Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Free Radic Biol Med ; 162: 478-489, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33189867

RESUMEN

The immunoinhibitory effect of glucocorticoid and immunoenhancing attributes of melatonin (MEL) are well known, however, the involvement of glucocorticoid receptor (GR) in melatonin modulation of bacterial toxins caused-inflammation has not been studied in colon. Pyocyanin (PCN), a toxin released by Pseudomonas aeruginosa, can destroy cells through generating superoxide products and inflammatory response. Here we report that PCN treatment elevated the generation of reactive oxygen species (ROS), which further lead to mitochondrial swelling and caspase cascades activation both in vivo and in vitro. However, MEL treatment alleviated the oxidative stress caused by PCN on cells through scavenging ROS and restoring the expression of antioxidant enzyme so that to effectively alleviate the apoptosis. Large amounts of ROS can activate the NLRP3 signaling pathway, so MEL inhibited PCN induced NLRP3 inflammasome activation and inflammatory cytokines (IL-1ß, IL-8, and TNF-α) secretion. In order to further investigate the molecular mechanism, goblet cells were exposed to MEL and PCN in the presence of luzindole and RU486, inhibitors of MEL receptors and GR respectively. It was found that PCN significantly inhibited the expression level of GR, and MEL effectively alleviated the inhibition phenomenon. Moreover, we found that MEL mainly upregulated the expression of GR to achieve its anti-inflammatory and anti-apoptotic functions rather than through its own receptor (MT2) in colon goblet cells. Therefore, MEL can reverse the inhibitory effects of PCN on GR/p-GR expression to present its anti-oxidative and anti-apoptotic function.


Asunto(s)
Toxinas Bacterianas , Melatonina , Animales , Apoptosis , Colon , Humanos , Inflamasomas , Melatonina/farmacología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Piocianina , Especies Reactivas de Oxígeno , Receptores de Glucocorticoides/genética
2.
Front Vet Sci ; 7: 315, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32671106

RESUMEN

On modern farms, animals are at high risk of bacterial invasion due to environmental stress factors. The adrenal gland is the terminal organ of the stress response. The crosstalk between adrenal endocrine stress and innate immune response is critical for the maintenance of immune homeostasis during inflammation. Thus, it's important to explore whether stresses play a pivotal role in lipopolysaccharide (LPS)-induced inflammatory response in the porcine adrenal gland. Thirty-days-old Duroc × Landrace × Large White crossbred piglets (12 ± 0.5 kg) were randomly allocated into four groups in a 2 × 2 factorial arrangement of treatments, including ACTH pretreatment (with or without ACTH injection) and LPS challenge (with or without LPS injection). Each group consisted of six male piglets. The results showed that our LPS preparation alone induced mRNA expressions of IL-1ß, IL-6, TNF-α, IL-10, COX-2, TLR2, TLR4, and GR (P < 0.05). ACTH pretreatment downregulated the TLR2 mRNA and IL-6 protein level induced by our LPS preparation significantly (P < 0.05) by one-way ANOVA analysis. Treatment with LPS alone extremely significantly decreased ssc-miR-338 levels (P < 0.01). Interaction of ACTH × LPS was significant for cNOS level (P = 0.011) and ssc-miR-338 expression (P = 0.04) by two-way ANOVA analysis. The LPS treatment significantly downregulated cNOS levels (P < 0.01), which was significantly attenuated by ACTH pretreatment (P < 0.05). Lipopolysaccharide alone did not affect ssc-miR-146b expression levels compared to that in the vehicle group. However, ACTH pretreatment in combination with LPS significantly increased this micro-RNA expression (P < 0.05). TLRs 1-10 were all expressed in adrenal tissue. The LPS challenge alone induced remarkable compensatory mitochondrial damages at the ultrastructural level, which was alleviated by ACTH pretreatment. Accordingly, ACTH pretreatment was able to block LPS-induced secretion of local adrenal cortisol (P < 0.05). Taken together, our results demonstrate that ACTH pretreatment seems to attenuate LPS-induced mitochondria damage and inflammation that decreased cNOS activity in the adrenal gland and ultimately returned local adrenal cortisol to basal levels at 6 h post LPS injection.

4.
Poult Sci ; 99(6): 3111-3120, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32475448

RESUMEN

Maternal betaine was reported to regulate offspring hepatic cholesterol metabolism in mammals. However, it is unclear whether and how feeding betaine to laying hens affects hepatic cholesterol metabolism in offspring chickens. Rugao yellow-feathered laying hens (n = 120) were fed basal or 0.5% betaine-supplemented diet for 28 D before the eggs were collected for incubation. Maternal betaine significantly decreased the hepatic cholesterol content (P < 0.05) in offspring chickens. Accordingly, the cholesterol biosynthetic enzymes, sterol regulator element-binding protein 2 (SREBP2) and 3-hydroxy-3-methylglutaryl coenzyme A reductase, were decreased, while cholesterol-7alpha-hydroxylase (CYP7A1), which converts cholesterol to bile acids, was increased at both mRNA and protein levels in betaine-treated offspring chickens. Hepatic mRNA and protein expression of low-density lipoprotein receptor was significantly (P < 0.05) increased, while the mRNA abundance of cholesterol acyltransferase 1 (ACAT1) that mediates cholesterol esterification was significantly (P < 0.05) decreased in the betaine group. Meanwhile, hepatic protein contents of DNA methyltransferases 1 and betaine homocysteine methyltransferase were increased (P < 0.05), which was associated with modifications of CpG methylation on affected cholesterol metabolic genes. Furthermore, the level of CpG methylation on gene promoters was increased (P < 0.05) for sterol regulator element-binding protein 2 and abundance of cholesterol acyltransferase 1 yet decreased (P < 0.05) for cholesterol-7alpha-hydroxylase. These results indicate that maternal betaine supplementation significantly decreases hepatic cholesterol deposition through epigenetic regulation of cholesterol metabolic genes in offspring juvenile chickens.


Asunto(s)
Proteínas Aviares/genética , Betaína/metabolismo , Pollos/metabolismo , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol/metabolismo , Metilación de ADN , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Alimentación Animal/análisis , Animales , Proteínas Aviares/metabolismo , Betaína/administración & dosificación , Pollos/genética , Colesterol 7-alfa-Hidroxilasa/metabolismo , Metilación de ADN/efectos de los fármacos , Dieta/veterinaria , Suplementos Dietéticos/análisis , Epigénesis Genética , Hígado/metabolismo , Masculino , Herencia Materna , Regiones Promotoras Genéticas/efectos de los fármacos , Distribución Aleatoria , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo
5.
Poult Sci ; 99(5): 2533-2542, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32359589

RESUMEN

In documents, maternal betaine modulates hypothalamic cholesterol metabolism in chicken posthatchings, but it remains unclear whether this effect can be passed on by generations. In present study, eggs were injected with saline or betaine at 2.5 mg/egg, and the hatchlings (F1) were raised under the same condition until sexual maturation. Both the control group and the betaine group used artificial insemination to collect sperm from their cockerels. Fertilized eggs were incubated, and the hatchlings of the following generation (F2) were raised up to 64 D of age. F2 cockerels in betaine group showed significantly (P < 0.05) lower body weight, which was associated with significantly decreased (P < 0.05) hypothalamic content of total cholesterol and cholesterol ester. Concordantly, hypothalamic expression of cholesterol biosynthetic genes, SREBP2 and HMGCR, were significantly downregulated (P < 0.05), together with cholesterol conversion-related and excretion-related genes, CYP46A1 and ABCA1. These changes coincided with a significant downregulation in mRNA expression of regulatory neuropeptides including brain-derived neurotrophic factor, neuropeptide Y, and corticotropin-releasing hormone. Moreover, genes involved in methyl transfer cycle were also modified. Betaine homocysteine methyltransferase (P < 0.05) was downregulated, yet DNA methyltransferase1 tended to be upregulated (P = 0.06). S-adenosyl methionine/S-adenosylhomocysteine ratio was higher in the hypothalamus of betaine-treated F2 cockerels, which was associated with significantly modified CpG methylation on the promoter of those affected genes. These results suggested that betaine might regulate central cholesterol metabolism and hypothalamic expression of genes related to brain function by altering promoter DNA methylation in F2 cockerels.


Asunto(s)
Proteínas Aviares/genética , Betaína/administración & dosificación , Embrión de Pollo/efectos de los fármacos , Colesterol/genética , Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Animales , Proteínas Aviares/metabolismo , Pollos , Colesterol/metabolismo , Metilación de ADN , Masculino , Regiones Promotoras Genéticas/genética
6.
Cell Stress Chaperones ; 25(1): 47-56, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31745845

RESUMEN

Our previous study had shown that chronic corticosterone (CORT) exposure causes excessive fat deposition in chicken liver, yet it remains unknown whether it is associated with inflammation and fibrosis. In general, heat shock proteins (HSPs) are activated in response to acute stress to play a cytoprotective role, and this activation is associated with m6A-mediated post-transcriptional regulation. However, changes of HSPs and the m6A methylation on their mRNAs in response to chronic CORT treatment in chicken liver have not been reported. In this study, chronic CORT exposure induced inflammation and fibrosis in chicken liver, associated with significantly modulated expression of HSPs that was significantly upregulated at mRNA level yet downregulated at protein level. Concurrently, m6A methyltransferases METTL3 content was upregulated together with the level of m6A methylation on HSPs transcripts. The m6A-seq analysis revealed 2-6 significantly (P < 0.05) hypermethylated m6A peaks in the mRNA of 4 different species of HSPs in CORT-treated chicken liver. HSP90B1 transcript had 6 differentially methylated m6A peaks among which peaks on exon 16 and exon 17 showed 3.14- and 4.72-fold of increase, respectively. Mutation of the 8 predicted m6A sites on exon 16 and exon 17 resulted in a significant (P < 0.05) increase in eGFP-fused content of HSP90B1 exon 16 and exon 17 fragment in 293 T cells, indicating a possible role of m6A in post-transcriptional regulation of HSPs. In conclusion, chronic CORT exposure induces inflammation and fibrosis in chicken liver along with an increase in the levels and m6A methylation of several HSPs mRNAs; HSPs levels were however reduced under the indicated conditions. Results presented suggest that the reduction in HSPs levels may be associated with m6A methylation in CORT-exposed chickens.


Asunto(s)
Corticosterona/farmacología , Fibrosis/inducido químicamente , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Choque Térmico/efectos de los fármacos , Inflamación/inducido químicamente , Animales , Pollos , Metilación de ADN/efectos de los fármacos , Fibrosis/tratamiento farmacológico , Proteínas de Choque Térmico/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/genética , Cirrosis Hepática/tratamiento farmacológico , Metiltransferasas/genética
7.
Aging (Albany NY) ; 11(6): 1832-1849, 2019 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-30923260

RESUMEN

Autophagy plays an important role in cell survival under diverse stress conditions. Here, we show that white LED light exposure for 24 h significantly activated autophagy-related genes and increased autophagosome formation in hippocampal neural cells (HT-22). Concurrently, the rhythmic pattern of clock-related gene expression was disrupted, which was associated with augmented expression of SIRT1, AMPK and retinoid-related orphan receptor alpha (RORα). SR1001, a specific inhibitor of RORα, protected the cells from light-induced activation of autophagy. Moreover, light exposure increased glucocorticoid receptor (GR) phosphorylation and nuclear translocation. GR inhibitor RU486 prevented light-induced up-regulation of RORα and the activation of autophagy. These changes were associated with enhanced glycogen synthase kinase-3 (GSK-3) activity and its specific inhibitor CHIR-99021 significantly rescued light-induced autophagy and augmented GR, RORα and autophagy-related proteins. Furthermore, GSK-3 was identified as an upstream regulator of GR/RORα signaling as it was not affected by GR or RORα inhibitors. Taken together, our data demonstrate that GSK-3-mediated GR/RORα signaling pathway is involved in white LED light-induced autophagy in hippocampal neuron cells.


Asunto(s)
Autofagia/genética , Supervivencia Celular , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/metabolismo , Luminiscencia , Animales , Autofagia/fisiología , Humanos , Ratones , Neuronas/metabolismo , Fosforilación , Transducción de Señal
8.
J Cell Physiol ; 234(4): 3621-3633, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30471106

RESUMEN

N-(3-oxododecanoyl)-l-homoserine lactone (3-oxo-C12-HSL), a quorum-sensing (QS) molecule produced by Gram-negative bacteria in the gastrointestinal tract, adversly impacts host cells. Our previous study demonstrated that 3-oxo-C12-HSL induced a decrease in cell viability via cell apoptosis and eventually disrupted mucin synthesis from LS174T goblet cells. However, the molecular mechanism underlying cell apoptosis and whether pyroptosis was involved in this process are still unknown. In this study, we emphasized on the caspases signal pathway and sterile inflammation to reveal the harmful effects of 3-oxo-C12-HSL on LS174T goblet cells. Our data showed that 3-oxo-C12-HSL is a major inducer of oxidative stress indicated by a high level of intracellular reactive oxygen species (ROS). However, TQ416, an inhibitor of paraoxonase 2, can effectively block oxidative stress. A higher ROS level is the trigger for activating the caspase-1 and 3 cascade signal pathways. Blockade of ROS synthesis and caspase-1 and 3 cascades can obviously rescue the viability of LS174T cells after 3-oxo-C12-HSL treatment. We also found that paralleled with a higher level of ROS and caspases activation, an abnormal expression of proinflammatory cytokines was induced by 3-oxo-C12-HSL treatment; however, the blockage of TLRs-NF-κB pathway cannot restore cell viability and secretary function. These data collectively indicate that 3-oxo-C12-HSL exposure induces damages to cell viability and secretary function of LS174T goblet cells, which is mediated by oxidative stress, cell apoptosis, and sterile inflammation. Overall, the data in this study will provide a better understanding of the harmful impacts of some QS molecules on host cells and their underlying mechanism.


Asunto(s)
4-Butirolactona/análogos & derivados , Caspasa 1/metabolismo , Células Caliciformes/efectos de los fármacos , Homoserina/análogos & derivados , Piroptosis/efectos de los fármacos , Percepción de Quorum , 4-Butirolactona/toxicidad , Arildialquilfosfatasa/metabolismo , Caspasa 3/metabolismo , Línea Celular , Activación Enzimática , Células Caliciformes/metabolismo , Células Caliciformes/patología , Homoserina/toxicidad , Humanos , Mediadores de Inflamación/metabolismo , Mucinas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
9.
J Agric Food Chem ; 66(29): 7633-7642, 2018 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-29961332

RESUMEN

Peroxisome proliferator-activated receptor α (PPARα) plays a protective role against non-alcoholic fatty liver disease (NAFLD). Sodium butyrate (NaB) has been shown to alleviate NAFLD, yet whether and how PPARα is involved in the action of NaB remains elusive. In this study, NaB administration alleviated high-fat-diet-induced NAFLD in adult rats, with a decrease of hepatic triglyceride content from 108.18 ± 5.77 to 81.34 ± 7.94 µg/mg ( p < 0.05), which was associated with a significant activation of PPARα. Nuclear factor κ-light-chain-enhancer of activated B cell (NF-κB)-mediated nucleotide-binding domain-like receptor protein 3 signaling and pro-inflammatory cytokine release were diminished by NaB treatment. NaB-induced PPARα upregulation coincided with a reduced protein content of histone deacetylase 1 and promoted histone H3 acetyl K9 (H3K9Ac) modification on the promoter of PPARα, whereas NaB-induced suppression of inflammation was linked to significantly increased PPARα binding with p-p65. NaB acts as a histone deacetylase inhibitor to upregulate PPARα expression with enhanced H3K9Ac modification on it promoter. NaB-induced PPARα activation stimulates fatty acid ß oxidation and inhibits NF-κB-mediated inflammation pathways via protein-protein interaction, thus contributing to amelioration of high-fat-diet-induced NAFLD in adult rats.


Asunto(s)
Ácido Butírico/administración & dosificación , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , PPAR alfa/metabolismo , Animales , Citocinas/genética , Citocinas/metabolismo , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos/metabolismo , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histonas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Hígado/metabolismo , Masculino , FN-kappa B/genética , FN-kappa B/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Oxidación-Reducción , PPAR alfa/genética , Ratas , Ratas Sprague-Dawley , Triglicéridos/metabolismo
10.
J Nutr ; 148(3): 316-325, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29546310

RESUMEN

Background: We have shown previously that in ovo betaine injection can prevent nonalcoholic fatty liver induced by glucocorticoid exposure in chickens; yet it remains unknown whether feeding betaine to laying hens may exert similar effects in their progeny. Objective: In this study, we fed laying hens a betaine-supplemented diet, and the progeny were later exposed chronically to corticosterone (CORT) to test hepatoprotective effects and further elucidate underlying mechanisms. Methods: Rugao yellow-feathered laying hens (n = 120) were fed a basal (control, C) diet or a 0.5% betaine-supplemented (B) diet for 28 d before their eggs were collected for incubation. At 49 d of age, male chickens selected from each group were daily injected subcutaneously with solvent (15% ethanol; vehicle, VEH) or CORT (4.0 mg/kg body mass) for 7 d to establish a fatty liver model. Chickens in the 4 groups (C-VEH, C-CORT, B-VEH, and B-CORT) were killed at day 57. Plasma and hepatic triglyceride (TG) concentrations, as well as the hepatic expression of genes involved in lipogenesis and lipophagy, were determined. Results: CORT induced a 1.6-fold increase in the plasma TG concentration (P < 0.05) and a 1.8-fold increment in the hepatic TG concentration (P < 0.05), associated with activation of lipogenic genes (70-780%). In contrast, lipophagy and mitochondrial ß-oxidation genes were inhibited by 30-60% (P < 0.05) in CORT-treated chickens. These CORT-induced changes were completely normalized by maternal betaine supplementation or were partially normalized to intermediate values that were significantly different from those in the C-VEH and C-CORT groups. These effects were accompanied by modifications in CpG methylation and glucocorticoid receptor binding to the promoters of major lipogenic and lipophagic genes (P < 0.05). Conclusions: These results indicate that maternal betaine supplementation protects male juvenile chickens from CORT-induced TG accumulation in the liver via epigenetic modulation of lipogenic and lipophagic genes.


Asunto(s)
Betaína/uso terapéutico , Corticosterona/efectos adversos , Suplementos Dietéticos , Hígado Graso/prevención & control , Hígado/efectos de los fármacos , Fenómenos Fisiologicos de la Nutrición Prenatal , Triglicéridos/metabolismo , Animales , Betaína/farmacología , Pollos , Corticosterona/metabolismo , Metilación de ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Epigénesis Genética , Hígado Graso/etiología , Hígado Graso/metabolismo , Femenino , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Hígado/metabolismo , Masculino , Mitocondrias , Proteínas Mitocondriales/genética , Embarazo , Regiones Promotoras Genéticas , Receptores de Glucocorticoides/metabolismo
11.
Artículo en Inglés | MEDLINE | ID: mdl-29452261

RESUMEN

As a feed additive, betaine is widely used in livestock production for its ability to promote growth. Our previous studies had reported that maternal betaine supplementation altered hepatic metabolism in offspring. But it remains unknown whether and how maternal betaine modulates metabolism of thyroid hormones in the offspring chickens by epigenetic modification. In this study, one hundred and twenty Rugao yellow-feathered laying hens were randomly divided into two groups, and were fed basal diet with or without 0.5% betaine supplementation for 28 days. After that, all the hens were artificially inseminated and then four hundreds fertilized eggs were selected. After hatching, the newborn chicks were raised until 56 days old. Betaine fed female chicks showed significantly lower body weight and lower level of biologically active thyroid hormone in plasma compared to control group, which was associated with significantly decrease in expression of type 1 iodothyronine deiodinase (Dio1). Moreover, betaine also changed hepatic expression of betaine-homocysteine -S-methyltransferase (BHMT) and DNA methyltransferase 1 (DNMT1), which may contribute to hypermethylation of the Dio1 promoter. Interestingly, betaine treatments of hens caused none of these effects in male chicks except Dio1 expression. These results indicate that maternal betaine administration effects growth of offspring through differential modification of Dio1 gene methylation and expression in liver and this model of transgenerational effects may help elucidate the mechanisms of maternal effects arise in natural systems.


Asunto(s)
Betaína/farmacología , Pollos , Epigénesis Genética/efectos de los fármacos , Yoduro Peroxidasa/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Madres , Animales , Betaína/administración & dosificación , Metilación de ADN/efectos de los fármacos , Femenino , Masculino , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tiroxina/sangre , Triyodotironina/sangre
12.
Artículo en Inglés | MEDLINE | ID: mdl-29247843

RESUMEN

Betaine is widely used in animal nutrition to promote growth, development and methyl donor during methionine metabolism through nutritional reprogramming via regulation of gene expression. Prenatal betaine exposure is reported to modulate hypothalamic cholesterol metabolism in chickens, yet it remains unknown whether feeding hens with betaine-supplemented diet may affect hypothalamic cholesterol metabolism in F1 offspring. In this study, hens were fed with basal or betaine-supplemented (0.5%) for 30days, and the eggs were collected for incubation. The hatchlings were raised under the same condition up to 56days of age. Betaine-treated group showed significantly (P<0.05) higher plasma concentration of total cholesterol and HDL-cholesterol, together with increased hypothalamic content of total cholesterol and cholesterol ester. Concordantly, hypothalamic gene expression of SREBP2, HMGCR, and LDLR was significantly up regulated (P<0.05). Also, mRNA abundances of SREBP1, ACAT1 and APO-A1 were up-regulated, while that of CYP46A1 was significantly down-regulated (P<0.05). These changes coincided with a significant down-regulation of BDNF and CRH, and a significant up-regulation of NPY mRNA expression. Moreover, genes involved in methyl transfer cycle were also modulated. DNMT1 and BHMT were up-regulated (P<0.05) at both mRNA and protein levels, which was associated with significant modifications of CpG methylation on the promoter of SREBP-1, SREBP-2 and APO-A1 genes as detected by bisulfate sequencing. These results indicate that feeding betaine to hens modulates hypothalamic expression of genes involved in cholesterol metabolism and brain functions in F1 cockerels with modification of promoter DNA methylation.


Asunto(s)
Betaína/farmacología , Pollos/genética , Metilación de ADN/efectos de los fármacos , Suplementos Dietéticos , Hipotálamo/efectos de los fármacos , Animales , Western Blotting , Colesterol/metabolismo , Femenino , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
J Agric Food Chem ; 66(2): 540-550, 2018 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-29224337

RESUMEN

Spermidine is an acetyltransferase inhibitor and a specific inducer of autophagy. Recently, spermidine is identified as a potential therapeutic agent for age-related muscle atrophy and inherited myopathies. However, the effect of spermidine on nonpathological skeletal muscle remains unclear. In this study, long-term spermidine administration in mice lowered the mean cross-sectional area of the gastrocnemius muscle and reduced the expression of myosin heavy chain isoforms in the muscle, which was associated with ubiquitination. Moreover, spermidine supplementation induced autophagy in satellite cells and enhanced satellite cell proliferation. ChIP assay revealed that spermidine repressed H3K56ac in the promoter of ACVR2B and lowered the binding affinity of Smad3 to the promoters of Myf5 and MyoD. Altogether, our results indicate that long-term administration of spermidine can activate satellite cells, as well as enhance autophagy, eventually resulting in muscle atrophy. In addition, H3K56ac and Smad3 emerged as key determinants of satellite cell activation.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Proteína smad3/metabolismo , Espermidina/farmacología , Acetilación , Receptores de Activinas Tipo II/genética , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Unión Proteica/efectos de los fármacos , Células Satélite del Músculo Esquelético/efectos de los fármacos , Proteína smad3/genética
14.
Free Radic Biol Med ; 113: 413-423, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29106990

RESUMEN

Light emitting diode (LED) light has been tested to treat traumatic brain injury, neural degenerative diseases and psychiatric disorders. Previous studies indicate that blue LED light affects cell proliferation and apoptosis in photosensitive cells and cancer cells. In this study, we demonstrate that white LED light exposure impaired proliferation and induced apoptosis in HeLa and HT-22 hippocampal neural cells, but not C2C12 cells. Furthermore, the mechanisms underlying the effect of white LED light exposure on HT-22 cells were elucidated. In HeLa and HT-22 cells, white LED light activated mitochondrial cytochrome c oxidase (Cco), in association with enhanced ATP synthase activity and elevated intracellular ATP concentration. Also, reactive oxygen species (ROS) and nitric oxide (NO) production were increased, accompanied by higher calcium concentration and lower mitochondrial membrane potential. HT-22 cells exposed to white LED light for 24h showed reduced viability, with higher apoptotic rate and a cell cycle arrest at G0/G1 phase. Concurrently, the mRNA expression and the concentration of IGF-1 were decreased, while that of TNF-α were increased, in light-exposed cells, which was supported by the luciferase activity of both gene promoters. The down-stream mitogen-activated protein kinase (MAPK), AKT/mTOR pathways were inhibited, in association with an activation of apoptotic caspase 3. N-Acetylcysteine, a ROS scavenger, protected the cells from LED light-induced cellular damage, with rescued cell viability and restored mRNA expression of IGF-1 and TNF-α. Our data demonstrate that white LED light suppresses proliferation and induces apoptosis in hippocampal neuron cells through mitochondrial Cco/ROS-mediated IGF-1 and TNF-α pathways.


Asunto(s)
Apoptosis/efectos de la radiación , Complejo IV de Transporte de Electrones/genética , Factor I del Crecimiento Similar a la Insulina/genética , Mioblastos/efectos de la radiación , Neuronas/efectos de la radiación , Factor de Necrosis Tumoral alfa/genética , Animales , Calcio/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Puntos de Control del Ciclo Celular/efectos de la radiación , Línea Celular , Línea Celular Transformada , Supervivencia Celular/efectos de la radiación , Complejo IV de Transporte de Electrones/metabolismo , Regulación de la Expresión Génica , Células HeLa , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/efectos de la radiación , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Luz , Potencial de la Membrana Mitocondrial/efectos de la radiación , Ratones , Mioblastos/citología , Mioblastos/metabolismo , Neuronas/citología , Neuronas/metabolismo , Óxido Nítrico/agonistas , Óxido Nítrico/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
15.
Poult Sci ; 96(12): 4389-4398, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29053854

RESUMEN

Betaine, an important methyl donor, is known to execute epigenetic regulation of gene expression via nutritional reprogramming. Herein, we explore whether feeding a betaine-supplemented diet to laying hens would affect corticosteroid biosynthesis in the adrenal gland and corticosterone deposition in eggs, in correlation with the expression of methyl transfer enzymes and the promoter DNA methylation status of affected genes. Rugao yellow-feathered laying hens at 38 weeks of age were assigned to Control and Betaine groups, fed basal and betaine-supplemented diets, respectively, for four weeks. Betaine supplementation significantly increased (P < 0.05) the average laying rate, while the body weight and egg quality remained unchanged. Plasma concentrations of cholesterol and low-density lipoprotein-cholesterol were also higher (P < 0.05) in the Betaine group. Moreover, eggs in the Betaine group contained higher corticosterone in the yolk, which was associated with up-regulation of steroidogenesis genes in adrenal glands. Steroidogenic acute regulatory protein (StAR), the rate-limiting protein responsible for transporting cholesterol to the inner mitochondrial membrane, was significantly activated (P < 0.05), together with its transcription factors steroidogenic factor-1 (SF-1) and glucocorticoid receptor. Also, betaine supplementation significantly up-regulated (P < 0.05) the adrenal mRNA expression of adenosyl homocysteinase-like 1 and DNA methyltransferases1 and 3a. Bisulfite sequencing analysis revealed significant hypomethylation in several CpG sites within the promoter region of SF-1 gene in the adrenal gland. These results indicate that dietary supplementation of betaine in hens activates adrenal expression of StAR, possibly through epigenetic regulation of SF-1 gene.


Asunto(s)
Proteínas Aviares/genética , Betaína/metabolismo , Pollos/genética , Pollos/metabolismo , Corticosterona/metabolismo , Yema de Huevo/química , Fosfoproteínas/genética , Glándulas Suprarrenales/metabolismo , Alimentación Animal/análisis , Animales , Proteínas Aviares/metabolismo , Betaína/administración & dosificación , Metilación de ADN , Dieta/veterinaria , Suplementos Dietéticos/análisis , Femenino , Expresión Génica , Fosfoproteínas/metabolismo
16.
Gen Comp Endocrinol ; 246: 241-248, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28039067

RESUMEN

Sterol 27-hydroxylase (CYP27A1) plays an important role in cholesterol homeostasis by degrading cholesterol to bile acids. Betaine can alleviate high-fat diet-induced hepatic cholesterol accumulation and maternal betaine treatment programs the hepatic expression of CYP27A1 in offspring. Excessive corticosterone (CORT) exposure causes hepatic cholesterol deposition in chickens, yet it remains unknown whether prenatal betaine modulates CORT-induced cholesterol accumulation in chicken liver later in life and whether it involves epigenetic gene regulation of CYP27A1. In this study, fertilized eggs were injected with saline or betaine at 2.5mg/egg before incubation, and the hatchlings were raised under the same condition till 56days of age followed by 7days of subcutaneous CORT injection. Plasma concentrations of total cholesterol (Tch), HDL- and LDL-cholesterol were significantly increased (P<0.05), after CORT challenge, in both control and betaine groups. However, prenatal betaine exposure prevented CORT-induced increase (P<0.05) in hepatic Tch content. Hepatic expression of cholesterol biosynthesis genes and ACAT1 protein that esterifies cholesterol for storage, were activated in both control and betaine groups upon CORT challenge. However, betaine-treated chickens were protected from CORT-induced repression (P<0.05) in LXR and CYP27A1 expression in the liver. CORT-induced down-regulation of LXR and CYP27A1 coincided with significantly increased (P<0.05) CpG methylation on their promoters, which was significantly ameliorated in betaine-treated chickens. These results suggest that in ovo betaine injection alleviates CORT-induced hepatic cholesterol deposition most probably through epigenetic regulation of CYP27A1 and LXR genes in juvenile chickens.


Asunto(s)
Betaína/administración & dosificación , Colestanotriol 26-Monooxigenasa/antagonistas & inhibidores , Corticosterona/farmacología , Metilación de ADN/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hígado/metabolismo , Regiones Promotoras Genéticas/genética , Envejecimiento , Animales , Animales Recién Nacidos , Antiinflamatorios/farmacología , Western Blotting , Pollos/crecimiento & desarrollo , Pollos/metabolismo , Colestanotriol 26-Monooxigenasa/genética , Colestanotriol 26-Monooxigenasa/metabolismo , Colesterol/metabolismo , Epigénesis Genética/efectos de los fármacos , Femenino , Fármacos Gastrointestinales/administración & dosificación , Inmunoprecipitación , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Sci Rep ; 7: 40251, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-28059170

RESUMEN

Betaine alleviates high-fat diet-induced fatty liver and prenatal betaine programs offspring hepatic lipid metabolism. Excessive corticosterone (CORT) exposure causes fatty liver in chickens, yet it remains unknown whether and how prenatal betaine modulates the susceptibility of CORT-induced fatty liver later in life. In this study, fertilized eggs were injected with saline or betaine before incubation, and the hatchlings were raised at 8 weeks of age followed by 7 days of subcutaneous CORT injection. CORT-induced fatty liver was less severe in betaine-treated chickens, with significantly reduced oil-red staining and hepatic triglyceride content (P < 0.05). The protective effect of prenatal betaine was associated with significantly up-regulated expression of PPARα and CPT1α, as well as mitochondrial DNA (mtDNA)-encoded genes (P < 0.05). Moreover, betaine rescued CORT-induced alterations in methionine cycle genes, which coincided with modifications of CpG methylation on CPT1α gene promoter and mtDNA D-loop regions. Furthermore, the elevation of hepatic GR protein content after CORT treatment was significantly reduced (P < 0.05), while the reduction of GR binding to the control region of affected genes was significantly increased (P < 0.05), in betaine-treated chickens. These results indicate that in ovo betaine injection protects the juvenile chickens from CORT-induced fatty liver.


Asunto(s)
Betaína/administración & dosificación , Corticosterona/administración & dosificación , Epigénesis Genética , Hígado Graso/inducido químicamente , Hígado Graso/genética , Animales , Peso Corporal/efectos de los fármacos , Pollos , Metilación de ADN , ADN Mitocondrial/genética , Ingestión de Alimentos/efectos de los fármacos , Hígado Graso/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Metionina/metabolismo , Cigoto/efectos de los fármacos , Cigoto/metabolismo
18.
Poult Sci ; 96(6): 1715-1724, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-27965408

RESUMEN

Cholesterol is essential for neuronal development and brain function. Previously we reported that in ovo administration of betaine modulates hepatic cholesterol metabolism in the chicken, yet it remains unknown whether maternal betaine affects the cholesterol content and the expression of cholesterol metabolic genes in chicken hypothalamus. In this study, eggs were injected with saline or betaine at 2.5 mg/egg, and the hatchlings were raised under the same condition until 64 d of age. Maternal betaine significantly (P = 0.05) increased the body weight and suppressed aggressive behavior of 64-day-old cockerels, in association with significantly (P < 0.05) up-regulated expression of 5-HTR1A receptor in the hypothalamus. Concurrently, betaine in ovo significantly increased (P < 0.05) the hypothalamic content of total cholesterol and cholesterol ester, which coincided with significantly up-regulated (P < 0.05) hypothalamic expression of cholesterol biosynthetic genes, such as sterol-regulatory element binding protein 2 and 3-hydroxy-3-methyl-glutaryl-CoA reductase as well as acetyl-CoA cholesterol acyltransferase 1, which converts free cholesterol to cholesterol ester for storage. In contrast, low density lipoprotein receptor, which mediates the cholesterol uptake, was significantly down-regulated (P < 0.05). In ovo betaine administration significantly enhanced the expression of betaine-homocysteine methyltransferase and DNA methyltransferase 1 (P < 0.05), which was associated with alterations of CpG methylation on the promoter of modified cholesterol metabolic genes. These results indicate that maternal betaine modulates hypothalamic cholesterol metabolism in cockerels through modifying DNA methylation on the promoter of cholesterol metabolic genes.


Asunto(s)
Betaína/farmacología , Pollos/metabolismo , Colesterol/biosíntesis , Metilación de ADN/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Agresión/efectos de los fármacos , Animales , Betaína/administración & dosificación , Peso Corporal/efectos de los fármacos , Pollos/genética , Pollos/crecimiento & desarrollo , Colesterol/metabolismo , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Masculino , Óvulo/efectos de los fármacos , Óvulo/metabolismo
19.
Int J Biochem Cell Biol ; 79: 222-230, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27592453

RESUMEN

Folate deficiency contributes to impaired adult hippocampal neurogenesis, yet the mechanisms remain unclear. Here we use HT-22 hippocampal neuron cells as model to investigate the effect of folate deprivation (FD) on cell proliferation and apoptosis, and to elucidate the underlying mechanism. FD caused cell cycle arrest at G0/G1 phase and increased the rate of apoptosis, which was associated with disrupted expression of folate transport and methyl transfer genes. FOLR1 and SLC46A1 were (P<0.01) down-regulated, while SLC19A1 was up-regulated (P<0.01) in FD group. FD cells exhibited significantly (P<0.05) higher protein content of BHMT, MAT2b and DNMT3a, as well as increased SAM/SAH concentrations and global DNA hypermethylation. The expression of the total and all the 3 classes of IGF-1 mRNA variants was significantly (P<0.01) down-regulated and IGF-1 concentration was decreased (P<0.05) in the culture media. IGF-1 signaling pathway was also compromised with diminished activation (P<0.05) of STAT3, AKT and mTOR. CpG hypermethylation was detected in the promoter regions of IGF-1 and FOLR1 genes, while higher SLC19A1 mRNA corresponded to hypomethylation of its promoter. IGF-1 supplementation in FD media significantly abolished FD-induced decrease in cell viability. However, IGF-1 had limited effect in rescuing the cell phenotype when added 24h after FD. Taken together, down-regulation of IGF-1 expression and signaling is involved in FD-induced cell cycle arrest and apoptosis in HT-22 hippocampal neuron cells, which is associated with an abnormal activation of methyl transfer pathway and hypermethylation of IGF-1 gene promoter.


Asunto(s)
Apoptosis , Deficiencia de Ácido Fólico/patología , Puntos de Control de la Fase G1 del Ciclo Celular , Hipocampo/patología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neuronas/patología , Fase de Descanso del Ciclo Celular , Animales , Transporte Biológico , Línea Celular , Proliferación Celular , Supervivencia Celular , Islas de CpG/genética , Metilación de ADN , Regulación hacia Abajo , Epigénesis Genética , Receptor 1 de Folato/genética , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Transportadores de Ácido Fólico/genética , Ratones , Regiones Promotoras Genéticas/genética , Proteína Portadora de Folato Reducido/genética , Transducción de Señal
20.
Steroids ; 115: 26-33, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27500692

RESUMEN

Prenatal exposure to excessive glucocorticoids (GCs) leads to intrauterine growth retardation and fetal programming of adult health and disease through deregulation of placental functions. Placental secretion of insulin-like growth factor-I (IGF-I) plays a critical role in the regulation of placental development and function. However, it remains elusive whether GCs affect placental functions through glucocorticoid receptor (GR)-mediated transcriptional regulation of IGF-I gene. In this study, human placental choriocarcinoma (BeWo) cells before and after syncytialization were used as cytotrophoblast and syncytiotrophoblast models, respectively, to explore the effects of dexamethasone (Dex) on transcriptional regulation of IGF-I gene at both stages. Dex significantly inhibited (P<0.05) cell proliferation in cytotrophoblasts and down-regulated amino acid transporter SLC7A5 in syncytiotrophoblasts. Concurrently, the abundance of IGF-I mRNA and its transcript variants, together with IGF-I level in culture media, were significantly reduced, in association with significantly enhanced (P<0.05) GR phosphorylation. GR antagonist RU486 was able to abolish all these effects. Two glucocorticoid response elements (GREs) were predicted in the promoter regions of IGF-I gene. GR binding to GRE1 was significantly enriched (P<0.05) in both cytotrophoblasts and syncytiotrophoblasts, but that to GRE2 was significantly diminished (P<0.05) in cytotrophoblasts but not in syncytiotrophoblasts, in response to Dex treatment. IGF-I supplementation completely rescued Dex-induced cell cycle arrest but not SLC7A5 down-regulation, indicating different regulatory mechanisms. Taken together, our results suggest that GR-mediated transcriptional regulation of IGF-I is involved in Dex-induced inhibition of placental cell proliferation and function.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Receptores de Glucocorticoides/metabolismo , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Dexametasona/farmacología , Técnica del Anticuerpo Fluorescente , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Transportador de Aminoácidos Neutros Grandes 1/genética , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Radioinmunoensayo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Glucocorticoides/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA