocampo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , ARN Largo no Codificante/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Anesthesiology ; 128(1): 117-129, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29040168

RESUMEN

BACKGROUND: Diabetes impairs the cardioprotective effect of volatile anesthetics, yet the mechanisms are still murky. We examined the regulatory effect of isoflurane on microRNA-21, endothelial nitric-oxide synthase, and mitochondrial respiratory complex I in type 2 diabetic mice. METHODS: Myocardial ischemia/reperfusion injury was produced in obese type 2 diabetic (db/db) and C57BL/6 control mice ex vivo in the presence or absence of isoflurane administered before ischemia. Cardiac microRNA-21 was quantified by real-time quantitative reverse transcriptional-polymerase chain reaction. The dimers and monomers of endothelial nitric-oxide synthase were measured by Western blot analysis. Mitochondrial nicotinamide adenine dinucleotide fluorescence was determined in Langendorff-perfused hearts. RESULTS: Body weight and fasting blood glucose were greater in db/db than C57BL/6 mice. Isoflurane decreased left ventricular end-diastolic pressure from 35 ± 8 mmHg in control to 23 ± 9 mmHg (P = 0.019, n = 8 mice/group, mean ± SD) and elevated ±dP/dt 2 h after post-ischemic reperfusion in C57BL/6 mice. These beneficial effects of isoflurane were lost in db/db mice. Isoflurane elevated microRNA-21 and the ratio of endothelial nitric-oxide synthase dimers/monomers and decreased mitochondrial nicotinamide adenine dinucleotide levels 5 min after ischemia in C57BL/6 but not db/db mice. MicroRNA-21 knockout blocked these favorable effects of isoflurane, whereas endothelial nitric-oxide synthase knockout had no effect on the expression of microRNA-21 but blocked the inhibitory effect of isoflurane preconditioning on nicotinamide adenine dinucleotide. CONCLUSIONS: Failure of isoflurane cardiac preconditioning in obese type 2 diabetic db/db mice is associated with aberrant regulation of microRNA-21, endothelial nitric-oxide synthase, and mitochondrial respiratory complex I.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Complejo I de Transporte de Electrón/fisiología , Precondicionamiento Isquémico Miocárdico/métodos , Isoflurano/administración & dosificación , MicroARNs/fisiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Obesidad/metabolismo , Animales , Diabetes Mellitus Tipo 2/terapia , Complejo I de Transporte de Electrón/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/deficiencia , Obesidad/terapia , Técnicas de Cultivo de Órganos , Insuficiencia del Tratamiento
5.
BMC Cardiovasc Disord ; 18(1): 197, 2018 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-30342478

RESUMEN

Long noncoding RNAs (lncRNAs) are endogenous RNA transcripts longer than 200 nucleotides which regulate epigenetically the expression of genes but do not have protein-coding potential. They are emerging as potential key regulators of diabetes mellitus and a variety of cardiovascular diseases. Diabetic cardiomyopathy (DCM) refers to diabetes mellitus-elicited structural and functional abnormalities of the myocardium, beyond that caused by ischemia or hypertension. The purpose of this review was to summarize current status of lncRNA research for DCM and discuss the challenges and possible strategies of lncRNA research for DCM. A systemic search was performed using PubMed and Google Scholar databases. Major conference proceedings of diabetes mellitus and cardiovascular disease occurring between January, 2014 to August, 2018 were also searched to identify unpublished studies that may be potentially eligible. The pathogenesis of DCM involves elevated oxidative stress, myocardial inflammation, apoptosis, and autophagy due to metabolic disturbances. Thousands of lncRNAs are aberrantly regulated in DCM. Manipulating the expression of specific lncRNAs, such as H19, metastasis-associated lung adenocarcinoma transcript 1, and myocardial infarction-associated transcript, with genetic approaches regulates potently oxidative stress, myocardial inflammation, apoptosis, and autophagy and ameliorates DCM in experimental animals. The detail data regarding the regulation and function of individual lncRNAs in DCM are limited. However, lncRNAs have been considered as potential diagnostic and therapeutic targets for DCM. Overexpression of protective lncRNAs and knockdown of detrimental lncRNAs in the heart are crucial for defining the role and function of lncRNAs of interest in DCM, however, they are technically challenging due to the length, short life, and location of lncRNAs. Gene delivery vectors can provide exogenous sources of cardioprotective lncRNAs to ameliorate DCM, and CRISPR-Cas9 genome editing technology may be used to knockdown specific lncRNAs in DCM. In summary, current data indicate that LncRNAs are a vital regulator of DCM and act as the promising diagnostic and therapeutic targets for DCM.


Asunto(s)
Cardiomiopatías Diabéticas/genética , Miocardio/metabolismo , ARN Largo no Codificante/genética , Animales , Cardiomiopatías Diabéticas/diagnóstico , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/terapia , Regulación de la Expresión Génica , Terapia Genética/métodos , Humanos , Miocardio/patología , ARN Largo no Codificante/metabolismo , ARN Largo no Codificante/uso terapéutico
6.
J Ind Microbiol Biotechnol ; 45(9): 827-838, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29936578

RESUMEN

Maltose metabolism of baker's yeast (Saccharomyces cerevisiae) in lean dough is suppressed by the glucose effect, which negatively affects dough fermentation. In this study, differences and interactions among SNF4 (encoding for the regulatory subunit of Snf1 kinase) overexpression and REG1 and REG2 (which encodes for the regulatory subunits of the type I protein phosphatase) deletions in maltose metabolism of baker's yeast were investigated using various mutants. Results revealed that SNF4 overexpression and REG1 and REG2 deletions effectively alleviated glucose repression at different levels, thereby enhancing maltose metabolism and leavening ability to varying degrees. SNF4 overexpression combined with REG1/REG2 deletions further enhanced the increases in glucose derepression and maltose metabolism. The overexpressed SNF4 with deleted REG1 and REG2 mutant ΔREG1ΔREG2 + SNF4 displayed the highest maltose metabolism and strongest leavening ability under the test conditions. Such baker's yeast strains had excellent potential applications.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Proteínas Portadoras/genética , Eliminación de Gen , Maltosa/metabolismo , Proteína Fosfatasa 1/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Factores de Transcripción/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Fermentación , Regulación Fúngica de la Expresión Génica , Glucosa/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Simportadores/genética , Simportadores/metabolismo , Factores de Transcripción/metabolismo , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo
7.
J Cell Physiol ; 232(1): 216-24, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27138089

RESUMEN

Contradictory reports on the effects of diabetes and hyperglycemia on myocardial infarction range from cytotoxicity to cytoprotection. The study was designed to investigate acute effects of high glucose-driven changes in mitochondrial metabolism and osmolarity on adaptive mechanisms and resistance to oxidative stress of isolated rat cardiomyocytes. We examined the effects of high glucose on several parameters of mitochondrial bioenergetics, including changes in oxygen consumption, mitochondrial membrane potential, and NAD(P)H fluorometry. Effects of high glucose on the endogenous cytoprotective mechanisms elicited by anesthetic preconditioning (APC) and the mediators of cell injury were also tested. These experiments included real-time measurements of reactive oxygen species (ROS) production and mitochondrial permeability transition pore (mPTP) opening in single cells by laser scanning fluorescence confocal microscopy, and cell survival assay. High glucose rapidly enhanced mitochondrial energy metabolism, observed by increase in NAD(P)H fluorescence intensity, oxygen consumption, and mitochondrial membrane potential. This substantially elevated production of ROS, accelerated opening of the mPTP, and decreased survival of cells exposed to oxidative stress. Abrogation of high glucose-induced mitochondrial hyperpolarization with 2,4 dinitrophenol (DNP) significantly, but not completely, attenuated ROS production to a level similar to hyperosmotic mannitol control. DNP treatment reversed high glucose-induced cytotoxicity to cytoprotection. Hyperosmotic mannitol treatment also induced cytoprotection. High glucose abrogated APC-induced mitochondrial depolarization, delay in mPTP opening and cytoprotection. In conclusion, high glucose-induced mitochondrial hyperpolarization abolishes APC and augments cell injury. Attenuation of high glucose-induced ROS production by eliminating mitochondrial hyperpolarization protects cardiomyocytes. J. Cell. Physiol. 232: 216-224, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Anestésicos , Glucosa/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Glucosa/metabolismo , Masculino , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar
8.
Microb Cell Fact ; 16(1): 194, 2017 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-29121937

