Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Phytomedicine ; 108: 154495, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36257219

RESUMEN

BACKGROUND: Zhen Wu Decoction (ZWD) is a prescription from the classical text "Treatise on Exogenous Febrile Disease" and has been extensively used to control kidney diseases since the time of the Eastern Han Dynasty. HYPOTHESIS: We hypothesized that ZWD limits tubular fibrogenesis by reinvigorating tubular bio-energetic capacity. STUDY DESIGN / METHODS: A mouse model of chronic kidney disease (CKD) was established using unilateral ureteral obstruction (UUO). Three concentrations of ZWD, namely 25.2 g/kg (high dosage), 12.6 g/kg (middle dosage), and 6.3 g/kg (low dosage), were included to study the dose-effect relationship. Real-time qPCR was used to observe gene transcription in blood samples from patients with CKD. Different siRNAs were designed to study the role of mitochondrial transcription factor A (TFAM) and nuclear factor (erythroid-derived 2)-related factor 2 (NRF2) in transforming growth factor (TGF)-ß1 induced fibrogenesis and mitochondrial damage. RESULTS: We showed that ZWD efficiently attenuates renal function impairment and reduces renal interstitial fibrosis. TFAM and NRF2 were repressed, and the stimulator of interferon genes (STING) was activated in CKD patient blood sample. We further confirmed that ZWD activated TFAM depended on NRF2 as an important negative regulator of STING in mouse kidneys. Treatment with ZWD significantly reduced oxidative stress and inflammation by regulating the levels of oxidative phosphorylation (OXPHOS) and pro-inflammatory factors, such as interleukin-6, interleukin-1ß, tumor necrosis factor receptor 1, and mitochondrial respiratory chain subunits. NRF2 inhibitors can weaken the ability of ZWD to increase TFAM expression and heal injured mitochondria, playing a similar role to that of STING inhibitors. Our study showed that ZWD elevates the expression of TFAM and mitochondrial respiratory chain subunits by promoting NRF2 activation, after suppressing mitochondrial membrane damage and cristae breakdown and restricting mitochondrial DNA (mtDNA) leakage into the cytoplasm to reduce STING activation. CONCLUSION: ZWD maintains mitochondrial integrity and improves OXPHOS which represents an innovative insight into "strengthening Yang-Qi" theory. ZWD limits tubular fibrogenesis by reinvigorating tubular bioenergetic capacity.


Asunto(s)
Proteínas de Unión al ADN , Medicamentos Herbarios Chinos , Proteínas del Grupo de Alta Movilidad , Factor 2 Relacionado con NF-E2 , Insuficiencia Renal Crónica , Obstrucción Ureteral , Animales , Ratones , ADN Mitocondrial/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Metabolismo Energético , Fibrosis , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Riñón , Mitocondrias/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Insuficiencia Renal Crónica/tratamiento farmacológico , Obstrucción Ureteral/patología , Medicamentos Herbarios Chinos/farmacología
2.
Theranostics ; 11(14): 6983-7004, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093866

RESUMEN

Rationale: We recently demonstrated that the 'Metabesity' factor HMG20A regulates islet beta-cell functional maturity and adaptation to physiological stress such as pregnancy and pre-diabetes. HMG20A also dictates central nervous system (CNS) development via inhibition of the LSD1-CoREST complex but its expression pattern and function in adult brain remains unknown. Herein we sought to determine whether HMG20A is expressed in the adult CNS, specifically in hypothalamic astrocytes that are key in glucose homeostasis and whether similar to islets, HMG20A potentiates astrocyte function in response to environmental cues. Methods: HMG20A expression profile was assessed by quantitative PCR (QT-PCR), Western blotting and/or immunofluorescence in: 1) the hypothalamus of mice exposed or not to either a high-fat diet or a high-fat high-sucrose regimen, 2) human blood leukocytes and adipose tissue obtained from healthy or diabetic individuals and 3) primary mouse hypothalamic astrocytes exposed to either high glucose or palmitate. RNA-seq and cell metabolic parameters were performed on astrocytes treated or not with a siHMG20A. Astrocyte-mediated neuronal survival was evaluated using conditioned media from siHMG20A-treated astrocytes. The impact of ORY1001, an inhibitor of the LSD1-CoREST complex, on HMG20A expression, reactive astrogliosis and glucose metabolism was evaluated in vitro and in vivo in high-fat high-sucrose fed mice. Results: We show that Hmg20a is predominantly expressed in hypothalamic astrocytes, the main nutrient-sensing cell type of the brain. HMG20A expression was upregulated in diet-induced obesity and glucose intolerant mice, correlating with increased transcript levels of Gfap and Il1b indicative of inflammation and reactive astrogliosis. Hmg20a transcript levels were also increased in adipose tissue of obese non-diabetic individuals as compared to obese diabetic patients. HMG20A silencing in astrocytes resulted in repression of inflammatory, cholesterol biogenesis and epithelial-to-mesenchymal transition pathways which are hallmarks of reactive astrogliosis. Accordingly, HMG20A depleted astrocytes exhibited reduced mitochondrial bioenergetics and increased susceptibility to apoptosis. Neuron viability was also hindered in HMG20A-depleted astrocyte-derived conditioned media. ORY1001 treatment rescued expression of reactive astrogliosis-linked genes in HMG20A ablated astrocytes while enhancing cell surface area, GFAP intensity and STAT3 expression in healthy astrocytes, mimicking the effect of HMG20A. Furthermore, ORY1001 treatment protected against obesity-associated glucose intolerance in mice correlating with a regression of hypothalamic HMG20A expression, indicative of reactive astrogliosis attenuation with improved health status. Conclusion: HMG20A coordinates the astrocyte polarization state. Under physiological pressure such as obesity and insulin resistance that induces low grade inflammation, HMG20A expression is increased to induce reactive astrogliosis in an attempt to preserve the neuronal network and re-establish glucose homeostasis. Nonetheless, a chronic metabesity state or functional mutations will result in lower levels of HMG20A, failure to promote reactive astrogliosis and increase susceptibility of neurons to stress-induced apoptosis. Such effects could be reversed by ORY1001 treatment both in vitro and in vivo, paving the way for a new therapeutic approach for Type 2 Diabetes Mellitus.


Asunto(s)
Astrocitos/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Gliosis/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Adulto , Animales , Supervivencia Celular/efectos de los fármacos , Proteínas Co-Represoras/antagonistas & inhibidores , Dieta Alta en Grasa , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucosa/metabolismo , Proteínas del Grupo de Alta Movilidad/antagonistas & inhibidores , Proteínas del Grupo de Alta Movilidad/genética , Histona Demetilasas/antagonistas & inhibidores , Humanos , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , ARN Interferente Pequeño , RNA-Seq
3.
J Agric Food Chem ; 68(44): 12284-12294, 2020 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-33094608

RESUMEN

The initiation and development of alcoholic liver disease (ALD) is mediated, at least partly, by mitochondria dysfunction, which is regulated by PPARγ coactivator-1α (PGC-1α) via mitochondria transcription factor A (TFAM). Then, PGC-1α expression was regulated by several microRNAs. This research investigated the hepatoprotective effects of the rice bran phenolic extract (RBPE) on mice fed with an ethanol-containing diet via the microRNAs-PGC-1α-TFAM signal pathway. RBPE treatment protected against alcoholic liver injury, as indicated by decreased serum aminotransferase activities and hepatic triglyceride accumulation, together with alleviated oxidative stress in serum and the liver. RBPE treatment alleviated ethanol-induced mitochondrial dysfunction through altering the membrane potential, mtDNA content, and respiratory chain complex enzyme activities in mitochondria, resulting in increased hepatic ATP production. Decreased cytoplasmic cytochrome c contents, caspase-3 activity, and Bax/Bcl-2 ratio were detected in the liver of RBPE-treated mice, indicating that the RBPE might inhibit ethanol-induced hepatocellular apoptosis. Furthermore, ethanol-induced decreases in the mRNA and protein expression of PGC-1α and TFAM were remarkably alleviated in RBPE-treated mice. RBPE treatment to ethanol-fed mice could also downregulate the expression of microRNA-494-3p, which regulates PGC-1α expression directly. Therefore, the RBPE might exert protection against ALD by alleviating mitochondrial dysfunction and the resulting hepatocyte apoptosis via the PGC-1α-TFAM signal pathway mediated by microRNA-494-3p.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Hepatopatías Alcohólicas/prevención & control , MicroARNs/metabolismo , Mitocondrias/efectos de los fármacos , Oryza/química , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fenoles/administración & dosificación , Extractos Vegetales/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Sustancias Protectoras/administración & dosificación , Semillas/química
4.
Molecules ; 25(4)2020 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-32079067

RESUMEN

Red ginseng has been reported to elicit various therapeutic effects relevant to cancer, diabetes, neurodegenerative diseases, and inflammatory diseases. However, the effect of red ginseng on exercise endurance and skeletal muscle function remains unclear. Herein, we sought to investigate whether red ginseng could affect exercise endurance and examined its molecular mechanism. Mice were fed with red ginseng extract (RG) and undertook swimming exercises to determine the time to exhaustion. Animals fed with RG had significantly longer swimming endurance. RG treatment was also observed to enhance ATP production levels in myoblasts. RG increased mRNA expressions of mitochondrial biogenesis regulators, NRF-1, TFAM, and PGC-1α, which was accompanied by an elevation in mitochondrial DNA, suggesting an enhancement in mitochondrial energy-generating capacity. Importantly, RG treatment induced phosphorylation of p38 and AMPK and upregulated PGC1α expression in both myoblasts and in vivo muscle tissue. In addition, RG treatment also stimulated C2C12 myogenic differentiation. Our findings show that red ginseng improves exercise endurance, suggesting that it may have applications in supporting skeletal muscle function and exercise performance.


Asunto(s)
Mitocondrias/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Mioblastos/efectos de los fármacos , Panax/química , Resistencia Física/efectos de los fármacos , Extractos Vegetales/farmacología , Adenosina Trifosfato/biosíntesis , Animales , Diferenciación Celular/efectos de los fármacos , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Ratones , Ratones Endogámicos ICR , Mitocondrias/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Factor Nuclear 1 de Respiración/genética , Factor Nuclear 1 de Respiración/metabolismo , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Condicionamiento Físico Animal , Resistencia Física/fisiología , Extractos Vegetales/aislamiento & purificación , Natación/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
J Cell Mol Med ; 24(3): 2229-2239, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31960591

RESUMEN

Blockade of cell cycle re-entry in quiescent cancer cells is a strategy to prevent cancer progression and recurrence. We investigated the action and mode of action of CPF mixture (Coptis chinensis, Pinellia ternata and Fructus trichosanthis) in impeding a proliferative switch in quiescent lung cancer cells. The results indicated that CPF impeded cell cycle re-entry in quiescent lung cancer cells by reduction of FACT and c-MYC mRNA and protein levels, with concomitant decrease in H3K4 tri-methylation and RNA polymerase II occupancy at FACT and c-MYC promoter regions. Animals implanted with quiescent cancer cells that had been exposed to CPF had reduced tumour volume/weight. Thus, CPF suppresses proliferative switching through transcriptional suppression of FACT and the c-MYC, providing a new insight into therapeutic target and intervention method in impeding cancer recurrence.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas del Grupo de Alta Movilidad/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-myc/genética , Transcripción Genética/efectos de los fármacos , Factores de Elongación Transcripcional/genética , Células A549 , Animales , Araceae/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/genética , Ranunculaceae/química , Trichosanthes/química
6.
Epigenomics ; 8(12): 1689-1708, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27855486

RESUMEN

Dysregulation of histone methylation has emerged as a major driver of neurodevelopmental disorders including intellectual disabilities and autism spectrum disorders. Histone methyl writer and eraser enzymes generally act within multisubunit complexes rather than in isolation. However, it remains largely elusive how such complexes cooperate to achieve the precise spatiotemporal gene expression in the developing brain. Histone H3K4 methylation (H3K4me) is a chromatin signature associated with active gene-regulatory elements. We review a body of literature that supports a model in which the RAI1-containing H3K4me writer complex counterbalances the LSD1-containing H3K4me eraser complex to ensure normal brain development. This model predicts H3K4me as the nexus of previously unrelated neurodevelopmental disorders.


Asunto(s)
Encéfalo/metabolismo , Histonas/metabolismo , Anomalías Múltiples/genética , Animales , Trastornos de los Cromosomas/genética , Duplicación Cromosómica/genética , Ritmo Circadiano/genética , Proteínas Co-Represoras/genética , Expresión Génica , Proteínas del Grupo de Alta Movilidad/genética , Histona Demetilasas/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Metilación , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas del Tejido Nervioso/genética , Síndrome de Smith-Magenis/genética , Transactivadores , Factores de Transcripción/genética
7.
J Nutr Biochem ; 26(11): 1193-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26166229

RESUMEN

Mitochondrial dysfunction in muscles leads to a wide range of metabolic and age-related disorders. Recently, it has been reported that a natural polyphenol, resveratrol, affects mitochondrial biogenesis. This study aimed to identify other natural polyphenolic compounds that regulate mitochondrial biogenesis in muscles. For this purpose, we used the C2C12 murine muscle cell line. Screening involved a reporter assay based on the promoter of mitochondrial transcription factor A (Tfam). We found that several polyphenols exhibited the ability to increase Tfam promoter activity and that the soy isoflavone daidzein was a most potent candidate that regulated mitochondrial biogenesis. When C2C12 myotubes were treated with 25-50 µM daidzein for 24h, there were significant increases in the expression of Tfam and mitochondrial genes such as COX1 and Cytb as well as the mitochondrial content. Using several mutant Tfam promoter fragments, we found that the transcription factor, nuclear respiratory factor (NRF) and its coactivator, PGC1α, were necessary for the effect of daidzein on Tfam expression. Finally, silencing of sirtuin-1 (SIRT1) by shRNA resulted in inhibition of the daidzein effects on mitochondrial gene expression. In conclusion, daidzein regulates mitochondrial biogenesis in muscle cells by regulating transcriptional networks through a SIRT1-associated pathway. These results suggest that daidzein would be beneficial to protect against a wide range of diseases caused by muscle mitochondrial dysfunction.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas del Grupo de Alta Movilidad/genética , Isoflavonas/farmacología , Mitocondrias Musculares/efectos de los fármacos , Mioblastos/efectos de los fármacos , Animales , Sitios de Unión , Línea Celular , Suplementos Dietéticos , Evaluación Preclínica de Medicamentos/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Mitocondrias Musculares/genética , Mitocondrias Musculares/metabolismo , Mioblastos/metabolismo , Factor 1 Relacionado con NF-E2/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Polifenoles/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Sirtuina 1/genética , Sirtuina 1/metabolismo
8.
J Appl Physiol (1985) ; 116(8): 1078-87, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24610533

RESUMEN

While exercise benefits have been well documented in patients with chronic diseases, the mechanistic understanding of cachectic muscle's response to contraction is essentially unknown. We previously demonstrated that treadmill exercise training attenuates the initiation of cancer cachexia and the development of metabolic syndrome symptoms (Puppa MJ, White JP, Velazquez KT, Baltgalvis KA, Sato S, Baynes JW, Carson JA. J Cachexia Sarcopenia Muscle 3: 117-137, 2012). However, cachectic muscle's metabolic signaling response to a novel, acute bout of low-frequency contraction has not been determined. The purpose of this study was to determine whether severe cancer cachexia disrupts the acute contraction-induced response to low-frequency muscle contraction [low-frequency stimulation (LoFS)]. Metabolic gene expression and signaling was examined 3 h after a novel 30-min bout of contraction (10 Hz) in cachectic Apc(Min/+) (Min) and C57BL/6 (BL-6) mice. Pyrrolidine dithiocarbamate, a STAT/NF-κB inhibitor and free radical scavenger, was administered systemically to a subset of mice to determine whether this altered the muscle contraction response. Although glucose transporter-4 mRNA was decreased by cachexia, LoFS increased muscle glucose transporter-4 mRNA in both BL-6 and Min mice. LoFS also induced muscle peroxisome proliferator-activated receptor-γ and peroxisome proliferator-activated receptor-α coactivator-1 mRNA. However, in Min mice, LoFS was not able to induce muscle proliferator-activated receptor-α coactivator-1 targets nuclear respiratory factor-1 and mitochondrial transcription factor A mRNA. LoFS induced phosphorylated-S6 in BL-6 mice, but this induction was blocked by cachexia. Administration of pyrrolidine dithiocarbamate for 24 h rescued LoFS-induced phosphorylated-S6 in cachectic muscle. LoFS increased muscle phosphorylated-AMP-activated protein kinase and p38 in BL-6 and Min mice. These data demonstrate that cachexia alters the muscle metabolic response to acute LoFS, and combination therapies in concert with muscle contraction may be beneficial for improving muscle mass and function during cachexia.


Asunto(s)
Caquexia/fisiopatología , Caquexia/terapia , Terapia por Estimulación Eléctrica/métodos , Músculo Esquelético/fisiopatología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Caquexia/genética , Citocromos c/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Genes APC , Fuerza de la Mano/fisiología , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Contracción Muscular/fisiología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Factor Nuclear 1 de Respiración/genética , Factor Nuclear 1 de Respiración/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Pirrolidinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Tiocarbamatos/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Cardiovasc Res ; 103(1): 100-10, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24675724

RESUMEN

AIM: MicroRNAs (miRNAs) play key roles in inflammatory responses of macrophages. However, the function of miRNAs in macrophage-derived foam cell formation is unclear. Here, we investigated the role of miRNAs in macrophage-derived foam cell formation and atherosclerotic development. METHODS AND RESULTS: Using quantitative reverse transcription-PCR (qRT-PCR), we found that the level of miR-155 expression was increased significantly in both plasma and macrophages from atherosclerosis (ApoE(-/-)) mice. We identified that oxidized low density lipoprotein (oxLDL) induced the expression and release of miR-155 in macrophages, and that miR-155 was required to mediate oxLDL-induced lipid uptake and reactive oxygen species (ROS) production of macrophages. Furthermore, ectopic overexpression and knockdown experiments identified that HMG box-transcription protein1 (HBP1) is a novel target of miR-155. Knockdown of HBP1 enhanced lipid uptake and ROS production in oxLDL-stimulated macrophages, and overexpression of HBP1 repressed these effects. Furthermore, bioinformatics analysis identified three YY1 binding sites in the promoter region of pri-miR-155 and verified YY1 binding directly to its promoter region. Detailed analysis showed that the YY1/HDAC2/4 complex negatively regulated the expression of miR-155 to suppress oxLDL-induced foam cell formation. Importantly, inhibition of miR-155 by a systemically delivered antagomiR-155 decreased clearly lipid-loading in macrophages and reduced atherosclerotic plaques in ApoE(-/-) mice. Moreover, we observed that the level of miR-155 expression was up-regulated in CD14(+) monocytes from patients with coronary heart disease. CONCLUSION: Our findings reveal a new regulatory pathway of YY1/HDACs/miR-155/HBP1 in macrophage-derived foam cell formation during early atherogenesis and suggest that miR-155 is a potential therapeutic target for atherosclerosis.


Asunto(s)
Aterosclerosis/etiología , Células Espumosas/metabolismo , Células Espumosas/patología , Proteínas del Grupo de Alta Movilidad/antagonistas & inhibidores , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Sitios de Unión/genética , Estudios de Casos y Controles , Enfermedad Coronaria/genética , Enfermedad Coronaria/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Histona Desacetilasa 2/metabolismo , Histona Desacetilasas/metabolismo , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Ratones , Ratones Noqueados , Monocitos/metabolismo , Regiones Promotoras Genéticas , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción YY1/metabolismo
10.
Apoptosis ; 19(4): 603-14, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24337820

RESUMEN

Coenzyme Q10 (CoQ10) acts by scavenging reactive oxygen species for protecting neuronal cells against oxidative stress in neurodegenerative diseases. We tested whether a diet supplemented with CoQ10 ameliorates oxidative stress and mitochondrial alteration, as well as promotes retinal ganglion cell (RGC) survival in ischemic retina induced by intraocular pressure elevation. A CoQ10 significantly promoted RGC survival at 2 weeks after ischemia. Superoxide dismutase 2 (SOD2) and heme oxygenase-1 (HO-1) expression were significantly increased at 12 h after ischemic injury. In contrast, the CoQ10 significantly prevented the upregulation of SOD2 and HO-1 protein expression in ischemic retina. In addition, the CoQ10 significantly blocked activation of astroglial and microglial cells in ischemic retina. Interestingly, the CoQ10 blocked apoptosis by decreasing caspase-3 protein expression in ischemic retina. Bax and phosphorylated Bad (pBad) protein expression were significantly increased in ischemic retina at 12 h. Interestingly, while CoQ10 significantly decreased Bax protein expression in ischemic retina, CoQ10 showed greater increase of pBad protein expression. Of interest, ischemic injury significantly increased mitochondrial transcription factor A (Tfam) protein expression in the retina at 12 h, however, CoQ10 significantly preserved Tfam protein expression in ischemic retina. Interestingly, there were no differences in mitochondrial DNA content among control- or CoQ10-treated groups. Our findings demonstrate that CoQ10 protects RGCs against oxidative stress by modulating the Bax/Bad-mediated mitochondrial apoptotic pathway as well as prevents mitochondrial alteration by preserving Tfam protein expression in ischemic retina. Our results suggest that CoQ10 may provide neuroprotection against oxidative stress-mediated mitochondrial alterations in ischemic retinal injury.


Asunto(s)
Isquemia/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Retina/metabolismo , Ubiquinona/análogos & derivados , Vitaminas/farmacología , Animales , Apoptosis/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dieta , Femenino , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Presión Intraocular/efectos de los fármacos , Isquemia/metabolismo , Isquemia/patología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Fosforilación , Retina/efectos de los fármacos , Retina/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Ubiquinona/farmacología , Ubiquinona/uso terapéutico , Vitaminas/uso terapéutico , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proteína Letal Asociada a bcl/metabolismo
11.
Oxid Med Cell Longev ; 2012: 835970, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22655115

RESUMEN

Skeletal muscle function largely depend on intact energy metabolism, stress response, and antioxidant defense mechanisms. In this study, we tested the effect of a combined supplementation of α-lipoic acid (LA) plus coenzyme Q10 (Q10) on PPARγ-coactivator α (PGC1α) activity, expression of glutathione-related phase II enzymes and glutathione (GSH) levels in cultured C2C12 myotubes. Supplementation of myotubes with 250 µmol/L LA plus 100 µmol/L Q10 significantly increased nuclear levels of PGC1α, a master switch of energy metabolism and mitochondrial biogenesis. The increase of nuclear PGC1α was accompanied by an increase in PPARγ transactivation, a downstream target of PGC1α, and an increase in mitochondrial transcription factor A mRNA centrally involved in mitochondrial replication and transcription. Furthermore, supplementation of myotubes with LA plus Q10 resulted in an increase of genes encoding proteins involved in stress response, GSH synthesis, and its recycling. In LA-plus-Q10-treated myotubes a significant 4-fold increase in GSH was evident. This increase in GSH was accompanied by increased nuclear Nrf2 protein levels, partly regulating γGCS and GST gene expression. Present data suggest that the combined supplementation of skeletal muscle cells with LA plus Q10 may improve energy homeostasis, stress response, and antioxidant defense mechanisms.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Glutatión/metabolismo , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Estrés Fisiológico/efectos de los fármacos , Ácido Tióctico/farmacología , Transactivadores/metabolismo , Ubiquinona/análogos & derivados , Animales , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Ratones , Modelos Biológicos , Células Musculares/citología , Células Musculares/efectos de los fármacos , Músculo Esquelético/citología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estrés Fisiológico/genética , Ácido Tióctico/administración & dosificación , Factores de Transcripción , Ubiquinona/administración & dosificación , Ubiquinona/farmacología
12.
PLoS One ; 7(2): e32305, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22384209

RESUMEN

Plant pathogenic bacteria utilize an array of effector proteins to cause disease. Among them, transcriptional activator-like (TAL) effectors are unusual in the sense that they modulate transcription in the host. Although target genes and DNA specificity of TAL effectors have been elucidated, how TAL proteins control host transcription is poorly understood. Previously, we showed that the Xanthomonas citri TAL effectors, PthAs 2 and 3, preferentially targeted a citrus protein complex associated with transcription control and DNA repair. To extend our knowledge on the mode of action of PthAs, we have identified new protein targets of the PthA4 variant, required to elicit canker on citrus. Here we show that all the PthA4-interacting proteins are DNA and/or RNA-binding factors implicated in chromatin remodeling and repair, gene regulation and mRNA stabilization/modification. The majority of these proteins, including a structural maintenance of chromosomes protein (CsSMC), a translin-associated factor X (CsTRAX), a VirE2-interacting protein (CsVIP2), a high mobility group (CsHMG) and two poly(A)-binding proteins (CsPABP1 and 2), interacted with each other, suggesting that they assemble into a multiprotein complex. CsHMG was shown to bind DNA and to interact with the invariable leucine-rich repeat region of PthAs. Surprisingly, both CsHMG and PthA4 interacted with PABP1 and 2 and showed selective binding to poly(U) RNA, a property that is novel among HMGs and TAL effectors. Given that homologs of CsHMG, CsPABP1, CsPABP2, CsSMC and CsTRAX in other organisms assemble into protein complexes to regulate mRNA stability and translation, we suggest a novel role of TAL effectors in mRNA processing and translational control.


Asunto(s)
Proteínas Bacterianas/fisiología , Poli U/química , ARN/química , Xanthomonas/metabolismo , Arabidopsis/metabolismo , Proteínas Bacterianas/química , Cromosomas/ultraestructura , Citrus , Reparación del ADN , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/genética , Sistemas de Lectura Abierta , Extractos Vegetales/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Efectores Tipo Activadores de la Transcripción , Transcripción Genética , Técnicas del Sistema de Dos Híbridos
13.
Mol Cancer ; 10: 74, 2011 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-21679440

RESUMEN

BACKGROUND: Platinum-containing chemotherapy produces specific DNA damage and is used to treat several human solid tumors. Tumors initially sensitive to platinum-based drugs frequently become resistant. Inhibition of DNA repair is a potential strategy to enhance cisplatin effectiveness. After cisplatin treatment, a balance between repair and apoptosis determines whether cancer cells proliferate or die. DNA-dependent protein kinase (DNA-PK) binds to DNA double strand breaks (DSBs) through its Ku subunits and initiates non-homologous end joining. Inhibition of DNA-PK sensitizes cancer cells to cisplatin killing. The goal of this study is to elucidate the mechanism underlying the effects of DNA-PK on cisplatin sensitivity. RESULTS: Silencing the expression of the catalytic subunit of DNA-PK (DNA-PKcs) increased sensitivity to cisplatin and decreased the appearance of γH2AX after cisplatin treatment. We purified DNA-PK by its Ku86 subunit and identified interactors by tandem mass spectrometry before and after cisplatin treatment. The structure specific recognition protein 1 (SSRP1), Spt16 and γH2AX appeared in the Ku86 complex 5 hours after cisplatin treatment. SSRP1 and Spt16 form the facilitator of chromatin transcription (FACT). The cisplatin-induced association of FACT with Ku86 and γH2AX was abrogated by DNase treatment. In living cells, SSRP1 and Ku86 were recruited at sites of DSBs induced by laser beams. Silencing SSRP1 expression increased sensitivity to cisplatin and decreased γH2AX appearance. However, while silencing SSRP1 in cisplatin-treated cells increased both apoptosis and necrosis, DNA-PKcs silencing, in contrast, favored necrosis over apoptosis. CONCLUSIONS: DNA-PK and FACT both play roles in DNA repair. Therefore both are putative targets for therapeutic inhibition. Since DNA-PK regulates apoptosis, silencing DNA-PKcs redirects cells treated with cisplatin toward necrosis. Silencing FACT however, allows both apoptosis and necrosis. Targeting DNA repair in cancer patients may have different therapeutic effects depending upon the roles played by factors targeted.


Asunto(s)
Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Reparación del ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas del Grupo de Alta Movilidad/fisiología , Factores de Elongación Transcripcional/fisiología , Antineoplásicos/farmacología , Apoptosis/genética , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Reparación del ADN/genética , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Evaluación Preclínica de Medicamentos , Células HEK293 , Células HeLa , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Necrosis/inducido químicamente , Necrosis/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
14.
Dev Biol ; 315(1): 18-27, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18206866

RESUMEN

The epicardium is the last layer of the vertebrate heart to form, surrounding the heart muscle during embryogenesis and providing signaling cues essential to the continued growth and differentiation of the heart. This outer layer of the heart develops from a transient structure, the proepicardial organ (PEO). Despite its essential roles, the early signals required for the formation of the PEO and the epicardium remain poorly understood. The molecular markers wt1 and tcf21 are used to identify the epicardial layer in the zebrafish heart, to trace its development and to determine genes required for its normal development. Disruption of lateral plate mesoderm (LPM) migration through knockdown of miles apart or casanova leads to cardia bifida with each bilateral heart associated with its own PEO, suggesting that the earliest progenitors of the epicardium lie in the LPM. Using a gene knockdown approach, a genetic framework for PEO development is outlined. The pandora/spt6 gene is required for multiple cardiac lineages, the zinc-finger transcription factor wt1 is required for the epicardial lineage only and finally, the cell polarity genes heart and soul and nagie oko are required for proper PEO morphogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Organogénesis , Pericardio/embriología , Proteínas de Pez Cebra/metabolismo , Animales , Biomarcadores , Linaje de la Célula , Polaridad Celular/genética , ADN Complementario , Embrión no Mamífero , Femenino , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Hibridación in Situ , Isoenzimas/genética , Isoenzimas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Microinyecciones , Modelos Biológicos , Mutación , Miocardio/metabolismo , Oligonucleótidos Antisentido/farmacología , Pericardio/citología , Pericardio/metabolismo , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Factores de Transcripción SOX , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética
15.
Methods Mol Biol ; 329: 353-69, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16846004

RESUMEN

The process of bringing a new pharmacologically active drug to market is laborious, time consuming, and costly. From drug discovery to safety assessment, new methods are constantly sought to develop faster and more efficient procedures to eliminate drugs from further investigation because of their limited effectiveness or high toxicity. Because in vitro cell assays are an important arm of this discovery process, it is therefore somewhat unsurprising that there is an emerging contribution of embryonic stem (ES) cell technology to this area. This technology utilizes the in vitro differentiation of ES cells into somatic cell target populations that, when coupled to the use of "lineage selection" protocols, allows for the production of infinite numbers of pure populations of the desired cells for both bioactivity and toxicological screens. Unlike the use of transformed cell lines, ES-derived cells remain karyotypically normal and therefore better reflect the potential responses of cells in vivo, and when selected are more homogeneous than those obtained using primary cultures. In this chapter we discuss the use of ES cell-derived somatic cells in pharmacological screens, with particular emphasis on neural cells, and describe the methods and protocols associated with the development of ES cell-derived neural cell assays.


Asunto(s)
Embrión de Mamíferos/citología , Neuronas/citología , Neuronas/efectos de los fármacos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Medios de Cultivo , Proteínas de Unión al ADN/genética , Evaluación Preclínica de Medicamentos/métodos , Marcación de Gen , Genes Reporteros , Vectores Genéticos , Proteínas del Grupo de Alta Movilidad/genética , Ratones , Plásmidos/genética , Factores de Transcripción SOXB1
16.
Biotechnol Bioeng ; 93(6): 1152-63, 2006 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-16470881

RESUMEN

Bone marrow mesenchymal stem cells (MSCs) are candidate cells for cartilage tissue engineering. This is due to their ability to undergo chondrogenic differentiation after extensive expansion in vitro and stimulation with various biomaterials in three-dimensional (3-D) systems. Collagen type II is one of the major components of the hyaline cartilage and plays a key role in maintaining chondrocyte function. This study aimed at analyzing the MSC chondrogenic response during culture in different types of extracellular matrix (ECM) with a focus on the influence of collagen type II on MSC chondrogenesis. Bovine MSCs were cultured in monolayer as well as in alginate and collagen type I and II hydrogels, in both serum free medium and medium supplemented with transforming growth factor (TGF) beta1. Chondrogenic differentiation was detected after 3 days of culture in 3-D hydrogels, by examining the presence of glycosaminoglycan and newly synthesized collagen type II in the ECM. Differentiation was most prominent in cells cultured in collagen type II hydrogel, and it increased in a time-dependent manner. The expression levels of the of chondrocyte specific genes: sox9, collagen type II, aggrecan, and COMP were measured by quantitative "Real Time" RT-PCR, and genes distribution in the hydrogel beads were localized by in situ hybridization. All genes were upregulated by the presence of collagen, particularly type II, in the ECM. Additionally, the chondrogenic influence of TGF beta1 on MSCs cultured in collagen-incorporated ECM was analyzed. TGF beta1 and dexamethasone treatment in the presence of collagen type II provided more favorable conditions for expression of the chondrogenic phenotype. In this study, we demonstrated that collagen type II alone has the potential to induce and maintain MSC chondrogenesis, and prior interaction with TGF beta1 to enhance the differentiation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Condrocitos/metabolismo , Hidrogeles/farmacología , Células Madre Mesenquimatosas/citología , Agrecanos , Alginatos/farmacología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Bovinos , Diferenciación Celular/fisiología , Proliferación Celular/efectos de los fármacos , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Condrogénesis/fisiología , Proteoglicanos Tipo Condroitín Sulfato/genética , Colágeno Tipo I/genética , Colágeno Tipo I/farmacología , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Colágeno Tipo II/farmacología , Colágeno Tipo X/genética , Medio de Cultivo Libre de Suero/farmacología , Matriz Extracelular/fisiología , Proteínas de la Matriz Extracelular/genética , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Ácido Glucurónico/farmacología , Glicoproteínas/genética , Ácidos Hexurónicos/farmacología , Proteínas del Grupo de Alta Movilidad/genética , Hibridación in Situ , Lectinas Tipo C/genética , Proteínas Matrilinas , Células Madre Mesenquimatosas/efectos de los fármacos , Proteínas/farmacología , Factor de Transcripción SOX9 , Ingeniería de Tejidos/métodos , Factores de Transcripción/genética
17.
J Nutr Biochem ; 17(6): 385-95, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16311027

RESUMEN

Dietary phosphorus (P) is essential to bone growth and turnover; however, little research has focused on the genetic mechanisms controlling P utilization. Understanding the interactions between genetics and dietary P that optimize bone integrity could provide novel interventions for osteoporosis. Thirty-six pigs from two sire lines known to differ in bone structure [heavier boned (HB) and lighter boned (LB)] were assigned to one of the three diets (P adequate, P repletion or P deficient). After 14 days, bone marrow and intact radial bones were collected. Differences between these lines in growth rate, bone integrity and gene expression within bone marrow were observed. In HB, but not LB, pigs, the P-deficient diet decreased weight gain (P<.01). For both lines, P deficiency caused a reduction in radial bone strength (P<.01), but HB P-deficient animals had greater (P<.10) bone integrity than P-deficient LB pigs. In HB, but not LB, pigs, dietary treatment affected the expression of CALCR (calcitonin receptor) (P<.05), VDR (vitamin D receptor) (P<.04) and IGFBP3 (insulin-like growth factor binding protein 3) (P<.06). There was also a trend of increased IL6 (interleukin-6), TFIIB (transcription initiation factor IIB) and SOX9 (sex determining region Y-box 9) expression with P deficiency in HB, but not LB, pigs. Both genetic backgrounds responded similarly to P deficiency with an increase in the expression of OXTR (oxytocin receptor) and IGF1 (insulin-like growth factor 1). Differences in growth rate, bone integrity and gene expression within the bone marrow suggest a difference in the homeorhetic control of P utilization between these genetic lines. Understanding these differences could lead to novel treatments for osteoporosis and aid in the development of tests for identifying those at risk for this disease.


Asunto(s)
Genotipo , Osteoporosis/genética , Fósforo Dietético/administración & dosificación , Fósforo/deficiencia , Animales , Huesos/fisiopatología , Calcio/sangre , Femenino , Proteínas del Grupo de Alta Movilidad/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/genética , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , Receptores de Calcitonina/genética , Receptores de Calcitriol/genética , Receptores de Oxitocina/genética , Factor de Transcripción SOX9 , Porcinos , Resistencia a la Tracción , Factor de Transcripción TFIIB/genética , Factores de Transcripción/genética
18.
Ann Med ; 38(8): 560-77, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17438671

RESUMEN

The anterior pituitary gland is a central regulator of growth, reproduction and homeostasis, and is the end-product of a carefully orchestrated pattern of expression of signalling molecules and transcription factors leading to the development of this complex organ secreting six hormones from five different cell types. Naturally occurring and transgenic murine models have demonstrated a role for many of these molecules in the aetiology of combined pituitary hormone deficiency (CPHD). These include the transcription factors HESX1, PROP1, POU1FI, LHX3, LHX4, TBX19 (TPIT), SOX3 and SOX2. The expression pattern of these transcription factors, their interaction with co-factors and their impact on target genes dictate the phenotype that results when the gene encoding the relevant transcription factor is mutated. The highly variable phenotype may consist of isolated hypopituitarism, or more complex disorders such as septo-optic dysplasia (SOD) and holoprosencephaly. Since mutations in any one transcription factor are uncommon, and since the overall incidence of mutations in known transcription factors is low in patients with CPHD, it is clear that many genes remain to be identified, and characterization of these will further elucidate the pathogenesis of these complex conditions, and also shed light on normal pituitary development.


Asunto(s)
Hipopituitarismo/congénito , Hipopituitarismo/genética , Animales , Cromosomas Humanos X/genética , Proteínas de Unión al ADN/genética , Proteínas HMGB/genética , Proteínas Hedgehog/genética , Proteínas del Grupo de Alta Movilidad/genética , Proteínas de Homeodominio/genética , Humanos , Hipotálamo/crecimiento & desarrollo , Factores de Transcripción de Tipo Kruppel/genética , Proteínas con Homeodominio LIM , Mutación , Proteínas Nucleares/genética , Adenohipófisis/crecimiento & desarrollo , Factores de Transcripción SOXB1 , Proteínas de Dominio T Box/genética , Factor de Transcripción Pit-1/genética , Factores de Transcripción/genética , Proteína Gli2 con Dedos de Zinc , Proteína del Homeodomínio PITX2
19.
Development ; 132(24): 5491-502, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16291787

RESUMEN

Gonadotropin-releasing hormone (GnRH) is found in a wide range of vertebrate tissues, including the nervous system. In general, GnRH has two functions: endocrine, acting as a releasing hormone; and neuromodulatory, affecting neural activity in the peripheral and central nervous system. The best understood population of GnRH cells is that of the hypothalamus, which is essential for reproduction. Less well understood are the populations of GnRH cells found in the terminal nerve and midbrain, which appear to be neuromodulatory in function. The GnRH-containing cells of the midbrain are proposed to arise from the mesencephalic region of the neural tube. Previously, we showed that neuromodulatory GnRH cells of the terminal nerve arise from cranial neural crest. To test the hypothesis that neuromodulatory GnRH cells of the midbrain also arise from neural crest, we used gene knockdown experiments in zebrafish to disrupt neural crest development. We demonstrate that decrement of the function of foxd3 and/or sox10, two genes important for the development and specification of neural crest, resulted in a reduction and/or loss of GnRH cells of the midbrain, as well as a reduction in the number of terminal nerve GnRH cells. Therefore, our data support a neural crest origin for midbrain GnRH cells. Additionally, we demonstrate that knockdown of kallmann gene function resulted in the loss of endocrine GnRH cells of the hypothalamus, but not of neuromodulatory GnRH cells of the midbrain and terminal nerve, thus providing additional evidence for separate pathways controlling the development of neuromodulatory and endocrine GnRH cells.


Asunto(s)
Proteínas Portadoras/fisiología , Diferenciación Celular/fisiología , Factores de Transcripción Forkhead/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Proteínas del Grupo de Alta Movilidad/fisiología , Mesencéfalo/citología , Cresta Neural/citología , Proteínas de Pez Cebra/fisiología , Pez Cebra/fisiología , Animales , Proteínas Portadoras/genética , Nervios Craneales/citología , Nervios Craneales/embriología , Embrión no Mamífero/citología , Embrión no Mamífero/fisiología , Femenino , Factores de Transcripción Forkhead/genética , Proteínas del Grupo de Alta Movilidad/genética , Hipotálamo/citología , Hipotálamo/embriología , Masculino , Mesencéfalo/embriología , Mutación , Cresta Neural/embriología , Factores de Transcripción SOXE , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
20.
PLoS Biol ; 3(12): e415, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16296893

RESUMEN

Normal aging in humans and rodents is accompanied by a progressive increase in adiposity. To investigate the role of hypothalamic neuronal circuits in this process, we used a Cre-lox strategy to create mice with specific and progressive degeneration of hypothalamic neurons that express agouti-related protein (Agrp) or proopiomelanocortin (Pomc), neuropeptides that promote positive or negative energy balance, respectively, through their opposing effects on melanocortin receptor signaling. In previous studies, Pomc mutant mice became obese, but Agrp mutant mice were surprisingly normal, suggesting potential compensation by neuronal circuits or genetic redundancy. Here we find that Pomc-ablation mice develop obesity similar to that described for Pomc knockout mice, but also exhibit defects in compensatory hyperphagia similar to what occurs during normal aging. Agrp-ablation female mice exhibit reduced adiposity with normal compensatory hyperphagia, while animals ablated for both Pomc and Agrp neurons exhibit an additive interaction phenotype. These findings provide new insight into the roles of hypothalamic neurons in energy balance regulation, and provide a model for understanding defects in human energy balance associated with neurodegeneration and aging.


Asunto(s)
Metabolismo Energético , Hipotálamo/patología , Hipotálamo/fisiopatología , Degeneración Nerviosa/fisiopatología , Proteína Relacionada con Agouti , Animales , Peso Corporal/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ingestión de Alimentos , Eliminación de Gen , Genes Reporteros/genética , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Proopiomelanocortina/deficiencia , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , ARN Mensajero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA