Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nat Commun ; 15(1): 2102, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38453901

ABSTRACT

Nicotinamide adenine dinucleotide (NAD)+ serves as a crucial coenzyme in numerous essential biological reactions, and its cellular availability relies on the activity of the nicotinamide phosphoribosyltransferase (NAMPT)-catalyzed salvage pathway. Here we show that treatment with saturated fatty acids activates the NAD+ salvage pathway in hypothalamic astrocytes. Furthermore, inhibition of this pathway mitigates hypothalamic inflammation and attenuates the development of obesity in male mice fed a high-fat diet (HFD). Mechanistically, CD38 functions downstream of the NAD+ salvage pathway in hypothalamic astrocytes burdened with excess fat. The activation of the astrocytic NAMPT-NAD+-CD38 axis in response to fat overload induces proinflammatory responses in the hypothalamus. It also leads to aberrantly activated basal Ca2+ signals and compromised Ca2+ responses to metabolic hormones such as insulin, leptin, and glucagon-like peptide 1, ultimately resulting in dysfunctional hypothalamic astrocytes. Our findings highlight the significant contribution of the hypothalamic astrocytic NAD+ salvage pathway, along with its downstream CD38, to HFD-induced obesity.


Subject(s)
Dietary Fats , NAD , Male , Mice , Animals , NAD/metabolism , Dietary Fats/metabolism , Astrocytes/metabolism , Obesity/metabolism , Hypothalamus/metabolism , Cytokines/metabolism
2.
Endocrinol Metab (Seoul) ; 39(1): 1-11, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38356211

ABSTRACT

5´-Adenosine monophosphate (AMP)-activated protein kinase (AMPK), a cellular energy sensor, is an essential enzyme that helps cells maintain stable energy levels during metabolic stress. The hypothalamus is pivotal in regulating energy balance within the body. Certain neurons in the hypothalamus are sensitive to fluctuations in food availability and energy stores, triggering adaptive responses to preserve systemic energy equilibrium. AMPK, expressed in these hypothalamic neurons, is instrumental in these regulatory processes. Hypothalamic AMPK activity is modulated by key metabolic hormones. Anorexigenic hormones, including leptin, insulin, and glucagon-like peptide 1, suppress hypothalamic AMPK activity, whereas the hunger hormone ghrelin activates it. These hormonal influences on hypothalamic AMPK activity are central to their roles in controlling food consumption and energy expenditure. Additionally, hypothalamic AMPK activity responds to variations in glucose concentrations. It becomes active during hypoglycemia but is deactivated when glucose is introduced directly into the hypothalamus. These shifts in AMPK activity within hypothalamic neurons are critical for maintaining glucose balance. Considering the vital function of hypothalamic AMPK in the regulation of overall energy and glucose balance, developing chemical agents that target the hypothalamus to modulate AMPK activity presents a promising therapeutic approach for metabolic conditions such as obesity and type 2 diabetes mellitus.


Subject(s)
AMP-Activated Protein Kinases , Diabetes Mellitus, Type 2 , Humans , AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Glucose
3.
BMB Rep ; 55(6): 293-298, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35651327

ABSTRACT

Antipsychotics have been widely accepted as a treatment of choice for psychiatric illnesses such as schizophrenia. While atypical antipsychotics such as aripiprazole are not associated with obesity and diabetes, olanzapine is still widely used based on the anticipation that it is more effective in treating severe schizophrenia than aripiprazole, despite its metabolic side effects. To address metabolic problems, metformin is widely prescribed. Hypothalamic proopiomelanocortin (POMC) neurons have been identified as the main regulator of metabolism and energy expenditure. Although the relation between POMC neurons and metabolic disorders is well established, little is known about the effects of olanzapine and metformin on hypothalamic POMC neurons. In the present study, we investigated the effect of olanzapine and metformin on the hypothalamic POMC neurons in female mice. Olanzapine administration for 5 days significantly decreased Pomc mRNA expression, POMC neuron numbers, POMC projections, and induced leptin resistance before the onset of obesity. It was also observed that coadministration of metformin with olanzapine not only increased POMC neuron numbers and projections but also improved the leptin response of POMC neurons in the olanzapine-treated female mice. These findings suggest that olanzapine-induced hypothalamic POMC neuron abnormality and leptin resistance, which can be ameliorated by metformin administration, are the possible causes of subsequent hyperphagia. [BMB Reports 2022; 55(6): 293-298].


Subject(s)
Antipsychotic Agents , Metformin , Animals , Antipsychotic Agents/metabolism , Antipsychotic Agents/pharmacology , Aripiprazole/metabolism , Aripiprazole/pharmacology , Female , Hypothalamus/metabolism , Leptin/metabolism , Metformin/metabolism , Metformin/pharmacology , Mice , Neurons/metabolism , Obesity/drug therapy , Obesity/metabolism , Olanzapine/metabolism , Olanzapine/pharmacology , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/pharmacology
4.
Mol Cells ; 45(4): 169-176, 2022 Apr 30.
Article in English | MEDLINE | ID: mdl-35387896

ABSTRACT

A primary cilium, a hair-like protrusion of the plasma membrane, is a pivotal organelle for sensing external environmental signals and transducing intracellular signaling. An interesting linkage between cilia and obesity has been revealed by studies of the human genetic ciliopathies Bardet-Biedl syndrome and Alström syndrome, in which obesity is a principal manifestation. Mouse models of cell type-specific cilia dysgenesis have subsequently demonstrated that ciliary defects restricted to specific hypothalamic neurons are sufficient to induce obesity and hyperphagia. A potential mechanism underlying hypothalamic neuron cilia-related obesity is impaired ciliary localization of G protein-coupled receptors involved in the regulation of appetite and energy metabolism. A well-studied example of this is melanocortin 4 receptor (MC4R), mutations in which are the most common cause of human monogenic obesity. In the paraventricular hypothalamus neurons, a blockade of ciliary trafficking of MC4R as well as its downstream ciliary signaling leads to hyperphagia and weight gain. Another potential mechanism is reduced leptin signaling in hypothalamic neurons with defective cilia. Leptin receptors traffic to the periciliary area upon leptin stimulation. Moreover, defects in cilia formation hamper leptin signaling and actions in both developing and differentiated hypothalamic neurons. The list of obesity-linked ciliary proteins is expending and this supports a tight association between cilia and obesity. This article provides a brief review on the mechanism of how ciliary defects in hypothalamic neurons facilitate obesity.


Subject(s)
Cilia , Leptin , Animals , Cilia/metabolism , Humans , Hyperphagia/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Mice , Obesity/genetics , Obesity/metabolism
5.
Undersea Hyperb Med ; 48(1): 43-51, 2021.
Article in English | MEDLINE | ID: mdl-33648032

ABSTRACT

Background: Hyperbaric oxygen (HBO2) therapy is a safe and well-tolerated treatment modality. Seizures, one of the most severe central nervous system side effects of HBO2 therapy, can occur. Episodes of seizures during HBO2 therapy have not yet been reported in countries such as Korea, where hyperbaric medicine is still in the developmental stage. Methods: The registry data of all patients treated with HBO2 therapy in a tertiary academic hospital in Korea were prospectively collected, and patients who developed seizures during HBO2 therapy between October 2016 and December 2019 were evaluated. In addition, we reviewed previous studies on occurrence of seizures during HBO2 therapy. Results: During the study period, a total of 10,425 treatments were provided to 1,308 patients. The most frequently treated indication was carbon monoxide (CO) poisoning ABSTRACT (n=547, 41.8%). During the HBO2 therapy sessions (total: 10,425), five seizure episodes occurred (patients with CO poisoning: n=4; patients with arterial gas embolism [AGE]: n=1). The frequency of seizures in patients with CO poisoning (0.148%) and AGE (3.448%) was significantly higher than that in patients with all indications (0.048%) (p=0.001). None of the patients had lasting effects due to the seizures. Conclusion: Our study revealed a similar frequency rate in terms of all indications and CO poisoning, and a slightly higher rate in AGE. Seizures were observed in patients with CO poisoning and AGE. Therefore, if clinicians plan to operate a hyperbaric center in a country like Korea, where there are several patients with acute CO poisoning, they should be prepared to handle seizures that may occur during HBO2 therapy.


Subject(s)
Hyperbaric Oxygenation/adverse effects , Seizures/epidemiology , Adult , Carbon Monoxide Poisoning/therapy , Embolism, Air/therapy , Female , Humans , Hyperbaric Oxygenation/statistics & numerical data , Male , Middle Aged , Prospective Studies , Registries , Republic of Korea/epidemiology , Seizures/etiology
6.
Cell Metab ; 33(2): 334-349.e6, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33535098

ABSTRACT

Low-grade mitochondrial stress can promote health and longevity, a phenomenon termed mitohormesis. Here, we demonstrate the opposing metabolic effects of low-level and high-level mitochondrial ribosomal (mitoribosomal) stress in hypothalamic proopiomelanocortin (POMC) neurons. POMC neuron-specific severe mitoribosomal stress due to Crif1 homodeficiency causes obesity in mice. By contrast, mild mitoribosomal stress caused by Crif1 heterodeficiency in POMC neurons leads to high-turnover metabolism and resistance to obesity. These metabolic benefits are mediated by enhanced thermogenesis and mitochondrial unfolded protein responses (UPRmt) in distal adipose tissues. In POMC neurons, partial Crif1 deficiency increases the expression of ß-endorphin (ß-END) and mitochondrial DNA-encoded peptide MOTS-c. Central administration of MOTS-c or ß-END recapitulates the adipose phenotype of Crif1 heterodeficient mice, suggesting these factors as potential mediators. Consistently, regular running exercise at moderate intensity stimulates hypothalamic MOTS-c/ß-END expression and induces adipose tissue UPRmt and thermogenesis. Our findings indicate that POMC neuronal mitohormesis may underlie exercise-induced high-turnover metabolism.


Subject(s)
Hypothalamus/metabolism , Mitochondria/metabolism , Neurons/metabolism , Physical Conditioning, Animal , Pro-Opiomelanocortin/metabolism , Animals , Cell Line, Tumor , Energy Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
7.
Nat Commun ; 11(1): 5772, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33188191

ABSTRACT

Hypothalamic neurons including proopiomelanocortin (POMC)-producing neurons regulate body weights. The non-motile primary cilium is a critical sensory organelle on the cell surface. An association between ciliary defects and obesity has been suggested, but the underlying mechanisms are not fully understood. Here we show that inhibition of ciliogenesis in POMC-expressing developing hypothalamic neurons, by depleting ciliogenic genes IFT88 and KIF3A, leads to adulthood obesity in mice. In contrast, adult-onset ciliary dysgenesis in POMC neurons causes no significant change in adiposity. In developing POMC neurons, abnormal cilia formation disrupts axonal projections through impaired lysosomal protein degradation. Notably, maternal nutrition and postnatal leptin surge have a profound impact on ciliogenesis in the hypothalamus of neonatal mice; through these effects they critically modulate the organization of hypothalamic feeding circuits. Our findings reveal a mechanism of early life programming of adult adiposity, which is mediated by primary cilia in developing hypothalamic neurons.


Subject(s)
Adiposity , Cilia/metabolism , Hypothalamus/embryology , Hypothalamus/metabolism , Lysosomes/metabolism , Animals , Animals, Newborn , Arcuate Nucleus of Hypothalamus/metabolism , Axons/metabolism , Energy Metabolism , Female , Glucose/metabolism , Leptin/metabolism , Malnutrition/pathology , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neurogenesis , Obesity/metabolism , Obesity/pathology , Organogenesis , Pro-Opiomelanocortin/metabolism , Proteolysis
8.
Mol Cells ; 43(5): 431-437, 2020 May 31.
Article in English | MEDLINE | ID: mdl-32392909

ABSTRACT

The hypothalamus is a crucial organ for the maintenance of appropriate body fat storage. Neurons in the hypothalamic arcuate nucleus (ARH) detect energy shortage or surplus via the circulating concentrations of metabolic hormones and nutrients, and then coordinate energy intake and expenditure to maintain energy homeostasis. Malfunction or loss of hypothalamic ARH neurons results in obesity. Accumulated evidence suggests that hypothalamic inflammation is a key pathological mechanism that links chronic overconsumption of a high-fat diet (HFD) with the development of obesity and related metabolic complications. Interestingly, overnutrition-induced hypothalamic inflammation occurs specifically in the ARH, where microglia initiate an inflammatory response by releasing proinflammatory cytokines and chemokines in response to excessive fatty acid flux. Upon more prolonged HFD consumption, astrocytes and perivascular macrophages become involved and sustain hypothalamic inflammation. ARH neurons are victims of hypothalamic inflammation, but they may actively participate in hypothalamic inflammation by sending quiescence or stress signals to surrounding glia. In this mini-review, we describe the current state of knowledge regarding the contributions of neurons and glia, and their interactions, to HFD-induced hypothalamic inflammation.


Subject(s)
Adipose Tissue/immunology , Hypothalamus/immunology , Inflammation/metabolism , Macrophages/immunology , Microglia/immunology , Neurons/immunology , Obesity/immunology , Animals , Cytokines/metabolism , Diet, High-Fat , Energy Metabolism , Humans , Immunity, Cellular , Neurogenic Inflammation
9.
J Neuroinflammation ; 16(1): 221, 2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31727092

ABSTRACT

BACKGROUND: Obese mice on a high-fat diet (HFD) display signs of inflammation in the hypothalamic arcuate nucleus (ARC), a critical area for controlling systemic energy metabolism. This has been suggested as a key mechanism of obesity-associated hypothalamic dysfunction. We reported earlier that bone marrow-derived macrophages accumulate in the ARC to sustain hypothalamic inflammation upon chronic exposure to an HFD. However, the mechanism underlying hypothalamic macrophage accumulation has remained unclear. METHODS: We investigated whether circulating monocytes or myeloid precursors contribute to hypothalamic macrophage expansion during chronic HFD feeding. To trace circulating myeloid cells, we generated mice that express green fluorescent protein (GFP) in their lysozyme M-expressing myeloid cells (LysMGFP mice). We conducted parabiosis and bone marrow transplantation experiments using these animals. Mice received an HFD for 12 or 30 weeks and were then sacrificed to analyze LysMGFP cells in the hypothalamus. Hypothalamic vascular permeability in the HFD-fed obese mice was also tested by examining the extravascular leakage of Evans blue and fluorescence-labeled albumin. The timing of LysMGFP cell entry to the hypothalamus during development was also evaluated. RESULTS: Our parabiosis and bone marrow transplantation experiments revealed a significant infiltration of circulating LysMGFP cells into the liver, skeletal muscle, choroid plexus, and leptomeninges but not in the hypothalamic ARC during chronic HFD feeding, despite increased hypothalamic vascular permeability. These results suggested that the recruitment of circulating monocytes is not a major mechanism for maintaining and expanding the hypothalamic macrophage population in diet-induced obesity. We demonstrated instead that LysMGFP cells infiltrate the hypothalamus during its development. LysMGFP cells appeared in the hypothalamic area from the late embryonic period. This cellular pool suddenly increased immediately after birth, peaked at the postnatal second week, and adopted an adult pattern of distribution after weaning. CONCLUSIONS: Bone marrow-derived macrophages mostly populate the hypothalamus in early postnatal life and may maintain their pool without significant recruitment of circulating monocytes throughout life, even under conditions of chronic HFD feeding.


Subject(s)
Hypothalamus/metabolism , Macrophages/metabolism , Obesity/metabolism , Animals , Bone Marrow Transplantation , Capillary Permeability , Diet, High-Fat , Energy Metabolism , Insulin Resistance/physiology , Liver/metabolism , Male , Mice , Parabiosis
10.
Cell Rep ; 25(4): 934-946.e5, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30355499

ABSTRACT

Obesity-associated metabolic alterations are closely linked to low-grade inflammation in peripheral organs, in which macrophages play a central role. Using genetic labeling of myeloid lineage cells, we show that hypothalamic macrophages normally reside in the perivascular area and circumventricular organ median eminence. Chronic consumption of a high-fat diet (HFD) induces expansion of the monocyte-derived macrophage pool in the hypothalamic arcuate nucleus (ARC), which is significantly attributed to enhanced proliferation of macrophages. Notably, inducible nitric oxide synthase (iNOS) is robustly activated in ARC macrophages of HFD-fed obese mice. Hypothalamic macrophage iNOS inhibition completely abrogates macrophage accumulation and activation, proinflammatory cytokine overproduction, reactive astrogliosis, blood-brain-barrier permeability, and lipid accumulation in the ARC of obese mice. Moreover, central iNOS inhibition improves obesity-induced alterations in systemic glucose metabolism without affecting adiposity. Our findings suggest a critical role for hypothalamic macrophage-expressed iNOS in hypothalamic inflammation and abnormal glucose metabolism in cases of overnutrition-induced obesity.


Subject(s)
Hypothalamus/pathology , Inflammation/enzymology , Macrophages/enzymology , Nitric Oxide Synthase Type II/metabolism , Obesity/enzymology , Animals , Arcuate Nucleus of Hypothalamus/pathology , Blood-Brain Barrier/pathology , Cell Proliferation , Diet, High-Fat , Glucose/metabolism , Inflammation/pathology , Macrophage Activation , Mice , Mice, Inbred C57BL , Mice, Obese , Nitric Oxide Synthase Type II/antagonists & inhibitors , Obesity/pathology , RAW 264.7 Cells
11.
Metabolism ; 88: 51-60, 2018 11.
Article in English | MEDLINE | ID: mdl-30179604

ABSTRACT

BACKGROUND: Nicotinamide adenine dinucleotide (NAD)-dependent deacetylase SIRT1 is an important regulator of hypothalamic neuronal function. Thus, an adequate hypothalamic NAD content is critical for maintaining normal energy homeostasis. METHODS: We investigated whether NAD supplementation increases hypothalamic NAD levels and affects energy metabolism in mice. Furthermore, we investigated the mechanisms underlying the effects of exogenous NAD on central metabolism upon entering the hypothalamus. RESULTS: Central and peripheral NAD administration suppressed fasting-induced hyperphagia and weight gain in mice. Extracellular NAD was imported into N1 hypothalamic neuronal cells in a connexin 43-dependent and CD73-independent manner. Consistent with the in vitro data, inhibition of hypothalamic connexin 43 blocked hypothalamic NAD uptake and NAD-induced anorexia. Exogenous NAD suppressed NPY and AgRP transcriptional activity, which was mediated by SIRT1 and FOXO1. CONCLUSIONS: Exogenous NAD is effectively transported to the hypothalamus via a connexin 43-dependent mechanism and increases hypothalamic NAD content. Therefore, NAD supplementation is a potential therapeutic method for metabolic disorders characterized by hypothalamic NAD depletion.


Subject(s)
Connexin 43/metabolism , Energy Metabolism/drug effects , Hypothalamus/drug effects , NAD/pharmacology , Agouti-Related Protein/genetics , Animals , Biological Transport , Hyperphagia/prevention & control , Hypothalamus/cytology , Hypothalamus/metabolism , Injections, Intraperitoneal , Injections, Intraventricular , Male , Mice, Inbred C57BL , NAD/administration & dosage , Neurons/metabolism , Neuropeptide Y/genetics , Sirtuin 1/metabolism , Transcription, Genetic/drug effects , Weight Gain/drug effects
12.
Antiviral Res ; 144: 266-272, 2017 08.
Article in English | MEDLINE | ID: mdl-28668556

ABSTRACT

The aim of this study was to establish the effect of a 70% ethanol extract of Elaeocarpus sylvestris (ESE) on varicella-zoster virus (VZV) replication and identify the specific bioactive component(s) underlying its activity. ESE induced a significant reduction in replication of the clinical strain of VZV. Activity-guided fractionation indicated that the ethyl acetate (EtOAc) fraction of ESE contains the active compound(s) inhibiting VZV replication. High-Performance Liquid Chromatography coupled to Electrospray Ionization Quadrupole Time-of-Flight Mass Spectrometry (HPLC-Q-TOF-MS/MS) analysis of the EtOAc fraction of ESE facilitated the identification of 13 chemical components. Among these, 1,2,3,4,6-penta-O-galloyl-ß-D-glucose (PGG) markedly suppressed VZV-induced c-Jun N-terminal kinase (JNK) activation, expression of viral immediate-early 62 (IE62) protein and VZV replication. Our results collectively support the utility of PGG as a potential candidate anti-viral drug to treat VZV-associated diseases.


Subject(s)
Elaeocarpaceae/chemistry , Herpesvirus 3, Human/drug effects , Hydrolyzable Tannins/pharmacology , Plant Extracts/chemistry , Virus Replication/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Herpesvirus 3, Human/physiology , Humans , Hydrolyzable Tannins/isolation & purification , Immediate-Early Proteins/analysis , JNK Mitogen-Activated Protein Kinases/analysis , Spectrometry, Mass, Electrospray Ionization , Trans-Activators/analysis , Viral Envelope Proteins/analysis
13.
Diabetes ; 64(4): 1142-53, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25338813

ABSTRACT

Hypothalamic lipid sensing is important for the maintenance of energy balance. Angiopoietin-like protein 3 (Angptl3) critically regulates the clearance of circulating lipids by inhibiting lipoprotein lipase (LPL). The current study demonstrated that Angptl3 is highly expressed in the neurons of the mediobasal hypothalamus, an important area in brain lipid sensing. Suppression of hypothalamic Angptl3 increased food intake but reduced energy expenditure and fat oxidation, thereby promoting weight gain. Consistently, intracerebroventricular (ICV) administration of Angptl3 caused the opposite metabolic changes, supporting an important role for hypothalamic Angptl3 in the control of energy balance. Notably, ICV Angptl3 significantly stimulated hypothalamic LPL activity. Moreover, coadministration of the LPL inhibitor apolipoprotein C3 antagonized the effects of Angptl3 on energy metabolism, indicating that LPL activation is critical for the central metabolic actions of Angptl3. Increased LPL activity is expected to promote lipid uptake by hypothalamic neurons, leading to enhanced brain lipid sensing. Indeed, ICV injection of Angptl3 increased long-chain fatty acid (LCFA) and LCFA-CoA levels in the hypothalamus. Furthermore, inhibitors of hypothalamic lipid-sensing pathways prevented Angptl3-induced anorexia and weight loss. These findings identify Angptl3 as a novel regulator of the hypothalamic lipid-sensing pathway.


Subject(s)
Angiopoietins/metabolism , Energy Metabolism/physiology , Fatty Acids/metabolism , Hypothalamus/metabolism , Lipoprotein Lipase/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins , Angiopoietins/genetics , Angiopoietins/pharmacology , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Eating/drug effects , Eating/physiology , Energy Metabolism/drug effects , Hypothalamus/drug effects , Male , Mice , RNA Interference , Rats , Rats, Sprague-Dawley
14.
Clin Rheumatol ; 32(6): 829-37, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23397143

ABSTRACT

The aim of this study was to identify the factors associated with gout among South Koreans. A case control study of gout patients newly diagnosed between January 1, 2007, and December 31, 2008, and matching controls was conducted using the nationwide database (National Health Insurance Corporation and National Health Screening Exam (NHSE) database), which included the health-care records of 48.1 million individuals. Of 495,998 newly diagnosed patients, we included 18,123 who were ≥40 years old and had an NHSE before diagnosis of gout. To elucidate the factors associated with gout, multivariate conditional logistic analyses were performed. Gout was associated with drinking ≥1/week (p < 0.001), drinking ≥1 bottle of soju/session (p < 0.001), high body mass index (BMI) (p < 0.001), high blood pressure (p < 0.001), high total cholesterol (p < 0.001), proteinuria (multivariate odds ratio (OR) = 1.75; 95 % confidence interval (CI) = 1.53-2.00), and an elevated uric acid (multivariate OR = 1.54; 95 % CI = 1.22-1.94). Exercise frequency was not significantly associated with gout. Prediabetic blood sugar level (100-125 mg/dL) was associated with gout in the univariate analysis, but not in the multivariate analysis. Diabetic blood sugar level (≥126 mg/dL) was associated with a decreased odds of gout (multivariate OR = 0.79; 95 % CI = 0.73-0.86). Our nationwide South Korean study showed that frequent and excessive alcohol consumption, high BMI, high blood pressure, high total cholesterol, proteinuria, and high uric acid are associated with gout.


Subject(s)
Gout/epidemiology , Adult , Aged , Alcohol Drinking , Blood Pressure , Body Mass Index , Case-Control Studies , Databases, Factual , Female , Gout/diagnosis , Health Surveys , Humans , Male , Middle Aged , Multivariate Analysis , National Health Programs , Odds Ratio , Republic of Korea
15.
Acupunct Electrother Res ; 34(1-2): 27-40, 2009.
Article in English | MEDLINE | ID: mdl-19711773

ABSTRACT

Cancer pain impairs the quality of life of cancer patients, but opioid analgesics can not only cause inhibition of respiratory function, and constipation, but also other significant side effects such as addiction and tolerance that further decrease quality of life. Thus, in the present study, the effects of electro-acupuncture treatment (EA) on mechanical allodynia were examined in cancer pain mouse model. In order to induce neuropathic cancer pain model, S-180 sarcoma cells were inoculated around the sciatic nerve of left legs of Balb/c mice. The mass of S-180 cancer cells embedded around sciatic nerve in a time course was confirmed by Magnetic Resonance Imaging (MRI) scanning. Mechanical allodynia was most consistently induced in mouse sarcoma cell line S-180 (2 x 10(6) sarcoma cells) treated group among all groups. EA stimulation (2Hz) was daily given to ST36 (Zusanli) of S-180 bearing mice for 30 min for 9 days after S-180 inoculation. EA treatment significantly prolonged paw withdrawal latency from 5 days after inoculation as well as shortened cumulative lifting duration from 7 days after inoculation compared with tumor control. In addition, the overexpressions of pain peptide substance P in dorsal horn of spinal cord were significantly decreased in EA treated group compared with tumor control on Day 9 after inoculation. Furthermore, EA treatment effectively increased the concentration of beta endorphin in blood and brain of mice more than tumor control as well as normal group. The concentration of beta-endorphin for EA treatment group increased by 51.457% in blood 12.6% in brain respectively, compared with tumor control group. These findings suggest that S-180 cancer pain model can be a consistent and short time animal model and also EA treatment can be an alternative therapeutic method for cancer pain via decreased substance P and increased beta endorphin.


Subject(s)
Acupuncture Analgesia , Electroacupuncture , Neoplasms/complications , Pain Management , Substance P/metabolism , beta-Endorphin/metabolism , Acupuncture Points , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred BALB C , Pain/etiology , Pain/metabolism , Pain Threshold
SELECTION OF CITATIONS
SEARCH DETAIL