Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Life Sci ; 285: 119951, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34516994

RESUMEN

AIMS: We sought to evaluate the effects of overfeeding during lactation on the feeding behavior and expression of specific regulatory genes in brain areas associated with food intake in 22- and 60-day old male rats. METHODS: We evaluated body weight, food intake of standard and palatable diet, and mRNA expression of dopamine receptor D1 (DDR1), dopamine receptor (DDR2), melanocortin 4 receptor (MC4R), the µ-opioid receptor (MOR), neuropeptide Y (NPY), agouti-related protein (AGRP), proopiomelanocortin (POMC), cocaine-and amphetamine-regulated transcript (CART), serotonin (5-hydroxytryptamine; 5-HT) transporter (SERT), 5-hydroxytryptamine receptor 1B (5-HT1B), 5-hydroxytryptamine receptor 2C receptor (5-HT2C), Clock (CLOK), cryptochrome protein 1 (Cry1) and period circadian protein homolog 2 (Per2) in the striatum, hypothalamus and brainstem of male rats at post-natal days (PND) 22 and 60. KEY FINDINGS: Overfeeding resulted in significantly increased body weight through PND60, and a 2-fold increase in palatable food intake at PND22, but not at PND60. We observed significant increases in DDR1, DDR2, and MC4R gene expression in the striatum and brainstem and POMC/CART in the hypothalamus of the OF group at PND22 that were reversed by PND60. Hypothalamic levels of 5-HT1B, 5-HT2C and NPY/AGRP on the other hand were decreased at PND22 and increased at PND60 in OF animals. Clock genes were unaffected by OF at PND22, but were significantly elevated at PND60. SIGNIFICANCE: Overfeeding during early development of the rat brain results in obesity and altered feeding behavior in early adulthood. The altered behavior might be the consequence of the changes in food intake and reward gene expression.


Asunto(s)
Peso Corporal , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiopatología , Conducta Alimentaria , Hipernutrición/fisiopatología , Animales , Proteínas CLOCK/metabolismo , Criptocromos/metabolismo , Ingestión de Alimentos , Femenino , Lactancia , Masculino , Proteínas de Unión al ARN/metabolismo , Ratas , Ratas Wistar , Receptor de Serotonina 5-HT1B/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo
2.
Nutr Metab (Lond) ; 16: 63, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31528182

RESUMEN

BACKGROUND: Obesity can lead to a chronic systemic inflammatory state that increases the risk of cancer development. Therefore, this study aimed to evaluate the alterations in tumor non-infiltrated lymphocytes function and melanoma growth in animals maintained on a high-fat diet and/or moderate physical exercise program in a murine model of melanoma. METHODS: Female mice were randomly divided into eight groups: 1) normolipidic control (N), 2) normolipidic + melanoma (NM), 3) high-fat control (H), 4) high-fat + melanoma (HM), 5) normolipidic control + physical exercise (NE), 6) normolipidic melanoma + physical exercise (NEM), 7) high-fat control + physical exercise (HE), and 8) high-fat melanoma + physical exercise (HEM). After 8 weeks of diet treatment and/or moderate physical exercise protocol, melanoma was initiated by explanting B16F10 cells into one-half of the animals. RESULTS: Animals fed a high-fat diet presented high-energy consumption (30%) and body weight gain (H and HE vs N and NE, 37%; HM and HEM vs NM and NEM, 73%, respectively), whether or not they carried melanoma explants. Although the tumor growth rate was higher in animals from the HM group than in animals from any other sedentary group, it was reduced by the addition of a physical exercise regimen. We also observed an increase in stimulated peripheral lymphocyte proliferation and a decrease in the T-helper 1 response in the HEM group. CONCLUSIONS: The results of the present study support the hypothesis that altering function of tumor non-infiltrated lymphocytes via exercise-related mechanisms can slow melanoma progression, indicating that the incorporation of a regular practice of moderate-intensity exercises can be a potential strategy for current therapeutic regimens in treating advanced melanoma.

3.
Life Sci ; 232: 116579, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31252001

RESUMEN

AIMS: We sought to evaluate the effects of maternal protein restriction (LP) on oxidative balance and transcription factors for mitochondrial biogenesis in the hearts of young female rats of both the first (F1) and second (F2) generation. MAIN METHODS: We evaluated oxidative stress biomarkers (lipid peroxidation and protein oxidation), enzymatic antioxidant defense (activity of superoxide dismutase-SOD, catalase, and glutathione-S-transferase-GST), nonenzymatic antioxidant defense (reduced glutathione-GSH and sulfhydryl groups) and gene expression of AMPK, PGC-1α and TFAM. KEY FINDINGS: Interestingly, lipid peroxidation was decreased (49%, p < 0.001) in the LP-F1 group and 59% (p < 0.001) in LP-F2. In enzymatic defense, we observed increases in SOD activity in the LP-F1 group (79%, p = 0.036) and in CAT activity (approximately 40%, p = 0.041). GSH was increased in F2 in both groups (LP 546%, p < 0.0001 and in NP 491.7%, p < 0.0001). With respect to mitochondrial biogenesis gene transcription, we observed a decrease in AMPK (60%, p < 0. 0001) and an increase in PGC-1α (340%, p < 0.001) in LP compared to NP in the F1 generation. TFAM was decreased in LP-F2L compared to NP-F2L (42%, p = 0.0069) and increased in LP-F2 compared to LP-F1 (160%, p = 0.0037). SIGNIFICANCE: Our study contributes to knowledge of inheritance, showing that despite the potential mitochondrial 'inheritance' of cardiovascular damage caused by maternal malnutrition, that damage is not cross-generational and can be eliminated with proper nutrition in the F1 generation.


Asunto(s)
Miocardio/metabolismo , Estrés Oxidativo/fisiología , Desnutrición Proteico-Calórica/metabolismo , Animales , Antioxidantes/farmacología , Femenino , Glutatión/metabolismo , Corazón/efectos de los fármacos , Corazón/fisiología , Herencia/genética , Peroxidación de Lípido/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Biogénesis de Organelos , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Ratas , Superóxido Dismutasa/metabolismo , Factores de Transcripción/metabolismo
4.
Appl Physiol Nutr Metab ; 44(2): 164-171, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30058348

RESUMEN

There is a strong correlation between inadequate gestational and postpartum nutrition and the occurrence of cardiovascular diseases. The present study investigated the effects of a maternal low-protein diet and neonatal overfeeding on the oxidative balance and morphology of the renal cortex of male Wistar rats. Two independent protocols were used. First, pregnant Wistar rats received diets containing either 17% (normal protein) or 8% (low protein) casein throughout pregnancy and lactation. Second, the litter size was reduced by one-third on the third postnatal day to induce overnourishment in offspring. At 30 days, the oxidative balance and morphology of the renal cortex were analyzed. There was a small but significant increase in renal corpuscle area in the low protein (LP, 5%) and overnutrition (ON, 8%) groups. Glomerular tuft area also increased in LP (6%) and ON (9%), as did glomerular cellularity (LP, +11%; ON, +12%). In the oxidative stress analyses, both nutritional insults significantly elevated lipid peroxidation (LP, +18%; ON, +135%) and protein oxidation (LP, +40%; ON, +65%) while significantly reducing nonenzymatic antioxidant defenses, measured as reduced glutathione (LP, -32%; ON, -45%) and total thiol content (LP, -28%; ON, -24%). We also observed a decrease in superoxide dismutase (LP, -78%; ON, -51%), catalase (LP, -18%; ON, -61%), and glutathione S-transferase (only in ON, -44%) activities. Our results demonstrate that nutritional insults, even those of a very different nature, during perinatal development can result in similar changes in oxidative parameters and glomerular morphology in the renal cortex.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Corteza Renal/metabolismo , Glomérulos Renales/patología , Hipernutrición/metabolismo , Hipernutrición/patología , Estrés Oxidativo , Animales , Animales Recién Nacidos , Antioxidantes/metabolismo , Peso Corporal , Femenino , Corteza Renal/patología , Glomérulos Renales/metabolismo , Peroxidación de Lípido , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Wistar
5.
Oncol Lett ; 16(1): 1285-1290, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30061948

RESUMEN

Previous reports have indicated that the abundance of specific microRNAs (miRNA) contained within the exosome/microvesicle compartment of patient biofluids may be useful in diagnosing specific types of cancer. In the present study, the 786-O cell line, which is derived from a clear cell renal cell carcinoma (ccRCC), was used as an in vitro ccRCC tumor model and the human renal proximal tubule cell line HK-2 was used as its normal renal tissue control to investigate the similarities of exosomal content of selected ccRCC miRNA biomarkers in the supernatant with the content of those markers in the cells themselves. A PCR array identified miRNA biomarkers of solid RCC tumors (miR-210, MiR-34a, miR-155-5p and miR-150-5p) that were increased by 2-8 fold in 786-O exosomes compared with the control. These were subsequently chosen for further investigation using TaqMan RT-qPCR in addition to miR-15a and miR-205, which were selected based on prior interest as RCC biomarkers. MiR-15a, -34a, -210 and -155 levels were significantly lower in exosomes when compared with that in whole cells but did not differ between the HK-2 and 786-O cells in either the cytoplasmic, exosome or exosome-free supernatant fractions. By contrast, cytoplasmic miR-150 and miR-205 exhibited significant differences in concentration between the two cell lines. In addition, the cytoplasmic content of miR-150 and miR-205 was mirrored in the exosomal content of these miRNAs. Furthermore, the difference in exosomal miR-205 content was statistically significant. The present study indicated that measurements of the exosomal content of miR-205 and possibly miR-150, but not those of the other examined miRNAs, are proportional to their respective contents in the cells that secreted them. These findings suggest that in vitro RCC systems may be useful in identifying miRNAs with sufficiently high levels of exportation into exosomes; and with sufficiently different expression levels between tumor and normal cells to serve as ccRCC biomarkers in vivo.

6.
Nutr Neurosci ; 21(8): 580-588, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28494696

RESUMEN

Many studies have shown that a maternal low-protein diet increases the susceptibility of offspring to cardiovascular disease in later-life. Moreover, a lower incidence of cardiovascular disease in females than in males is understood to be largely due to the protective effect of high levels of estrogens throughout a woman's reproductive life. However, to our knowledge, the role of estradiol in moderating the later-life susceptibility of offspring of nutrient-deprived mothers to cardiovascular disease is not fully understood. The present study is aimed at investigating whether oxidative stress in the brainstem caused by a maternal low-protein diet administered during a critical period of fetal/neonatal brain development (i.e during gestation and lactation) is affected by estradiol levels. Female Wistar rat offspring were divided into four groups according to their mothers' diets and to the serum estradiol levels of the offspring at the time of testing: (1) 22 days of age/control diet: (2) 22 days of age/low-protein diet; (3) 122 days of age/control diet: (4) 122 days of age/low-protein diet. Undernutrition in the context of low serum estradiol compared to undernutrition in a higher estradiol context resulted in increased levels of oxidative stress biomarkers and a reduction in enzymatic and non-enzymatic antioxidant defenses. Total global oxy-score showed oxidative damage in 22-day-old rats whose mothers had received a low-protein diet. In the 122-day-old group, we observed a decrease in oxidative stress biomarkers, increased enzymatic antioxidant activity, and a positive oxy-score when compared to control. We conclude from these results that following a protein deficiency in the maternal diet during early development of the offspring, estrogens present at high levels at reproductive age may confer resistance to the oxidative damage in the brainstem that is very apparent in pre-pubertal rats.


Asunto(s)
Tronco Encefálico/metabolismo , Dieta con Restricción de Proteínas/efectos adversos , Desnutrición/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Neuronas/metabolismo , Neuroprotección , Estrés Oxidativo , Animales , Animales Recién Nacidos , Biomarcadores/sangre , Biomarcadores/metabolismo , Tronco Encefálico/enzimología , Estradiol/sangre , Femenino , Glutatión/metabolismo , Glutatión Transferasa/metabolismo , Lactancia , Peroxidación de Lípido , Desnutrición/sangre , Desnutrición/etiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/enzimología , Oxidación-Reducción , Oxidorreductasas/metabolismo , Embarazo , Carbonilación Proteica , Ratas Wistar
7.
Life Sci ; 192: 190-198, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29191645

RESUMEN

During their reproductive years women produce significant levels of estrogens, predominantly in the form of estradiol, that are thought to play an important role in cardioprotection. Mechanisms underlying this action include both estrogen-mediated changes in gene expression, and post-transcriptional activation of protein signaling cascades in the heart and in neural centers controlling cardiovascular function, in particular, in the brainstem. There, specific neurons, especially those of the bulbar region play an important role in the neuronal control of the cardiovascular system because they control the outflow of sympathetic activity and parasympathetic activity as well as the reception of chemical and mechanical signals. In the present review, we discuss how estrogens exert their cardioprotective effect in part by modulating the actions of internally generated products of cellular oxidation such as reactive oxygen species (ROS) in brain stem neurons. The significance of this review is in integrating the literature of oxidative damage in the brain with the literature of neuroprotection by estrogen in order to better understand both the benefits and limitations of using this hormone to prevent cardiovascular disease.


Asunto(s)
Química Encefálica/efectos de los fármacos , Encéfalo/fisiopatología , Cardiotónicos/farmacología , Enfermedades Cardiovasculares/fisiopatología , Estrógenos/farmacología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Cardiotónicos/uso terapéutico , Enfermedades Cardiovasculares/prevención & control , Estrógenos/uso terapéutico , Humanos , Fármacos Neuroprotectores/uso terapéutico
8.
J Thyroid Res ; 2016: 9276402, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27379194

RESUMEN

Exosomes are 30-100 nm, membrane-bound vesicles containing specific cellular proteins, mRNAs, and microRNAs that take part in intercellular communication between cells. A possible role for exosomes in thyroid function has not been fully explored. In the present study, FRTL-5 rat thyroid cells were grown to confluence and received medium containing either thyroid stimulating hormone (TSH), exogenous bovine thyroglobulin (bTg), or neither additive for 24 or 48 hours followed by collection of spent medium and ultracentrifugation to isolate small vesicles. Transmission electron microscopy and Western blotting for CD9 indicated the presence of exosomes. Western blotting of exosome extract using a monoclonal anti-Tg antibody revealed a Tg-positive band at ~330 kDa (the expected size of monomeric Tg) with a higher density in TSH-treated cells compared to that in untreated cells. These results are the first to show that normal thyroid cells in culture produce exosomes containing undegraded Tg.

9.
Microrna ; 4(1): 26-35, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26165467

RESUMEN

New therapies based on the targeting of signal pathways (such as VHL/HIF-1) common to most renal cell carcinomas (RCC), have greatly improved the outlook for sufferers of this disease. Given the growing reputation of many microRNAs (miRNAs) as both tumor suppressors and oncogenes, the measurement and manipulation of these small nucleotides in RCC patients may provide yet another valuable advance in renal cancer diagnosis and treatment. The present review summarizes the current literature on the role of microRNAs in RCC development and progression emphasizing the interaction of specific miRNAs with both oncogenic and tumor suppressor pathways of particular importance in renal cancer.


Asunto(s)
Carcinoma de Células Renales/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/genética , Riñón/patología , MicroARNs/genética , Animales , Carcinoma de Células Renales/diagnóstico , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Humanos , Riñón/metabolismo , Neoplasias Renales/diagnóstico , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , MicroARNs/análisis , MicroARNs/metabolismo , Pronóstico , Transporte de ARN , Transducción de Señal
10.
Mol Endocrinol ; 28(3): 368-79, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24479877

RESUMEN

Thyroglobulin (Tg), stored in the follicular lumen, has also been shown recently to perform two unexpected roles: as an autocrine negative-feedback suppressor of thyroid function in the presence of TSH and as a potent inducer of thyroid cell growth in the absence of TSH. However, the underlying molecular mechanism(s) remain unclear. To elucidate a molecular pathway linking Tg to increased cell proliferation, we examined the regulation of microRNAs (miRNAs) by Tg using an miRNA microarray. We identified 21 miRNAs whose expression was significantly suppressed by Tg in rat thyroid FRTL-5 cells. Using specific miRNA analogs, we determined that miR-16, miR-24, and miR-195 mediate the induction of thyroid cell growth by Tg. The expression of miR-16 and miR-195 target genes, Mapk8, Ccne1, and Cdc6, which were previously shown to be essential for TSH-stimulated thyroid cell growth, were also induced by Tg. Moreover, the Tg-induced expression of these genes was reduced by overexpression of miR-16 and miR-195. Similarly, the induction of c-Myc by Tg was reduced by miR-24 overexpression. These results suggest that Tg could alter thyroid cell proliferation by increasing the expression of cell division-related genes such as Mapk8, Ccne1, Cdc6, and c-Myc through its suppression of specific microRNAs (miR-16, miR-24, and miR-195). In addition, we identified phosphatidylinositol 3-kinase as a key signaling pathway, linking Tg with cell proliferation. The present data support an important role for miRNAs as effectors for the effect of Tg on cell proliferation and perhaps other functions of Tg in the thyroid cell.


Asunto(s)
Proliferación Celular , MicroARNs/metabolismo , Tiroglobulina/fisiología , Glándula Tiroides/citología , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , MicroARNs/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , Ratas , Transducción de Señal , Transcriptoma
11.
J Clin Endocrinol Metab ; 99(4): E694-702, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24433000

RESUMEN

CONTEXT: It was shown in the rat thyroid that thyroglobulin (Tg) stored in the follicular lumen is a potent regulator of thyroid-specific gene expression to maintain the function of individual follicles. However, the actions of Tg as a regulatory molecule in human thyroid have not been studied. OBJECTIVE: Our objective was to determine the effect of Tg on gene expression in normal and diseased human thyroid and to examine whether the proposed model of negative-feedback autocrine regulation of thyroid function by Tg is applicable in the human as well as the rat. DESIGN: Primary cultures of human thyrocytes were established from normal thyroid, Graves' disease thyroid, adenomatous goiter, follicular adenoma, and papillary carcinoma tissues obtained during surgery. Cells were stimulated with physiologic (ie, follicular) concentrations of Tg, and mRNA and protein expression of genes involved in thyroid hormonogenesis were evaluated. The effects of Tg on thyroid-specific gene expression were also assessed in 2 human papillary carcinoma cell lines. RESULTS: Transcript levels of genes participating in thyroid hormone biosynthesis were significantly reduced by Tg in thyrocyte cultures derived from normal and Graves' thyroid, but not in cultures derived from thyroid neoplasms and adenomatous goiter. CONCLUSION: It was confirmed that Tg acts as a negative-feedback regulator of gene expression in human thyrocytes, suggesting that Tg signaling may constitute a common mechanism for maintaining thyroid homeostasis in species with follicular thyroid morphology. However, certain diseases of intrinsic thyroid overgrowth appear to be associated with an escape from the regulatory mechanism of Tg.


Asunto(s)
Tiroglobulina/farmacología , Glándula Tiroides/citología , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Adenoma/genética , Adenoma/patología , Adulto , Autoantígenos/genética , Autoantígenos/metabolismo , Carcinoma Papilar/genética , Carcinoma Papilar/patología , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Bocio/genética , Bocio/patología , Enfermedad de Graves/genética , Enfermedad de Graves/patología , Humanos , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Masculino , Especificidad de Órganos/genética , Cultivo Primario de Células
12.
Thyroid ; 24(4): 625-38, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24251883

RESUMEN

BACKGROUND: The established paradigm for thyroglobulin (Tg) function is that of a high molecular weight precursor of the much smaller thyroid hormones, triiodothyronine (T3) and thyroxine (T4). However, speculation regarding the cause of the functional and morphologic heterogeneity of the follicles that make up the thyroid gland has given rise to the proposition that Tg is not only a precursor of thyroid hormones, but that it also functions as an important signal molecule in regulating thyroid hormone biosynthesis. SUMMARY: Evidence supporting this alternative paradigm of Tg function, including the up- or downregulation by colloidal Tg of the transcription of Tg, iodide transporters, and enzymes employed in Tg iodination, and also the effects of Tg on the proliferation of thyroid and nonthyroid cells, is examined in the present review. Also discussed in detail are potential mechanisms of Tg signaling in follicular cells. CONCLUSIONS: Finally, we propose a mechanism, based on experimental observations of Tg effects on thyroid cell behavior, that could account for the phenomenon of follicular heterogeneity as a highly regulated cycle of increasing and decreasing colloidal Tg concentration that functions to optimize thyroid hormone production through the transcriptional activation or suppression of specific genes.


Asunto(s)
Tiroglobulina/fisiología , Glándula Tiroides/fisiología , Animales , Receptor de Asialoglicoproteína/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Expresión Génica , Homeostasis , Humanos , Modelos Biológicos , Receptores de Superficie Celular/fisiología , Transducción de Señal , Tiroglobulina/genética , Glándula Tiroides/anatomía & histología , Glándula Tiroides/citología , Hormonas Tiroideas/biosíntesis , Factores de Transcripción/genética
13.
PLoS One ; 7(12): e50390, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23239980

RESUMEN

In this study, we investigated the effect of glutamine (Gln) supplementation on the signaling pathways regulating protein synthesis and protein degradation in the skeletal muscle of rats with streptozotocin (STZ)-induced diabetes. The expression levels of key regulatory proteins in the synthetic pathways (Akt, mTOR, GSK3 and 4E-BP1) and the degradation pathways (MuRF-1 and MAFbx) were determined using real-time PCR and Western blotting in four groups of male Wistar rats; 1) control, non-supplemented with glutamine; 2) control, supplemented with glutamine; 3) diabetic, non-supplemented with glutamine; and 4) diabetic, supplemented with glutamine. Diabetes was induced by the intravenous injection of 65 mg/kg bw STZ in citrate buffer (pH 4.2); the non-diabetic controls received only citrate buffer. After 48 hours, diabetes was confirmed in the STZ-treated animals by the determination of blood glucose levels above 200 mg/dL. Starting on that day, a solution of 1 g/kg bw Gln in phosphate buffered saline (PBS) was administered daily via gavage for 15 days to groups 2 and 4. Groups 1 and 3 received only PBS for the same duration. The rats were euthanized, and the soleus muscles were removed and homogenized in extraction buffer for the subsequent measurement of protein and mRNA levels. The results demonstrated a significant decrease in the muscle Gln content in the diabetic rats, and this level increased toward the control value in the diabetic rats receiving Gln. In addition, the diabetic rats exhibited a reduced mRNA expression of regulatory proteins in the protein synthesis pathway and increased expression of those associated with protein degradation. A reduction in the skeletal muscle mass in the diabetic rats was observed and was alleviated partially with Gln supplementation. The data suggest that glutamine supplementation is potentially useful for slowing the progression of muscle atrophy in patients with diabetes.


Asunto(s)
Glutamina/administración & dosificación , Músculo Esquelético , Biosíntesis de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Animales , Diabetes Mellitus Experimental/dietoterapia , Diabetes Mellitus Experimental/metabolismo , Suplementos Dietéticos , Humanos , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Proteínas , Ratas , Ratas Wistar , Transducción de Señal
14.
Vascul Pharmacol ; 57(2-4): 98-104, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22583821

RESUMEN

C type natriuretic peptide (CNP) functions as a paracrine/autocrine vasoprotectant. CNP mRNA is up-regulated in human vascular smooth muscle cells (SMC) by PDGF-BB via a protein kinase C (PKC)-dependent pathways, and by general PKC activation with phorbol myristate acetate (PMA). In this report we examine the calcium dependence and isotype specificity of these PKC/CNP pathways. The PKC-δ-specific inhibitor rottlerin blocked the increase in CNP mRNA and immunoreactive CNP following treatment of aortic SMC (AoSMC) with PDGF-BB. A 300-400-fold PMA-induced elevation of CNP transcript levels in AoSMC and a ~40-fold increase in human aortic endothelial cells (HAEC) were reduced by PKC-α- and PKC-δ-, but not PKC-ß-specific inhibitors. siRNA silencing of PKC-δ reduced PDGF-, but not PMA-stimulated CNP transcript in SMC. Inhibition of intracellular Ca(2+) mobilization abolished a PMA-stimulated increase in CNP transcript in both SMC and HAEC. The results of this study show that PDGF increases CNP in SMC via a protein kinase C-δ-dependent pathway. In contrast, PMA increases CNP expression using PKC-α- and PKC-δ-pathways in both SMC and HAEC. A 8-10-fold greater PMA-induced increase in CNP transcript in SMC than in HAEC suggests that smooth muscle cells could be selectively targeted for CNP up-regulation by PKC-α- and PKC-δ-activators.


Asunto(s)
Calcio/metabolismo , Péptido Natriurético Tipo-C/metabolismo , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-delta/metabolismo , Acetofenonas/farmacología , Aorta/efectos de los fármacos , Aorta/metabolismo , Becaplermina , Benzopiranos/farmacología , Células Cultivadas , Silenciador del Gen , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , ARN Mensajero/metabolismo , ARN Interferente Pequeño/administración & dosificación , Acetato de Tetradecanoilforbol/farmacología
15.
Peptides ; 32(9): 1964-71, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21816187

RESUMEN

C-type natriuretic peptide (CNP) is a member of the small family of natriuretic peptides that also includes atrial natriuretic peptide (ANP) and brain, or B-type natriuretic peptide (BNP). Unlike them, it performs its major functions in an autocrine or paracrine manner. Those functions, mediated through binding to the membrane guanylyl cyclase natriuretic peptide receptor B (NPR-B), or by signaling through the non-enzyme natriuretic peptide receptor C (NPR-C), include the regulation of endochondral ossification, reproduction, nervous system development, and the maintenance of cardiovascular health. To date, the regulation of CNP gene expression has not received the attention that has been paid to regulation of the ANP and BNP genes. CNP expression in vitro is regulated by TGF-ß and receptor tyrosine kinase growth factors in a cell/tissue-specific and sometimes species-specific manner. Expression of CNP in vivo is altered in diseased organs and tissues, including atherosclerotic vessels, and the myocardium of failing hearts. Analysis of the human CNP gene has led to the identification of a number of regulatory sites in the proximal promoter, including a GC-rich region approximately 50 base pairs downstream of the Tata box, and shown to be a binding site for several putative regulatory proteins, including transforming growth factor clone 22 domain 1 (TSC22D1) and a serine threonine kinase (STK16). The purpose of this review is to summarize the current literature on the regulation of CNP expression, emphasizing in particular the putative regulatory elements in the CNP gene and the potential DNA-binding proteins that associate with them.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Regulación de la Expresión Génica , Péptido Natriurético Tipo-C/metabolismo , Animales , Enfermedades Cardiovasculares/sangre , Sistema Cardiovascular/fisiopatología , Células Endoteliales/metabolismo , Genoma Humano , Humanos , Miocitos del Músculo Liso/metabolismo , Péptido Natriurético Tipo-C/sangre , Péptido Natriurético Tipo-C/genética , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Elementos Reguladores de la Transcripción , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Factores de Transcripción , Factor de Crecimiento Transformador beta/metabolismo
16.
Am J Physiol Heart Circ Physiol ; 299(6): H2018-27, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20802130

RESUMEN

C-type natriuretic peptide (CNP) possesses nitric oxide-like signaling mechanisms and actions in the vasculature, including the inhibition of fibrosis and vascular remodeling through counterregulation of transforming growth factor-ß (TGF-ß) signaling. The leucine zipper protein transforming growth factor stimulated clone 22 domain 1 (TSC22D1), cloned via its presumed binding to a GC-rich element in the CNP promoter, was the first protein to be described as a CNP transcription factor, but the lack of supporting evidence since its discovery and its lack of a classical DNA-binding site have left in question its role in the regulation of CNP by TGF-ß and other factors. To define a specific role for TSC22D1 in CNP transcription, we have examined the effects of the profibrotic growth factors TGF-ß1 and PDGF-BB on CNP mRNA expression in cultured human vascular smooth muscle cells (SMC) in which TSC22D1 expression was suppressed with small interfering RNA. Results showed that TGF-ß and PDGF-BB significantly increased CNP expression in all three SMC types. Twenty-four-hour TGF-ß-induced elevations in CNP were strongly correlated with changes in TSC22D1 mRNA levels, and both genes exhibited their greatest response to TGF-ß1 in coronary artery SMC. Furthermore, siRNA suppression of TSC22D1 expression in coronary artery and aortic SMC by ∼90% resulted in 45-65% reductions of both PDGF- and TGF-ß-stimulated CNP expression, respectively. These results support a postulated role of TSC22D1 as an enhancer of CNP transcription and suggest that TGF-ß-induced upregulation of CNP expression in SMC may be mediated in part by increased transcription of TSC22D1.


Asunto(s)
Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Péptido Natriurético Tipo-C/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Becaplermina , Células Cultivadas , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Humanos , Péptido Natriurético Tipo-C/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Interferencia de ARN , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Factores de Tiempo , Transcripción Genética , Regulación hacia Arriba
17.
Growth Horm IGF Res ; 20(4): 326-32, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20576456

RESUMEN

OBJECTIVE: Elevated neutral lipid content and mRNA expression of class A scavenger receptor (SRA) have been found in the renal cortex of the bovine growth hormone (bGH) mouse model of progressive glomerulosclerosis (GS). We hypothesize that the increased expression of SRA precedes glomerular scarring in this model. DESIGN: Real time RT-PCR and immunofluorescence were employed to measure SRA and collagen types I and IV in the bGH transgenic and control mice at 5 and 12 weeks (wk) of age to determine the chronology of change in SRA expression in relation to glomerular scarring. Alternative mechanisms for increasing glomerular lipid were assessed by measuring mRNA expression levels of low-density lipoprotein receptor (LDL-r), 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR), and ATP-binding cassette transporter A1 (ABCA1). In addition, the involvement of macrophages in early GS was assessed by CD68 mRNA expression in kidney cortex. RESULTS: Both mRNA and protein levels of SRA were significantly increased in 5-wk bGH compared with control mice, whereas the expression of collagen I and IV was unaltered. Unchanged levels of LDL-r and HMGR mRNA indicate that neither regulated cholesterol uptake via LDL-r nor the cholesterol synthetic pathway played a role in the early lipid increase. The finding of increased ABCA1 expression was an indicator of excess intracellular lipid in the renal cortex of bGH mice at 5 wk. CD68 expression in bGH did not differ significantly from that of controls at 5 wk suggesting that cortical macrophage infiltration was not increased in bGH mice at this time point. CONCLUSION: An early increase in SRA mRNA and protein expression in the bGH kidney precedes glomerular scarring and is independent of macrophage influx.


Asunto(s)
Hormona del Crecimiento/genética , Corteza Renal/metabolismo , Enfermedades Renales/metabolismo , Glomérulos Renales , Receptores Depuradores de Clase A/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Mesangio Glomerular/química , Hormona del Crecimiento/metabolismo , Glomérulos Renales/metabolismo , Metabolismo de los Lípidos , Lípidos/sangre , Ratones , Ratones Transgénicos , Modelos Animales , ARN Mensajero/metabolismo , Receptores de LDL/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptores Depuradores de Clase A/genética
18.
Biochem Biophys Res Commun ; 389(2): 269-73, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19716808

RESUMEN

Thyroglobulin (Tg) is an essential substrate for thyroid hormone biosynthesis whose production is primarily limited to the thyroid follicular cell. We have previously identified an approximately 1.2 kb fragment of Tg mRNA in cultured mouse mesangial cells, and in the present study provide evidence showing that this transcript is transcribed and translated into a unique protein (kTg) in the kidney, but not the thyroid gland. Cloning of kTg from a mouse kidney cDNA library showed that transcription starts in the middle of intron 41 of the Tg gene and continues in-frame with the remaining coding sequence of thyroid-derived Tg beginning with exon 42. Translation of this mRNA is predicted to yield a protein of 367 amino acids (40 kDa) containing a unique 13 amino acid sequence serving as a signal peptide followed by a 354 amino acid segment identical to the carboxy-terminal end of thyroid Tg. Western blot analysis with an antibody directed against the C-terminus of thyroid Tg detected a 40 kDa protein expressed in the kidney. Immunohistochemistry with this antibody showed that immunoreactive Tg was localized in podocytes and the mesangial area of the renal glomerulus. A part of a homologous transcript was also detected in human kidney, and the kTg protein was recognized by sera from Hashimoto's thyroiditis but not from controls. Together these results suggest that a unique low molecular weight variant of Tg is expressed in the kidney, where it could serve both physiological and pathological roles, including that of an autoantigen.


Asunto(s)
Glomérulos Renales/inmunología , Tiroglobulina/inmunología , Secuencia de Aminoácidos , Animales , Autoantígenos/genética , Autoantígenos/inmunología , Clonación Molecular , Biblioteca de Genes , Humanos , Ratones , Datos de Secuencia Molecular , Tiroglobulina/genética
19.
Vascul Pharmacol ; 51(1): 29-36, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19275966

RESUMEN

C-type natriuretic peptide (CNP) inhibits the migration of vascular smooth muscle cells (VSMC) and related cell types induced by oxidized LDL and its major atherogenic component, lysophosphatidylcholine (LPC). CNP expression is increased in smooth muscle in atherosclerotic and restenotic lesions, but mechanisms underlying this increase have not been elucidated. We therefore investigated the role of LPC in CNP expression in human aortic VSMC. Both LDL and HDL elevated CNP transcript levels approximately 3-fold in VSMC, but not in endothelial cells. Increasing LPC in both VSMC and endothelial cells induced significant elevations in CNP transcript levels that were correlated with the degree of LPC-induced membrane distortion, and that were abolished by stabilizing cell membranes with cholesterol:methyl-beta cyclodextrin complexes or by chelating intracellular Ca2+ with BAPTA/AM. CNP up-regulation in HAoSMC by both LPC and lysophosphatidic acid was dependent on intact tyrosine kinase and protein kinase C (PKC)-signaling, whereas it was independent of these pathways in endothelial cells. LPC-induced up-regulation of CNP mRNA in human VSMC results from membrane damage and Ca2+ influx and subsequent tyrosine kinase and PKC signaling, suggesting a means by which vascular damage by ox-LDL and LPC could initiate CNP-mediated vasoprotection.


Asunto(s)
Membrana Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Lisofosfatidilcolinas/farmacología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Péptido Natriurético Tipo-C/biosíntesis , Membrana Celular/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Péptido Natriurético Tipo-C/agonistas
20.
Amino Acids ; 34(4): 683-5, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18060527

RESUMEN

The proliferation of mesangial cells (MC) in the presence of glutamine (0-20 mM) was determined in both low (5 mM) and high (25 mM) glucose-containing medium. Glutamine in a high glucose (HG) environment increased cell proliferation in a dose-dependent manner. Inhibition of glutamine:fructose 6-phosphate amidotransferase (GFAT) and of phosphodiesterase significantly reduced glutamine-induced proliferation. Supraphysiologic levels of glutamine increase MC proliferation in a HG milieu via GFAT and cAMP-dependent pathways, suggesting that glutamine could pose a risk for diabetic nephropathy.


Asunto(s)
Glucosa/farmacología , Glutamina/farmacología , Células Mesangiales/citología , Células Mesangiales/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/antagonistas & inhibidores , Humanos , Hidrolasas Diéster Fosfóricas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA