Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Adv Exp Med Biol ; 1074: 11-17, 2018.
Article in English | MEDLINE | ID: mdl-29721922

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of blindness in older adults in developed countries. The molecular mechanisms of disease pathogenesis remain poorly understood; however, evidence suggests that mitochondrial dysfunction may contribute to the progression of the disease. Studies have shown that mitochondrial DNA lesions are increased in the retinal pigment epithelium (RPE) of human patients with the disease and that the number of these lesions increases with disease severity. Additionally, microscopy of human RPE from patients with dry AMD shows severe disruptions in mitochondrial inner and outer membrane structure, mitochondrial size, and mitochondrial cellular organization. Thus, improving our understanding of mitochondrial dysfunction in dry AMD pathogenesis may lead to the development of targeted therapies. We propose that mitochondrial dysfunction in the RPE can lead to the chronic oxidative stress associated with the disease. Therefore, one protective strategy may involve the use of small molecule therapies that target the regulation of mitochondrial biogenesis and mitochondrial fission and mitophagy.


Subject(s)
DNA, Mitochondrial/metabolism , Macular Degeneration/metabolism , Mitochondria/pathology , Molecular Targeted Therapy , Retinal Pigment Epithelium/pathology , Adenylate Kinase/physiology , Animals , DNA, Mitochondrial/genetics , Disease Models, Animal , Drug Evaluation, Preclinical , Geographic Atrophy/pathology , Humans , Iodates/toxicity , Macular Degeneration/drug therapy , Macular Degeneration/genetics , Metformin/pharmacology , Mice , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Oxidative Stress , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism
2.
Rejuvenation Res ; 19(6): 495-508, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27072567

ABSTRACT

The YiQiFuMai powder injection (YQFM), a traditional Chinese medicine (TCM) prescription re-developed based on the well-known TCM formula Sheng-maisan, showed a wide range of pharmacological activities in cardiovascular diseases in clinics. However, its role in protection against myocardial ischemia/reperfusion (MI/R) injury has not been elucidated. The present study not only evaluated the cardioprotective effect of YQFM from MI/R injury but also investigated the potential molecular mechanisms both in vivo and in vitro. The myocardium infarct size, production of lactate dehydrogenase (LDH), creatine kinase (CK), cardiac function, TUNEL staining, and caspase-3 activity were measured. Cell viability was determined, and cell apoptosis was measured by Hoechst 33342 staining and flow cytometry. Mitochondrial membrane potential (ΔΨm) was measured, and ATP content was quantified by bioluminescent assay. Expression of apoptosis-related proteins, including Caspase-3, Bcl-2, Bax, AMPKα, and phospho-AMPKα, was analyzed by western blotting. AMPKα siRNA transfection was also applied to the mechanism elucidation. YQFM at a concentration of 1.06 g/kg significantly reduced myocardium infarct size and the production of LDH, CK in serum, improved the cardiac function, and also produced a significant decrease of apoptotic index. Further, combined treatment with compound C partly attenuated the anti-apoptotic effect of YQFM. In addition, pretreatment with YQFM ranging from 25 to 400 µg/mL markedly improved cell viability and decreased LDH release. Moreover, YQFM inhibited H9c2 apoptosis, blocked the expression of caspase-3, and modulated Bcl-2 and Bax proteins, leading to an increased mitochondrial membrane potential and cellular ATP content. Mechanistically, YQFM activated AMP-activated protein kinase (AMPK) signaling pathways whereas pretreatment with AMPK inhibitor Compound C and application of transfection with AMPKα siRNA attenuated the anti-apoptotic effect of YQFM. Our results indicated that YQFM could provide significant cardioprotection against MI/R injury, and potential mechanisms might suppress cardiomyocytes apoptosis, at least in part, through activating the AMPK signaling pathways.


Subject(s)
Adenylate Kinase/physiology , Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Myocardial Reperfusion Injury/drug therapy , Myocardium/pathology , Animals , Cells, Cultured , Enzyme Activation/drug effects , Injections , Mice , Mice, Inbred ICR , Myocardial Reperfusion Injury/pathology , Powders , Proto-Oncogene Proteins c-bcl-2/analysis , bcl-2-Associated X Protein/analysis
3.
Metab Brain Dis ; 29(1): 47-58, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24435937

ABSTRACT

Here, we have investigated the effect of metformin pretreatment in the rat models of global cerebral ischemia. Cerebral ischemia which leads to brain dysfunction is one of the main causes of neurodegeneration and death worldwide. Metformin is used in clinical drug therapy protocols of diabetes. It is suggested that metformin protects cells under hypoxia and ischemia in non-neuronal contexts. Protective effects of metformin may be modulated via activating the AMP activated protein kinase (AMPK). Our results showed that induction of 30 min global cerebral I/R injury using 4-vesseles occlusion model led to significant cell death in the rat brain. Metformin pretreatment (200 mg kg/once/day, p.o., 2 weeks) attenuated apoptotic cell death and induced mitochondrial biogenesis proteins in the ischemic rats, analyzed using histological and Western blot assays. Besides, inhibition of AMPK by compound c showed that metformin resulted in apoptosis attenuation via AMPK activation. Interestingly, AMPK activation was also involved in the induction of mitochondrial biogenesis proteins using metformin, inhibition of AMPK by compound c reversed such effect, further supporting the role of AMPK upstream of mitochondrial biogenesis proteins. In summary, Metformin pretreatment is able to modulate mitochondrial biogenesis and apoptotic cell death pathways through AMPK activation in the context of global cerebral ischemia, conducting the outcome towards neuroprotection.


Subject(s)
Adenylate Kinase/physiology , Brain Ischemia/prevention & control , Brain/drug effects , Metformin/pharmacology , Neuroprotective Agents/pharmacology , Transcription Factors/physiology , Adenylate Kinase/antagonists & inhibitors , Animals , Apoptosis/drug effects , Brain/enzymology , Brain/pathology , Brain Ischemia/pathology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Male , Metformin/administration & dosage , Metformin/therapeutic use , Mitochondrial Turnover/drug effects , NF-E2-Related Factor 1/biosynthesis , NF-E2-Related Factor 1/genetics , Neuroprotective Agents/therapeutic use , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Premedication , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Transcription Factors/biosynthesis , Transcription Factors/genetics
4.
EMBO J ; 32(6): 844-57, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23403926

ABSTRACT

pRb is frequently inactivated in tumours by mutations or phosphorylation. Here, we investigated whether pRb plays a role in obesity. The Arcuate nucleus (ARC) in hypothalamus contains antagonizing POMC and AGRP/NPY neurons for negative and positive energy balance, respectively. Various aspects of ARC neurons are affected in high-fat diet (HFD)-induced obesity mouse model. Using this model, we show that HFD, as well as pharmacological activation of AMPK, induces pRb phosphorylation and E2F target gene de-repression in ARC neurons. Some affected neurons express POMC; and deleting Rb1 in POMC neurons induces E2F target gene de-repression, cell-cycle re-entry, apoptosis, and a hyperphagia-obesity-diabetes syndrome. These defects can be corrected by combined deletion of E2f1. In contrast, deleting Rb1 in the antagonizing AGRP/NPY neurons shows no effects. Thus, pRb-E2F1 is an obesity suppression mechanism in ARC POMC neurons and HFD-AMPK inhibits this mechanism by phosphorylating pRb in this location.


Subject(s)
Diet, High-Fat , Dietary Fats/pharmacology , Hypothalamus , Obesity/genetics , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/physiology , Adenylate Kinase/metabolism , Adenylate Kinase/physiology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/physiology , Diet, High-Fat/adverse effects , Down-Regulation/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/physiology , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/pathology , Ideal Body Weight/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Obesity/metabolism , Obesity/pathology , Phosphorylation/drug effects , Pro-Opiomelanocortin/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism
5.
J Endocrinol ; 214(2): 177-89, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22582096

ABSTRACT

Adiponectin secreted from adipose tissues plays a role in the regulation of energy homeostasis, food intake, and reproduction in the hypothalamus. We have previously demonstrated that adiponectin significantly inhibited GNRH secretion from GT1-7 hypothalamic GNRH neuron cells. In this study, we further investigated the effect of adiponectin on hypothalamic KISS1 gene transcription, which is the upstream signal of GNRH. We found that globular adiponectin (gAd) or AICAR, an artificial AMPK activator, decreased KISS1 mRNA transcription and promoter activity. Conversely, inhibition of AMPK by Compound C or AMPKα1-SiRNA augmented KISS1 mRNA transcription and promoter activity. Additionally, gAd and AICAR decreased the translocation of specificity protein-1 (SP1) from cytoplasm to nucleus; however, Compound C and AMPKα1-siRNA played an inverse role. Our experiments in vivo demonstrated that the expression of Kiss1 mRNA was stimulated twofold in the Compound C-treated rats and decreased about 60-70% in gAd- or AICAR-treated rats compared with control group. The numbers of kisspeptin immunopositive neurons in the arcuate nucleus region of Sprague Dawley rats mimicked the same trend seen in Kiss1 mRNA levels in animal groups with different treatments. In conclusion, our results provide the first evidence that adiponectin reduces Kiss1 gene transcription in GT1-7 cells through activation of AMPK and subsequently decreased translocation of SP1.


Subject(s)
Adenylate Kinase/physiology , Adiponectin/pharmacology , Hypothalamus/drug effects , Kisspeptins/genetics , Neurons/drug effects , Sp1 Transcription Factor/physiology , Adenylate Kinase/metabolism , Adiponectin/physiology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Hypothalamus/cytology , Hypothalamus/metabolism , Kisspeptins/metabolism , Neurons/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Protein Transport/genetics , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology , Sp1 Transcription Factor/metabolism , Transcription, Genetic/drug effects
6.
Diabetes Res Clin Pract ; 97(3): 453-60, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22560150

ABSTRACT

AIMS: To determine whether exendin-4 might directly improve endothelial dysfunction in aorta isolated from high-fat diet-induced obese rats. METHODS: Wistar rats were randomly divided into control and obesity (OB) groups and fed. Vascular segments of obese rats were incubated in organ bath in the presence or absence of exendin-4. Nitric oxide (NO) production and nuclear transcription factor kappa B expression in vascular rings were measured. The aortic rings of the obese rats were then incubated in an organ bath with exendin-4 in the presence or absence of the following inhibitors: the AMPK, the adenylate cyclase and the NO synthase inhibitor. RESULTS: The maximum endothelium-dependent vasodilatation (EDV) value was severely reduced in the OB group. Exendin-4 treatment significantly increased the NO level, improved endothelium-dependent vasodilatation and reduced expression of NF-κB in the obese group. The beneficial effect of exendin-4 on EDV in obese rats was partly attenuated in the presence of the specific inhibitors. CONCLUSION: Exendin-4 directly improves impaired EDV of aortae from obese rats. The beneficial effect of exendin-4 appears to be mediated in part via stimulation of cAMP/AMPK-related signalling pathways and enhancement of endothelial nitric oxide synthase activity.


Subject(s)
Adenylate Kinase/metabolism , Aorta/drug effects , Cyclic AMP/metabolism , Endothelium, Vascular/drug effects , Nitric Oxide Synthase Type III/metabolism , Obesity/pathology , Peptides/pharmacology , Venoms/pharmacology , Adenylate Kinase/physiology , Animals , Aorta/pathology , Aorta/physiopathology , Cells, Cultured , Cyclic AMP/physiology , Drug Evaluation, Preclinical , Endothelium, Vascular/physiopathology , Exenatide , Hypoglycemic Agents/pharmacology , Male , Nitric Oxide Synthase Type III/physiology , Obesity/complications , Obesity/physiopathology , Organ Culture Techniques , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
7.
Regul Pept ; 173(1-3): 27-35, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-21963822

ABSTRACT

Ghrelin, released from the stomach, stimulates food intake through activation of the ghrelin receptor (GHS-R) located on neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons in the hypothalamus. A role for the energy sensor AMP-activated protein kinase (AMPK) and its downstream effector uncoupling protein 2 (UCP2) in the stimulatory effect of exogenous ghrelin on NPY/AgRP expression and food intake has been suggested. This study aimed to investigate whether a rise in endogenous ghrelin levels is able to influence hypothalamic AMPK activity, pACC, UCP2 and NPY/AgRP expression through activation of GHS-R. An increase in endogenous ghrelin levels was established by fasting (24h) or by induction of streptozotocin(STZ)-diabetes (15 days) in GHS-R(+/+) and GHS-R(-/-) mice. GHS-R(+/+) mice showed a significant increase in AgRP and NPY mRNA expression after fasting, which was not observed in GHS-R(-/-) mice. Fasting did not affect AMPK activity nor ACC phosphorylation in both genotypes and increased UCP2 mRNA expression. The hyperghrelinemia associated with STZ-induced diabetes was accompanied by an increased NPY and AgRP expression in GHS-R(+/+) but not in GHS-R(-/-) mice. AMPK activity and UCP2 expression in GHS-R(+/+) mice after induction of diabetes were decreased to a similar extent in both genotypes. Exogenous ghrelin administration tended to decrease hypothalamic AMPK activity. In conclusion, an increase in endogenous ghrelin levels triggered by fasting or STZ-induced diabetes stimulates the expression of AgRP and NPY via interaction with the GHS-R. The changes in AMPK activity, pACC and UCP2 occur independently from GHS-R suggesting that they do not play a major role in the orexigenic effect of endogenous ghrelin.


Subject(s)
Adenylate Kinase/physiology , Appetite , Ghrelin/physiology , Receptors, Ghrelin/genetics , Signal Transduction , Acetyl-CoA Carboxylase/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Adenylate Kinase/metabolism , Animals , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/metabolism , Eating , Fasting , Gene Knockout Techniques , Ghrelin/blood , Ghrelin/pharmacology , Hypothalamus/enzymology , Hypothalamus/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Male , Membrane Proteins , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Phosphorylation , Receptors, Ghrelin/metabolism , Uncoupling Protein 2
8.
An R Acad Nac Med (Madr) ; 129(2): 541-63; discussion 563, 2012.
Article in Spanish | MEDLINE | ID: mdl-24298863

ABSTRACT

Metabolic sensors play an important role in the control of food intake, utilization of nutrients and demonstration of feeding behaviour. In this work we describe the study done in our laboratory on glucokinase (GK) as brain glucose sensor, the AMP kinase (AMPK) as detector of the fall of intracellular energy charge and as the S6K in the signaling pathway of mTOR with opposite effects to AMPK. Glucose sensors are molecular designs that detect with accuracy glucose concentrations, facilitating therefore the homeostasis of this hexose. We consider GK as a component of a glucose sensor system that might modulates the feeding behaviour and indirectly the control of body weight. Our findings indicate that GK and GLUT-2 mRNAs and proteins are coexpressed mainly in areas of the hypothalamus implied in the control of food intake. We have also found a high glucose phosphorylating activity with kinetic properties similar to that reported in the liver, with a high apparent Km for glucose that displays no product inhibition by glucose-6-phosphate. GK may be also regulated by the presence of glucokinase regulatory protein (GKRP), which has been identified in the same brain areas than GK. The coexpression of these molecules might play a role as glucose sensors in which GLUT-2 has a permissive role and the interactions of GK with GKRP made possible a real sensor activity. Furthermore, the effects of anorexigenic peptides in this system should facilitate the transduction of signals required to produce a state of satiety. Thus, GLP-1 reduced significantly the glucose metabolism in areas of the hypothalamus and brainstem related with food intake, which open new ways to the study of pathophysiologicals aspects of feeding behaviour. Besides we have studied the functions of AMPK and mTOR pathway in the hypothalamic areas ventromedial (VMH) and lateral (LH) under situations with alterations of the nutritional status and energy balance. Our results revealed that the activation of AMPK and S6K in VMH y LH occur in response to the changes of glucose concentrations or in the changes in the nutritional state, as well as GLP-1/exendin-4 act by counteracting the activation/inactivation of these kinases, which support a modulating role of these peptides on the kinases. On the other hand, GLP-1/exendin-4 might contribute to the normalization of the altered values of these kinases in pathophysiological states such as obesity.


Subject(s)
Body Weight/physiology , Brain/metabolism , Energy Metabolism , Feeding Behavior/physiology , Adenylate Kinase/physiology , Exenatide , Glucagon-Like Peptide 1/physiology , Glucokinase/physiology , Homeostasis , Humans , Hypothalamus/physiology , Peptides/physiology , Ribosomal Protein S6 Kinases/physiology , Signal Transduction , TOR Serine-Threonine Kinases/physiology , Venoms
9.
Am J Physiol Endocrinol Metab ; 301(5): E922-30, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21810931

ABSTRACT

The plant-derived polyphenol resveratrol (RSV) modulates life span and metabolism, and it is thought that these effects are largely mediated by activating the deacetylase enzyme SIRT1. However, RSV also activates the cell energy sensor AMP-activated protein kinase (AMPK). We have previously reported that AMPK activators inhibit inducible nitric oxide synthase (iNOS), a key proinflammatory mediator of insulin resistance in endotoxemia and obesity. The aim of this study was to evaluate whether RSV inhibits iNOS induction in insulin target tissues and to determine the role of SIRT1 and AMPK activation in this effect. We found that RSV (40 mg/kg ip) treatment decreased iNOS induction and NO production in skeletal muscle and white adipose tissue, but not in liver, of endotoxin (LPS)-challenged mice. This effect of the polyphenol was recapitulated in vitro, where RSV (10-80 µM) robustly inhibited iNOS protein induction and NO production in cytokine/LPS-treated L6 myocytes and 3T3-L1 adipocytes. However, no effect of RSV was observed on iNOS induction in FAO hepatocytes. Further studies using inhibitors of SIRT1 revealed that the deacetylase enzyme is not involved in RSV action on iNOS. In marked contrast, RSV activates AMPK in L6 myocytes, and blunting its activation using Compound C or RNA interference partly blocked the inhibitory effect of RSV on NO production. These results show that RSV specifically inhibits iNOS induction in muscle through a mechanism involving AMPK but not SIRT1 activation. This anti-inflammatory action of RSV likely contributes to the therapeutic effect of this plant polyphenol.


Subject(s)
Adenylate Kinase/physiology , Muscle, Skeletal/drug effects , Nitric Oxide Synthase Type II/antagonists & inhibitors , Sirtuin 1/physiology , Stilbenes/pharmacology , 3T3-L1 Cells , Adenylate Kinase/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cells, Cultured , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Resveratrol , Signal Transduction/drug effects , Sirtuin 1/metabolism
10.
Am J Physiol Endocrinol Metab ; 300(4): E624-32, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21224484

ABSTRACT

Dysregulation of lipid metabolism is a key feature of metabolic disorder related to side effects of antipsychotic drugs. Here, we investigated the molecular mechanism by which second-generation atypical antipsychotic drugs (AAPDs) affect hepatic lipid metabolism in liver. AAPDs augmented hepatic lipid accumulation by activating expression of sterol regulatory element-binding protein (SREBP) transcription factors, with subsequent induction of downstream target genes involved in lipid and cholesterol synthesis in hepatocytes. We confirmed the direct involvement of SREBPs on AAPD-induced expression of lipogenic and cholesterogenic genes by utilization of adenovirus for dominant negative SREBP (Ad-SREBP-DN). Interestingly, AAPDs significantly decreased phosphorylation of AMPKα and expression of fatty acid oxidation genes. Treatment of constitutive active AMPK restored AAPD-mediated dysregulation of genes involved in both lipid synthesis and fatty acid oxidation. Moreover, AAPDs decreased transcriptional activity of PPARα, a critical transcriptional regulator for controlling hepatic fatty acid oxidation, via an AMPK-dependent manner. Close investigations revealed that mutations at the known p38 MAPK phosphorylation sites (S6/12/21A), but not mutations at the putative AMPKα phosphorylation sites (S167/373/453A), block AAPD-dependent reduction of PPARα transcriptional activity, suggesting that p38 MAPK might be also involved in the regulatory pathway as a downstream effector of AAPDs/AMPK. Taken together, these data suggest that AAPD-stimulated hepatic dysregulation of lipid metabolism could result from the inhibition of AMPK activity, and pharmaceutical means to potentiate AMPK activity would contribute to restore hepatic lipid homeostasis that occurs during AAPD treatment.


Subject(s)
Adenylate Kinase/physiology , Antipsychotic Agents/pharmacology , Lipid Metabolism/drug effects , Liver/drug effects , Adenylate Kinase/metabolism , Animals , Cells, Cultured , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lipid Metabolism/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism
11.
Am J Physiol Endocrinol Metab ; 296(4): E955-64, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19208854

ABSTRACT

Berberine (BBR) has been shown to improve several metabolic disorders, such as obesity, type 2 diabetes, and dyslipidemia, by stimulating AMP-activated protein kinase (AMPK). However, the effects of BBR on proinflammatory responses in macrophages are poorly understood. Here we show that BBR represses proinflammatory responses through AMPK activation in macrophages. In adipose tissue of obese db/db mice, BBR treatment significantly downregulated the expression of proinflammatory genes such as TNF-alpha, IL-1beta, IL-6, monocyte chemoattractant protein-1 (MCP-1), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2). Consistently, BBR inhibited LPS-induced expression of proinflammatory genes including IL-1beta, IL-6, iNOS, MCP-1, COX-2, and matrix metalloprotease-9 in peritoneal macrophages and RAW 264.7 cells. Upon various proinflammatory signals including LPS, free fatty acids, and hydrogen peroxide, BBR suppressed the phosphorylation of MAPKs, such as p38, ERK, and JNK, and the level of reactive oxygen species in macrophages. Moreover, these inhibitory effects of BBR on proinflammatory responses were abolished by AMPK inhibition via either compound C, an AMPK inhibitor, or dominant-negative AMPK, implying that BBR would downregulate proinflammatory responses in macrophages via AMPK stimulation.


Subject(s)
Adenylate Kinase/physiology , Berberine/pharmacology , Inflammation Mediators/antagonists & inhibitors , Macrophages/drug effects , 3T3-L1 Cells , Adenylate Kinase/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Berberine/therapeutic use , Cells, Cultured , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Macrophages/enzymology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Receptors, Leptin/genetics
12.
Rev Invest Clin ; 59(6): 458-69, 2007.
Article in Spanish | MEDLINE | ID: mdl-18402338

ABSTRACT

The adenine monophosphate (AMP) activated protein kinase (AMPK), is a heterotrimeric complex that is activated by an increase in the AMP/ATP ratio, and is considered to be a cellular energy sensor that contributes to regulate energy balance and caloric intake. AMPK is activated by LKB1 hinase and it can phophorylate several enzymes involved in anabolism to prevent further ATP consumption, and induces some catabolic enzymes to increase ATP generation. Furthermore, AMPK regulates the expression of genes involved in lipogenesis and mitochondrial biogenesis, among others. AMPK is distributed in most organs including, liver, skeletal muscle, heart and hypothalamus; and even in adipose cells. In addition, AMPK is activated in the hypothalamus stimulating appetite due to energy depletion. AMPK also participates in glycolysis regulation, glucose uptake, lipid oxidation, fatty acid synthesis, cholesterol synthesis and gluconeogenesis, and it has been considered as a possible target enzyme in the treatment of some diseases such as obesity, type 2 diabetes and hepatic steatosis. This review provides a general overview of AMPK structure, its activators and its function in the organism.


Subject(s)
Adenylate Kinase/physiology , Energy Metabolism/physiology , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Adenylate Kinase/antagonists & inhibitors , Adenylate Kinase/chemistry , Adipokines/physiology , Adipose Tissue/metabolism , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/physiopathology , Enzyme Activation/drug effects , Fatty Liver/drug therapy , Fatty Liver/enzymology , Fatty Liver/physiopathology , Glucose/metabolism , Humans , Hypoglycemic Agents/pharmacology , Hypothalamus/metabolism , Lipogenesis/physiology , Liver/metabolism , Myocardium/metabolism , Obesity/drug therapy , Obesity/enzymology , Obesity/physiopathology , Organ Specificity , Phosphorylation , Physical Exertion/physiology , Protein Processing, Post-Translational
13.
Protein Eng ; 14(11): 903-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11742110

ABSTRACT

Guanylate kinase catalyzes the phosphorylation of either GMP to GDP or dGMP to dGDP and is an important enzyme in nucleotide metabolic pathways. Because of its essential intracellular role, guanylate kinase is a target for a number of cancer chemotherapeutic agents such as 6-thioguanine and 8-azaguanine and is involved in antiviral drug activation. Guanylate kinase shares a similarity in function and structure to other nucleoside monophosphate kinases especially with that of the well-studied adenylate kinase. Amino acid substitutions were made within the GMP binding site of mouse guanylate kinase to alter the polarity of the side chains that interact with GMP as a means of evaluating the role that these residues play on substrate interaction. One of these mutants, E72Q/D103N, was shown by functional complementation and enzyme assays to embody both guanylate kinase activity and a novel adenylate kinase activity.


Subject(s)
Adenylate Kinase/physiology , Mutation , Nucleoside-Phosphate Kinase/genetics , Adenosine Monophosphate/metabolism , Adenylate Kinase/metabolism , Animals , Asparagine/chemistry , Binding Sites , DNA, Complementary/metabolism , Escherichia coli/enzymology , Escherichia coli/metabolism , Genetic Complementation Test , Genetic Vectors , Glutamine/chemistry , Guanosine Monophosphate/metabolism , Guanylate Kinases , Mice , Models, Chemical , Mutagenesis , Nucleoside-Phosphate Kinase/metabolism , Spectrophotometry , Substrate Specificity , Time Factors
14.
J Clin Psychiatry ; 52 Suppl: 58-62, 1991 May.
Article in English | MEDLINE | ID: mdl-2030105

ABSTRACT

Available therapies for treating depression, even the specific serotonin reuptake blockers, may not act via a serotonergic mechanism. Regardless of the mechanism of action, limitations in efficacy, onset of action, and side effects of all available antidepressant drugs indicate the need for further efforts to develop superior treatments of depression. A number of novel approaches are currently being investigated, including beta-adrenergic agonists, alpha-adrenergic antagonists, tetrahydrobiopterin, GABAergic drugs and non-pharmacologic therapies such as light therapy. These treatments may act independently of serotonin, yet there are intriguing suggestions that even their action is linked to serotonin.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder/drug therapy , Serotonin/physiology , Adenylate Kinase/physiology , Antidepressive Agents/pharmacology , Biopterins/analogs & derivatives , Biopterins/pharmacology , Biopterins/therapeutic use , Depressive Disorder/physiopathology , Depressive Disorder/therapy , Humans , Phototherapy , Protein Kinases/physiology , Receptors, Serotonin/drug effects , Receptors, Serotonin/physiology , gamma-Aminobutyric Acid/physiology
SELECTION OF CITATIONS
SEARCH DETAIL