RESUMEN

BACKGROUND: Tup1 is a general transcriptional repressor of diverse gene families coordinately controlled by glucose repression, mating type, and other mechanisms in Saccharomyces cerevisiae. Several functional domains of Tup1 have been identified, each of which has differing effects on transcriptional repression. In this study, we aim to investigate the role of Tup1 and its domains in maltose metabolism of industrial baker's yeast. To this end, a battery of in-frame truncations in the TUP1 gene coding region were performed in the industrial baker's yeasts with different genetic background, and the maltose metabolism, leavening ability, MAL gene expression levels, and growth characteristics were investigated. RESULTS: The results suggest that the TUP1 gene is essential to maltose metabolism in industrial baker's yeast. Importantly, different domains of Tup1 play different roles in glucose repression and maltose metabolism of industrial baker's yeast cells. The Ssn6 interaction, N-terminal repression and C-terminal repression domains might play roles in the regulation of MAL transcription by Tup1 for maltose metabolism of baker's yeast. The WD region lacking the first repeat could influence the regulation of maltose metabolism directly, rather than indirectly through glucose repression. CONCLUSIONS: These findings lay a foundation for the optimization of industrial baker's yeast strains for accelerated maltose metabolism and facilitate future research on glucose repression in other sugar metabolism.


Asunto(s)
Maltosa/metabolismo , Proteínas Nucleares/genética , Proteínas Represoras/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Metabolismo de los Hidratos de Carbono , Fermentación , Eliminación de Gen , Regulación Fúngica de la Expresión Génica , Proteínas Nucleares/metabolismo , Dominios y Motivos de Interacción de Proteínas/genética , Proteínas Represoras/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
9.
Exp Brain Res ; 235(9): 2627-2638, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28573310

RESUMEN

Injury-induced neuropathic pain remains a serious clinical problem. Recent studies indicate that bone marrow stromal cells (BMSCs) effectively attenuate chronic neuropathic pain in animal models. Here, we examined the therapeutic effect of intrathecal administration of BMSCs isolated from young (1-month-old) rats on pain hypersensitivity induced by tibial nerve injury. Cerebrospinal fluid (CSF) was collected and analyzed to examine the effect of BMSC administration on the expression of 67 soluble factors in CSF. A sustained remission in injury-induced mechanical hyperalgesia was observed in BMSC-treated rats but not in control animals. Engrafted BMSCs were observed in spinal cords and dorsal root ganglia at 5 weeks after cell injection. Injury significantly decreased the levels of six soluble factors in CSF: intercellular adhesion molecule 1 (ICAM-1), interleukin-1ß (IL-1ß), IL-10, hepatocyte growth factor (HGF), Nope protein, and neurogenic locus notch homolog protein 1 (Notch-1). Intrathecal BMSCs significantly attenuated the injury-induced reduction of ICAM-1, IL-1ß, HGF, IL-10, and Nope. This study adds to evidence supporting the use of intrathecal BMSCs in pain control and shows that this effect is accompanied by the reversal of injury-induced reduction of multiple CSF soluble factors. Our findings suggest that these soluble factors may be potential targets for treating chronic pain.


Asunto(s)
Citocinas/líquido cefalorraquídeo , Ganglios Espinales , Hiperalgesia/líquido cefalorraquídeo , Hiperalgesia/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Neuralgia/líquido cefalorraquídeo , Neuralgia/terapia , Médula Espinal , Animales , Modelos Animales de Enfermedad , Ganglios Espinales/citología , Masculino , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología
10.
Anesth Analg ; 125(1): 241-254, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28622174

RESUMEN

BACKGROUND: Growing animal evidence demonstrates that prolonged exposure to propofol during brain development induces widespread neuronal cell death, but there is little information on the role of astrocytes. Astrocytes can release neurotrophic growth factors such as brain-derived neurotrophic factor (BDNF), which can exert the protective effect on neurons in paracrine fashion. We hypothesize that during propofol anesthesia, BDNF released from developing astrocytes may not be sufficient to prevent propofol-induced neurotoxicity. METHODS: Hippocampal astrocytes and neurons isolated from neonatal Sprague Dawley rats were exposed to propofol at a clinically relevant dose of 30 µM or dimethyl sulfoxide as control for 6 hours. Propofol-induced cell death was determined by propidium iodide (PI) staining in astrocyte-alone cultures, neuron-alone cultures, or cocultures containing either low or high density of astrocytes (1:9 or 1:1 ratio of astrocytes to neurons ratio [ANR], respectively). The astrocyte-conditioned medium was collected 12 hours after propofol exposure and measured by protein array assay. BDNF concentration in astrocyte-conditioned medium was quantified using enzyme-linked immunosorbent assay. Neuron-alone cultures were treated with BDNF, tyrosine receptor kinase B inhibitor cyclotraxin-B, glycogen synthase kinase 3ß (GSK3ß) inhibitor CHIR99021, or mitochondrial fission inhibitor Mdivi-1 before propofol exposure. Western blot was performed for quantification of the level of protein kinase B and GSK3ß. Mitochondrial shape was visualized through translocase of the outer membrane 20 staining. RESULTS: Propofol increased cell death in neurons by 1.8-fold (% of PI-positive cells [PI%] = 18.6; 95% confidence interval [CI], 15.2-21.9, P < .05) but did not influence astrocyte viability. The neuronal death was attenuated by a high ANR (1:1 cocultures; fold change [FC] = 1.17, 95% CI, 0.96-1.38, P < .05), but not with a low ANR [1:9 cocultures; FC = 1.87, 95% CI, 1.48-2.26, P > .05]). Astrocytes secreted BDNF in a cell density-dependent way and propofol decreased BDNF secretion from astrocytes. Administration of BDNF, CHIR99021, or Mdivi-1 significantly attenuated the propofol-induced neuronal death and aberrant mitochondria in neuron-alone cultures (FC = 0.8, 95% CI, 0.62-0.98; FC = 1.22, 95% CI, 1.11-1.32; FC = 1.35, 95% CI, 1.16-1.54, respectively, P < .05) and the cocultures with a low ANR (1:9; FC = 0.85, 95% CI, 0.74-0.97; FC = 1.08, 95% CI, 0.84-1.32; FC = 1.25, 95% CI, 1.1-1.39, respectively, P < .05). Blocking BDNF receptor or protein kinase B activity abolished astrocyte-induced neuroprotection in the cocultures with a high ANR (1:1). CONCLUSIONS: Astrocytes attenuate propofol-induced neurotoxicity through BDNF-mediated cell survival pathway suggesting multiple neuroprotective strategies such as administration of BDNF, astrocyte-conditioned medium, decreasing mitochondrial fission, or inhibition of GSK3ß.


Asunto(s)
Anestésicos Intravenosos/toxicidad , Astrocitos/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipocampo/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Dinámicas Mitocondriales/efectos de los fármacos , Neuronas/efectos de los fármacos , Comunicación Paracrina/efectos de los fármacos , Propofol/toxicidad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/enzimología , Astrocitos/patología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados/metabolismo , Relación Dosis-Respuesta a Droga , Hipocampo/enzimología , Hipocampo/patología , Mitocondrias/enzimología , Mitocondrias/patología , Neuronas/enzimología , Neuronas/patología , Proteínas Tirosina Quinasas/metabolismo , Ratas Sprague-Dawley , Receptor trkB , Transducción de Señal/efectos de los fármacos
11.
Nature ; 464(7291): 1058-61, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20348907

RESUMEN

Cancer chemoprevention uses natural, synthetic, or biological substances to reverse, suppress, or prevent either the initial phase of carcinogenesis or the progression of neoplastic cells to cancer. It holds promise for overcoming problems associated with the treatment of late-stage cancers. However, the broad application of chemoprevention is compromised at present by limited effectiveness and potential toxicity. To overcome these challenges, here we developed a new chemoprevention approach that specifically targets premalignant tumour cells for apoptosis. We show that a deficiency in the adenomatous polyposis coli (APC) gene and subsequent activation of beta-catenin lead to the repression of cellular caspase-8 inhibitor c-FLIP (also known as CFLAR) expression through activation of c-Myc, and that all-trans-retinyl acetate (RAc) independently upregulates tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptors and suppresses decoy receptors. Thus, the combination of TRAIL and RAc induces apoptosis in APC-deficient premalignant cells without affecting normal cells in vitro. In addition, we show that short-term and non-continuous TRAIL and RAc treatment induce apoptosis specifically in intestinal polyps, strongly inhibit tumour growth, and prolong survival in multiple intestinal neoplasms C57BL/6J-Apc(Min)/J (Apc(Min)) mice. With our approach, we further demonstrate that TRAIL and RAc induce significant cell death in human colon polyps, providing a potentially selective approach for colorectal cancer chemoprevention by targeting APC-deficient cells for apoptosis.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Apoptosis/efectos de los fármacos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Vitamina A/análogos & derivados , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Diterpenos , Regulación de la Expresión Génica/efectos de los fármacos , Genes APC , Humanos , Pólipos Intestinales/tratamiento farmacológico , Pólipos Intestinales/patología , Ratones , Ratones Endogámicos C57BL , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ésteres de Retinilo , Transducción de Señal/efectos de los fármacos , Tasa de Supervivencia , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Factores de Tiempo , Vitamina A/administración & dosificación , Vitamina A/farmacología , Vitamina A/uso terapéutico , beta Catenina/metabolismo
12.
Anesth Analg ; 123(5): 1286-1296, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27551735

RESUMEN

Mounting evidence has demonstrated that general anesthetics could induce developmental neurotoxicity, including acute widespread neuronal cell death, followed by long-term memory and learning abnormalities. Propofol is a commonly used intravenous anesthetic agent for the induction and maintenance of anesthesia and procedural and critical care sedation in children. Compared with other anesthetic drugs, little information is available on its potential contributions to neurotoxicity. Growing evidence from multiple experimental models showed a similar neurotoxic effect of propofol as observed in other anesthetic drugs, raising serious concerns regarding pediatric propofol anesthesia. The aim of this review is to summarize the current findings of propofol-induced developmental neurotoxicity. We first present the evidence of neurotoxicity from animal models, animal cell culture, and human stem cell-derived neuron culture studies. We then discuss the mechanism of propofol-induced developmental neurotoxicity, such as increased cell death in neurons and oligodendrocytes, dysregulation of neurogenesis, abnormal dendritic development, and decreases in neurotrophic factor expression. Recent findings of complex mechanisms of propofol action, including alterations in microRNAs and mitochondrial fission, are discussed as well. An understanding of the toxic effect of propofol and the underlying mechanisms may help to develop effective novel protective or therapeutic strategies for avoiding the neurotoxicity in the developing human brain.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Hipnóticos y Sedantes/toxicidad , Fármacos Neuroprotectores/uso terapéutico , Propofol/toxicidad , Animales , Encéfalo/patología , Células Cultivadas , Humanos , Fármacos Neuroprotectores/farmacología , Síndromes de Neurotoxicidad/patología , Síndromes de Neurotoxicidad/prevención & control
13.
Anesth Analg ; 122(5): 1269-79, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26991754

RESUMEN

BACKGROUND: Hyperglycemia can blunt the cardioprotective effects of isoflurane in the setting of ischemia-reperfusion injury. Previous studies suggest that reactive oxygen species (ROS) and increased mitochondrial fission play a role in cardiomyocyte death during ischemia-reperfusion injury. To investigate the role of glucose concentration in ROS production and mitochondrial fission during ischemia-reperfusion (with and without anesthetic protection), we used the novel platform of human-induced pluripotent stem-cell (iPSC)-derived cardiomyocytes (CMs). METHODS: Cardiomyocyte differentiation from iPSC was characterized by the expression of CM-specific markers using immunohistochemistry and by measuring contractility. iPSC-CMs were exposed to varying glucose conditions (5, 11, and 25 mM) for 24 hours. Mitochondrial permeability transition pore opening, cell viability, and ROS generation endpoints were used to assess the effects of various treatment conditions. Mitochondrial fission was monitored by the visualization of fragmented mitochondria using confocal microscopy. Expression of activated dynamin-related protein 1, a key protein responsible for mitochondrial fission, was assessed by Western blot. RESULTS: Cardiomyocytes were successfully differentiated from iPSC. Elevated glucose conditions (11 and 25 mM) significantly increased ROS generation, whereas only the 25-mM high glucose condition induced mitochondrial fission and increased the expression of activated dynamin-related protein 1 in iPSC-CMs. Isoflurane delayed mitochondrial permeability transition pore opening and protected iPSC-CMs from oxidative stress in 5- and 11-mM glucose conditions to a similar level as previously observed in various isolated animal cardiomyocytes. Scavenging ROS with Trolox or inhibiting mitochondrial fission with mdivi-1 restored the anesthetic cardioprotective effects in iPSC-CMs in 25-mM glucose conditions. CONCLUSIONS: Human iPSC-CM is a useful, relevant model for studying isoflurane cardioprotection and can be manipulated to recapitulate complex clinical perturbations. We demonstrate that the cardioprotective effects of isoflurane in elevated glucose conditions can be restored by scavenging ROS or inhibiting mitochondrial fission. These findings may contribute to further understanding and guidance for restoring pharmacological cardioprotection in hyperglycemic patients.


Asunto(s)
Anestésicos por Inhalación/farmacología , Glucosa/toxicidad , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Isoflurano/farmacología , Mitocondrias Cardíacas/enzimología , Dinámicas Mitocondriales/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Antioxidantes/farmacología , Biomarcadores/metabolismo , Diferenciación Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citoprotección , Relación Dosis-Respuesta a Droga , Dinaminas , GTP Fosfohidrolasas/metabolismo , Humanos , Hiperglucemia/metabolismo , Hiperglucemia/patología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Proteínas Mitocondriales/metabolismo , Contracción Miocárdica/efectos de los fármacos , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos
14.
Appl Microbiol Biotechnol ; 100(14): 6375-6383, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27041690

RESUMEN

Leavening ability in sweet dough is required for the commercial applications of baker's yeast. This property depends on many factors, such as glycolytic activity, sucrase activity, and osmotolerance. This study explored the importance of sucrase level on the leavening ability of baker's yeast in sweet dough. Furthermore, the baker's yeast strains with varying sucrase activities were constructed by deleting SUC2, which encodes sucrase or replacing the SUC2 promoter with the VPS8/TEF1 promoter. The results verify that the sucrase activity negatively affects the leavening ability of baker's yeast strains under high-sucrose conditions. Based on a certain level of osmotolerance, sucrase level plays a significant role in the fermentation performance of baker's yeast, and appropriate sucrase activity is an important determinant for the leavening property of baker's yeast in sweet dough. Therefore, modification on sucrase activity is an effective method for improving the leavening properties of baker's yeast in sweet dough. This finding provides guidance for the breeding of industrial baker's yeast strains for sweet dough leavening. The transformants BS1 with deleted SUC2 genetic background provided decreased sucrase activity (a decrease of 39.3 %) and exhibited enhanced leavening property (an increase of 12.4 %). Such a strain could be useful for industrial applications.


Asunto(s)
Pan/microbiología , Manipulación de Alimentos , Microbiología de Alimentos , Saccharomyces cerevisiae/metabolismo , Sacarasa/metabolismo , Biomasa , Pan/análisis , Medios de Cultivo/química , Fermentación , Eliminación de Gen , Glucosa/análisis , Glicerol/análisis , Factor 1 de Elongación Peptídica/genética , Factor 1 de Elongación Peptídica/metabolismo , Plásmidos/genética , Regiones Promotoras Genéticas , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Sacarosa/análisis , Trehalosa/análisis , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , beta-Fructofuranosidasa/genética , beta-Fructofuranosidasa/metabolismo
15.
Cell Mol Life Sci ; 72(10): 2005-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25504289

RESUMEN

The regulation of cardiac differentiation is critical for maintaining normal cardiac development and function. The precise mechanisms whereby cardiac differentiation is regulated remain uncertain. Here, we have identified a GATA-4 target, EGF, which is essential for cardiogenesis and regulates cardiac differentiation in a dose- and time-dependent manner. Moreover, EGF demonstrates functional interaction with GATA-4 in inducing the cardiac differentiation of P19CL6 cells in a time- and dose-dependent manner. Biochemically, GATA-4 forms a complex with STAT3 to bind to the EGF promoter in response to EGF stimulation and cooperatively activate the EGF promoter. Functionally, the cooperation during EGF activation results in the subsequent activation of cyclin D1 expression, which partly accounts for the lack of additional induction of cardiac differentiation by the GATA-4/STAT3 complex. Thus, we propose a model in which the regulatory cascade of cardiac differentiation involves GATA-4, EGF, and cyclin D1.


Asunto(s)
Diferenciación Celular/fisiología , Factor de Crecimiento Epidérmico/metabolismo , Factor de Transcripción GATA4/metabolismo , Corazón/embriología , Modelos Biológicos , Miocardio/citología , Transducción de Señal/fisiología , Animales , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Técnicas Histológicas , Inmunoprecipitación , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Tiempo
16.
Mol Pain ; 11: 5, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25888914

RESUMEN

BACKGROUND: Cell-based therapy may hold promise for treatment of chronic pain. Mesenchymal stem cells (MSCs) are readily available and robust, and their secretion of therapeutic peptides can be enhanced by genetically engineering. We explored the analgesic potential of transplanting bone marrow-derived MSCs that have been transduced with lentivectors. To optimize efficacy and safety, primary sensory neurons were targeted by MSC injection into the dorsal root ganglia (DRGs). RESULTS: MSCs were transduced using lentivectors to express enhanced green fluorescent protein (EGFP) or to co-express the analgesic peptide glial cell line-derived neurotrophic factor (GDNF) and EGFP by a viral 2A bicistronic transgene cassette. Engineered MSCs were injected into the 4(th) lumbar (L4) and L5 DRGs of adult allogeneic rats to evaluate survival in the DRGs. MSCs were detected by immunofluorescence staining up to 2-3 weeks after injection, distributed in the extracellular matrix space without disrupting satellite glial cell apposition to sensory neurons, suggesting well-tolerated integration of engrafted MSCs into DRG tissue. To examine their potential for inhibiting development of neuropathic pain, MSCs were injected into the L4 and L5 DRGs ipsilateral to a spinal nerve ligation injury. Animals injected with GDNF-engineered MSCs showed moderate but significant reduction in mechanical allodynia and hyperalgesia compared to controls implanted with MSCs expressing EGFP alone. We also observed diminished long-term survival of allografted MSCs at 3 weeks, and the development of a highly-proliferating population of MSCs in 12% of DRGs after transplantation. CONCLUSIONS: These data indicate that genetically modified MSCs secreting analgesic peptides could potentially be developed as a novel DRG-targeted cell therapy for treating neuropathic pain. However, further work is needed to address the challenges of MSC survival and excess proliferation, possibly with trials of autologous MSCs, evaluation of clonally selected populations of MSCs, and investigation of regulation of MSC proliferation.


Asunto(s)
Analgesia , Ganglios Espinales/trasplante , Células Madre Mesenquimatosas/citología , Neuralgia/terapia , Neuronas Aferentes/citología , Analgesia/métodos , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Ganglios Espinales/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas , Neuralgia/genética , Neuralgia/metabolismo , Manejo del Dolor/métodos , Ratas Sprague-Dawley , Nervios Espinales/metabolismo
17.
Anesthesiology ; 123(5): 1067-83, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26352374

RESUMEN

BACKGROUND: Studies in developing animals have shown that anesthetic agents can lead to neuronal cell death and learning disabilities when administered early in life. Development of human embryonic stem cell-derived neurons has provided a valuable tool for understanding the effects of anesthetics on developing human neurons. Unbalanced mitochondrial fusion and fission lead to various pathological conditions including neurodegeneration. The aim of this study was to dissect the role of mitochondrial dynamics in propofol-induced neurotoxicity. METHODS: Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate in situ nick-end labeling staining was used to assess cell death in human embryonic stem cell-derived neurons. Mitochondrial fission was assessed using TOM20 staining and electron microscopy. Expression of mitochondrial fission-related proteins was assessed by Western blot, and confocal microscopy was used to assess opening time of the mitochondrial permeability transition pore (mPTP). RESULTS: Exposure to 6 h of 20 µg/ml propofol increased cell death from 3.18 ± 0.17% in the control-treated group to 9.6 ± 0.95% and led to detrimental increases in mitochondrial fission (n = 5 coverslips per group) accompanied by increased expression of activated dynamin-related protein 1 and cyclin-dependent kinase 1, key proteins responsible for mitochondrial fission. Propofol exposure also induced earlier opening of the mPTP from 118.9 ± 3.1 s in the control-treated group to 73.3 ± 1.6 s. Pretreatment of the cells with mdivi-1, a mitochondrial fission blocker rescued the propofol-induced toxicity, mitochondrial fission, and mPTP opening time (n = 75 cells per group). Inhibiting cyclin-dependent kinase 1 attenuated the increase in cell death and fission and the increase in expression of activated dynamin-related protein 1. CONCLUSION: These data demonstrate for the first time that propofol-induced neurotoxicity occurs through a mitochondrial fission/mPTP-mediated pathway.


Asunto(s)
Anestésicos Intravenosos/toxicidad , Células Madre Embrionarias/efectos de los fármacos , Dinámicas Mitocondriales/efectos de los fármacos , Neuronas/efectos de los fármacos , Propofol/toxicidad , Muerte Celular/efectos de los fármacos , Células Cultivadas , Células Madre Embrionarias/patología , Células Madre Embrionarias/ultraestructura , Humanos , Neuronas/patología , Neuronas/ultraestructura
18.
Anesthesiology ; 122(4): 795-805, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25536091

RESUMEN

BACKGROUND: Anesthetic cardioprotection reduces myocardial infarct size after ischemia-reperfusion injury. Currently, the role of microRNA in this process remains unknown. MicroRNAs are short, noncoding nucleotide sequences that negatively regulate gene expression through degradation or suppression of messenger RNA. In this study, the authors uncovered the functional role of microRNA-21 (miR-21) up-regulation after anesthetic exposure. METHODS: MicroRNA and messenger RNA expression changes were analyzed by quantitative real-time polymerase chain reaction in cardiomyocytes after exposure to isoflurane. Lactate dehydrogenase release assay and propidium iodide staining were conducted after inhibition of miR-21. miR-21 target expression was analyzed by Western blot. The functional role of miR-21 was confirmed in vivo in both wild-type and miR-21 knockout mice. RESULTS: Isoflurane induces an acute up-regulation of miR-21 in both in vivo and in vitro rat models (n = 6, 247.8 ± 27.5% and 258.5 ± 9.0%), which mediates protection to cardiomyocytes through down-regulation of programmed cell death protein 4 messenger RNA (n = 3, 82.0 ± 4.9% of control group). This protective effect was confirmed by knockdown of miR-21 and programmed cell death protein 4 in vitro. In addition, the protective effect of isoflurane was abolished in miR-21 knockout mice in vivo, with no significant decrease in infarct size compared with nonexposed controls (n = 8, 62.3 ± 4.6% and 56.2 ± 3.2%). CONCLUSIONS: The authors demonstrate for the first time that isoflurane mediates protection of cardiomyocytes against oxidative stress via an miR-21/programmed cell death protein 4 pathway. These results reveal a novel mechanism by which the damage done by ischemia/reperfusion injury may be decreased.


Asunto(s)
Anestésicos por Inhalación/farmacología , Cardiotónicos/farmacología , Isoflurano/farmacología , MicroARNs/biosíntesis , Miocitos Cardíacos/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Embarazo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Regulación hacia Arriba/fisiología
19.
J Ind Microbiol Biotechnol ; 42(6): 939-48, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25877163

RESUMEN

Dough-leavening ability is one of the main aspects considered when selecting a baker's yeast strain for baking industry. Generally, modification of maltose metabolic pathway and known regulatory networks of maltose metabolism were used to increase maltose metabolism to improve leavening ability in lean dough. In this study, we focus on the effects of PGM2 (encoding for the phosphoglucomutase) and SNR84 (encoding for the H/ACA snoRNA) that are not directly related to both the maltose metabolic pathway and known regulatory networks of maltose metabolism on the leavening ability of baker's yeast in lean dough. The results show that the modifications on PGM2 and/or SNR84 are effective ways in improving leavening ability of baker's yeast in lean dough. Deletion of PGM2 decreased cellular glucose-1-phosphate and overexpression of SNR84 increased the maltose permease activity. These changes resulted in 11, 19 and 21% increases of the leavening ability for PGM2 deletion, SNR84 overexpression and SNR84 overexpression combining deleted PGM2, respectively.


Asunto(s)
Pan/microbiología , Metabolismo de los Hidratos de Carbono/genética , Maltosa/metabolismo , Fosfoglucomutasa/deficiencia , ARN Nucleolar Pequeño/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Industria de Alimentos/métodos , Eliminación de Gen , Regulación Fúngica de la Expresión Génica , Genes Fúngicos/genética , Glucofosfatos/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Fosfoglucomutasa/genética , Fosfoglucomutasa/metabolismo , Saccharomyces cerevisiae/enzimología
20.
Physiol Genomics ; 46(21): 789-97, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25159851

RESUMEN

MicroRNAs (miRNAs or miRs) are endogenous, small RNA molecules that suppress expression of targeted mRNA. miR-21, one of the most extensively studied miRNAs, is importantly involved in divergent pathophysiological processes relating to ischemia/reperfusion (I/R) injury, such as inflammation and angiogenesis. The role of miR-21 in renal I/R is complex, with both protective and pathological pathways being regulated by miR-21. Preconditioning-induced upregulation of miR-21 contributes to the protection against subsequent renal I/R injury through the targeting of genes such as the proapoptotic gene programmed cell death 4 and interactions between miR-21 and hypoxia-inducible factor. Conversely, long-term elevation of miR-21 may be detrimental to the organ by promoting the development of renal interstitial fibrosis following I/R injury. miR-21 is importantly involved in several pathophysiological processes related to I/R injury including inflammation and angiogenesis as well as the biology of stem cells that could be used to treat I/R injury; however, the effect of miR-21 on these processes in renal I/R injury remains to be studied.


Asunto(s)
Isquemia/genética , MicroARNs/fisiología , Daño por Reperfusión/genética , Animales , Supervivencia Celular/genética , Fibrosis/genética , Regulación de la Expresión Génica , Humanos , Hipoxia/genética , Inflamación/genética , Precondicionamiento Isquémico , Riñón/irrigación sanguínea , MicroARNs/genética , Neovascularización Fisiológica/genética , Trasplante de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA