Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
Add more filters

Publication year range
1.
Mol Biol Rep ; 47(4): 2703-2711, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32180086

ABSTRACT

The combination of sarcopenia and obesity (i.e., sarcopenic obesity) is more strongly associated with disability and metabolic/cardiovascular diseases than obesity or sarcopenia alone. Therefore, countermeasures that simultaneously suppress fat gain and muscle atrophy to prevent an increase in sarcopenic obesity are warranted. The aim of this study was to investigate the simultaneous effects of fucoxanthinol (FXOH) on fat loss in mature adipocytes and the inhibition of atrophy and loss in myotubes induced by oxidative stress. C2C12 myotubes were treated with FXOH for 24 h and further incubated with hydrogen peroxide (H2O2) for 24 h. The area of myosin heavy chain-positive myotubes and the ROS concentration were measured. Mature 3T3-L1 adipocytes were treated with FXOH for 72 h. The triacylglycerol (TG) content and glycerol and fatty acid (FA) release were biochemically measured. The myotube area was smaller in H2O2-treated cells than that in control cells. However, FXOH protected against the H2O2-induced decreases in myotube area. Further, the ROS concentration was significantly higher in the FXOH-treated cells compared with that in the control cells, although it was significantly lower than that in the H2O2-treated cells. On the other hand, in the mature adipocytes, the TG content was significantly decreased by FXOH treatment compared to that in the control. Moreover, FXOH treatment significantly increased glycerol and FA release compared with that of the control. These results suggest that FXOH inhibits H2O2-induced atrophy and loss in myotubes and activates lipolysis and decreases the TG content in mature adipocytes. Accordingly, FXOH has the potential to exert anti-sarcopenic obesity effects.


Subject(s)
Muscular Atrophy/metabolism , Oxidative Stress/drug effects , beta Carotene/analogs & derivatives , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Atrophy/metabolism , Cell Line , Fatty Acids/metabolism , Hydrogen Peroxide/metabolism , Lipolysis/drug effects , Metabolic Diseases/pathology , Mice , Muscle Fibers, Skeletal/metabolism , Obesity/metabolism , Oxidative Stress/physiology , Triglycerides/metabolism , beta Carotene/metabolism , beta Carotene/pharmacology
2.
Biometals ; 31(3): 415-424, 2018 06.
Article in English | MEDLINE | ID: mdl-29744695

ABSTRACT

Lactoferrin (LF) is a multifunctional protein in mammalian milk. We previously reported that enteric-coated bovine LF reduced the visceral fat in a double-blind clinical study. We further demonstrated that bovine LF (bLF) inhibited adipogenesis and promoted lipolysis in white adipocytes, but the effect of bLF on brown adipocytes has not been clarified. In this study, we investigated the effects of bLF on energy expenditure and cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling pathway using human reprogrammed brown adipocytes generated by gene transduction. bLF at concentrations of ≥ 100 µg/mL significantly increased uncoupling protein 1 (UCP1) mRNA levels, with the maximum value observed 4 h after bLF addition. At the same time point, bLF stimulation also significantly increased oxygen consumption. Signaling pathway analysis revealed rapid increases of intracellular cAMP and cAMP response element-binding protein (CREB) phosphorylation levels beginning 5 min after bLF addition. The mRNA levels of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) were also significantly increased after 1 h of bLF stimulation. H-89, a specific PKA inhibitor, abrogated bLF-induced UCP1 gene expression. Moreover, receptor-associated protein (Rap), an antagonist of low-density lipoprotein receptor-related protein 1 (LRP1), significantly reduced bLF-induced UCP1 gene expression in a dose-dependent manner. These results suggest that bLF promotes UCP1 gene expression in brown adipocytes through the cAMP-PKA signaling pathway via the LRP1 receptor, leading to increased energy expenditure.


Subject(s)
Adenosine Monophosphate/metabolism , Adipocytes, Brown/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Energy Metabolism , Lactoferrin/metabolism , Signal Transduction , Animals , Cattle , Humans , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
3.
Biochem Cell Biol ; 95(1): 142-147, 2017 02.
Article in English | MEDLINE | ID: mdl-28165288

ABSTRACT

Lactoferrin (LF) is a multifunctional cationic protein (pI 8.2-8.9) in mammalian milk. We previously reported that enteric-LF prevented hypercholesterolemia and atherosclerosis in a diet-induced atherosclerosis model using Microminipig, although the underlying mechanisms remain unclear. Because LF is assumed to electrostatically interact with bile acids to inhibit intestinal cholesterol absorption, LF could promote cholesterol excretion. In this study, we assessed the interaction between LF and taurocholate in vitro, and the effect of LF on cholesterol excretion in rats. The binding rate of taurocholate to LF was significantly higher than that to transferrin (pI 5.2-6.3). When rats were administered a high-cholesterol diet (HCD) containing 5% LF, LF was detected using ELISA in the upper small intestine from 7.5 to 60 min after the administration. Rats were fed one of the following diets: control, HCD, or HCD + 5% LF for 21 days. Fecal neutral steroids and hepatic cholesterol levels in the HCD group were significantly higher than those in the control group. The addition of LF to a HCD significantly increased fecal neutral steroids levels (22% increase, p < 0.05) and reduced hepatic cholesterol levels (17% decrease, p < 0.05). These parameters were inversely correlated (R = -0.63, p < 0.05). These results suggest that LF promotes cholesterol excretion via interactions with bile acids.


Subject(s)
Anti-Infective Agents/metabolism , Cholesterol/metabolism , Feces/chemistry , Lactoferrin/metabolism , Taurocholic Acid/metabolism , Animals , Cattle , Male , Rats , Rats, Sprague-Dawley
4.
J Sleep Res ; 25(6): 746-753, 2016 12.
Article in English | MEDLINE | ID: mdl-27338238

ABSTRACT

We have demonstrated previously that Japanese sake yeast improves sleep quality in humans. In the present study, we examined the molecular mechanisms of sake yeast to induce sleep by monitoring locomotor activity, electromyogram and electroencephalogram in mice. Oral administration of Japanese sake yeast (100, 200, and 300 mg kg-1 ) decreased the locomotor activity by 18, 46 and 59% and increased the amount of non-rapid eye movement (NREM) sleep by 1.5-, 2.3- and 2.4-fold (to 37 ± 6, 57 ± 8, and 60 ± 4 min from 25 ± 6 min in the vehicle-administered group, respectively) in a dose-dependent manner for 4 h after oral administration. However, Japanese sake yeast did not change the amount of rapid eye movement (REM) sleep, the electroencephalogram power density during NREM sleep or show any adverse effects, such as rebound of insomnia, during 24 h postadministration and on the next day. An intraperitoneal pretreatment with an adenosine A2A receptor-selective antagonist, ZM241385 (15 mg kg-1 ), reduced the amount of NREM sleep of sake yeast-administered mice to the basal level, without changing basal amount of sleep. Conversely, an A1 receptor-selective antagonist, 8-cyclopentyltheophylline (10 mg kg-1 ), did not affect the sleep-promoting effect of Japanese sake yeast. Thus, Japanese sake yeast promotes NREM sleep via activation of adenosine A2A but not A1 receptors.


Subject(s)
Alcoholic Beverages/microbiology , Eye Movements/physiology , Receptor, Adenosine A2A/metabolism , Saccharomyces cerevisiae/classification , Sleep/physiology , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Administration, Oral , Animals , Electroencephalography , Electromyography , Eye Movements/drug effects , Japan , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Sleep/drug effects , Sleep, REM/physiology , Theophylline/analogs & derivatives , Theophylline/pharmacology , Time Factors , Triazines/pharmacology , Triazoles/pharmacology
5.
J Sleep Res ; 25(1): 116-23, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26354605

ABSTRACT

Activation of adenosine A2a receptors in cerebral neurons induces sleep in various mammals. It was previously found that Japanese sake yeast enriched in adenosine analogues activates A2a receptors in vitro and induces sleep in mice. Here it is reported that sake yeast activated A2a receptors in a cultured human cell line and improved human sleep quality in a clinical trial. Sake yeast activated A2a receptors in HEK cells in a dose-dependent manner with an EC50 of 40 µg mL(-1), and the activation was attenuated almost completely by the A2a receptor antagonist ZM241385 with an IC50 of 73 nm. In a double-blind placebo-controlled crossover clinical study, 68 healthy participants ingested tablets containing either 500 mg of sake yeast powder or a placebo (cellulose) 1 h before sleep for 4 days. Electroencephalograms were recorded during sleep at home with a portable device for 4 week days. Electroencephalogram analyses revealed that sake yeast supplementation significantly (P = 0.03) increased delta power during the first cycle of slow-wave sleep by 110%, without changing other sleep parameters. Sake yeast supplementation also significantly increased growth hormone secretion in the urine on awakening by 137% from 3.17 ± 0.41 (placebo) to 4.33 ± 0.62 (sake yeast) pg mg(-1) creatinine (P = 0.03). Subjective sleepiness (P = 0.02) and fatigue (P = 0.06) in the morning were improved by sake yeast. Given these benefits and the absence of adverse effects during the study period, it was concluded that sake yeast supplementation is an effective and safe way to support daily high-quality, deep sleep.


Subject(s)
Alcoholic Beverages/microbiology , Cell Extracts/administration & dosage , Cell Extracts/pharmacology , Saccharomyces cerevisiae/chemistry , Sleep/drug effects , Sleep/physiology , Adenosine A2 Receptor Antagonists/pharmacology , Adult , Cell Extracts/adverse effects , Cross-Over Studies , Double-Blind Method , Electroencephalography , Female , HEK293 Cells , Humans , Male , Powders , Receptor, Adenosine A2A/metabolism , Sleep Stages/drug effects , Sleep Stages/physiology , Triazines/pharmacology , Triazoles/pharmacology
6.
Biosci Biotechnol Biochem ; 80(2): 295-303, 2016.
Article in English | MEDLINE | ID: mdl-26549014

ABSTRACT

Previously, we found that enteric lactoferrin (eLF) could reduce the visceral fat accumulation known to associate strongly with metabolic syndrome symptoms and consequently with an increased risk of atherosclerosis. In this study, the atherosclerosis-preventive potential of LF was assessed in a high-fat and high-cholesterol diet (HFCD)-induced hypercholesterolemia and atherosclerosis model using Microminipig™. Eight-week orally administered eLF remarkably reduced the HFCD-induced serum total and low-density lipoprotein cholesterol levels but not high-density lipoprotein cholesterol levels. A histological analysis of 15 arteries revealed that eLF systemically inhibited the development of atherosclerotic lesions. Pathway analysis using identified genes that characterized eLF administration in liver revealed significant changes in the steroid biosynthesis pathway (ssc00100) and all affected genes in this pathway were upregulated, suggesting that cholesterol synthesis inhibited by HFCD was recovered by eLF. In summary, eLF could potentially prevent the hypercholesterolemia and atherosclerosis through protecting homeostasis from HFCD-induced dysfunction of cholesterol metabolism.


Subject(s)
Atherosclerosis/diet therapy , Cholesterol, Dietary/adverse effects , Diet, High-Fat , Hypercholesterolemia/diet therapy , Lactoferrin/pharmacology , Administration, Oral , Animals , Arteries , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Gene Expression Regulation , Gene Ontology , Hypercholesterolemia/etiology , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Liver/drug effects , Liver/metabolism , Male , Molecular Sequence Annotation , Oligonucleotide Array Sequence Analysis , Signal Transduction , Swine , Swine, Miniature , Triglycerides/blood
7.
Biochem Cell Biol ; 93(6): 566-73, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26335364

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) describes a spectrum of lesions ranging from simple steatosis to non-alcoholic steatohepatitis (NASH). The excess influx of fatty acids (FAs) into the liver is recognized as a main cause of simple steatosis formation and progression to NASH. Recently, administration of lactoferrin (LF), a glycoprotein present in milk, was suggested to prevent NAFLD development. However, the effect of LF on the contribution of FA to NAFLD development remains unclear. In this study, the effects of LF on FA mixture (FAm)-induced lipotoxicity using human hepatocarcinoma G2 cells were assessed. FAm significantly decreased cell viability and increased intracellular lipid accumulation, whereas LF significantly recovered cell viability without affecting lipid accumulation. FAm-induced lactic dehydrogenase (LDH) and caspase-3/7 activities were significantly decreased by LF and SP600125, a c-Jun N-terminal kinase (JNK) specific inhibitor. We also found that LF added to FAm-treated cells induced Akt phosphorylation, which contributed to inhibition of JNK signaling pathway-dependent apoptosis. Akt inhibitor VIII, an allosteric Akt inhibitor, significantly attenuated the effect of LF on LDH activity and abrogated the ones on cell viability and caspase-3/7 activity. In summary, the present study has revealed that LF has a protective effect on FAm-induced lipotoxicity in a HepG2 model of NAFLD and identified the activation of the Akt signaling pathway as a possibly major mechanism.


Subject(s)
Lactoferrin/pharmacology , Lipid Metabolism/drug effects , Lipotropic Agents/pharmacology , Liver/drug effects , MAP Kinase Signaling System/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , Proto-Oncogene Proteins c-akt/agonists , Animals , Anthracenes/pharmacology , Apoptosis/drug effects , Benzimidazoles/pharmacology , Cattle , Fatty Acids, Nonesterified/adverse effects , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/metabolism , Hep G2 Cells , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Lactoferrin/antagonists & inhibitors , Lactoferrin/chemistry , Lactoferrin/metabolism , Lipotropic Agents/chemistry , Lipotropic Agents/metabolism , Liver/metabolism , Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Quinoxalines/pharmacology
8.
Nitric Oxide ; 44: 61-70, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25499030

ABSTRACT

Antipyretic analgesic drugs (including non-steroidal anti-inflammatory drugs) inhibit cyclooxygenase-2 and inducible nitric oxide synthase (iNOS), resulting in decreases of the proinflammatory mediators prostaglandin E2 and nitric oxide (NO), respectively. Both mediators are regulated by nuclear factor-kappa B (NF-κB), a key transcription factor in inflammation. Few reports have compared the efficacy and potency of anti-inflammatory drugs as NO inhibitors. In our study, we examined the effects of four popular antipyretic analgesic drugs on NO production induced in hepatocytes and macrophages. Mouse RAW264.7 macrophages treated with bacterial lipopolysaccharide showed the highest efficacy with regard to NO production; aspirin, loxoprofen, ibuprofen, and acetaminophen dose-dependently suppressed NO induction. Ibuprofen showed the highest potency in suppressing the induced production of NO. In rat hepatocytes, all the drugs inhibited interleukin 1ß-induced NO production and ibuprofen and loxoprofen inhibited NO induction effectively. Unexpectedly, the potency of NO suppression of each drug in hepatocytes did not always correlate with that observed in RAW264.7 cells. Microarray analyses of mRNA expression in hepatocytes revealed that the effects of the four antipyretic analgesic drugs modulated the NF-κB signaling pathway in a similar manner to the regulation of the expression of genes associated with inflammation, including the iNOS gene. However, the affected signal-transducing molecules in the NF-κB pathway were different for each drug. Therefore, antipyretic analgesic drugs may decrease NO production by modulating the NF-κB pathway in different ways, which could confer different efficacies and potencies with regard to their anti-inflammatory effects.


Subject(s)
Analgesics/pharmacology , Antipyretics/pharmacology , Hepatocytes/drug effects , Macrophages/drug effects , Nitric Oxide Synthase Type II/metabolism , Animals , Cell Line , Gene Expression/drug effects , Hepatocytes/enzymology , Macrophages/enzymology , Male , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Rats , Rats, Wistar , Signal Transduction/drug effects
9.
Exp Cell Res ; 319(18): 2835-44, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23948305

ABSTRACT

It has been suggested that nitric oxide (NO) derived from chronically inflamed tissues is a cause of carcinogenesis. We herein demonstrated that administration of an inducible NO synthase inhibitor, aminoguanidine, significantly suppressed the tumorigenic conversion of human colonic adenoma (FPCK-1-1) cells into adenocarcinoma (FPCK/Inflam) cells accelerated by foreign body-induced chronic inflammation in nude mice. To determine whether NO directly promotes carcinogenesis, we exposed FPCK-1-1 cells continuously to chemically generated NO (FPCK/NO), and periodically examined their tumorigenicity. FPCK/NO cells formed tumors, whereas vehicle-treated cells (FPCK/NaOH) did not. We selected a tumorigenic population from FPCK/NO cells kept it in three-dimensional (3D) culture where in vivo-like multicellular spheroidal growth was expected. FPCK/Inflam cells developed large spheroids whereas FPCK/NO cells formed tiny but growing compact aggregates in 3D culture. Meanwhile, FPCK-1-1 and FPCK/NaOH cells underwent anoikis (apoptotic cell death consequential on insufficient cell-to-substrate interactions) through activation of caspase 3. The survived cells in the 3D culture (FPCK/NO/3D), which were derived from FPCK/NO cells, showed a similar tumor incidence to that of FPCK/Inflam cells. These results showed that NO was one of the causative factors for the acceleration of colon carcinogenesis, especially in the conversion from adenoma to adenocarcinoma in the chronic inflammatory environment.


Subject(s)
Adenocarcinoma/pathology , Adenoma/pathology , Colonic Neoplasms , Inflammation , Nitric Oxide/metabolism , Adenocarcinoma/physiopathology , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Female , Guanidines/pharmacology , Humans , Immunohistochemistry , Male , Mice , Mice, Nude
10.
Int J Clin Oncol ; 19(1): 87-97, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23380957

ABSTRACT

BACKGROUND: To investigate the associations between serum concentrations of carotenoids and the presence of colorectal polyps and cancers in Japanese using a cross-sectional study. METHODS: 893 subjects who underwent colorectal endoscopy between 2001 and 2002 provided serum samples and information on lifestyle factors. Serum concentrations of six carotenoids were compared among patients with polyps, cancers, and controls. RESULTS: In males, high serum zeaxanthin was associated with decreased rates of polyps [odds ratio (OR) = 0.48, 95 % confidence interval (CI) 0.27-0.87] and cancer (OR = 0.35, 95 % CI 0.12-1.06), adjusting for age, body mass index, serum cholesterol, smoking status, and alcohol intake. In females, zeaxanthin (OR = 0.25, 95 % CI 0.07-0.82), lutein (OR = 0.30, 95 % CI 0.10-0.94), alpha-carotene (OR = 0.30, 95 % CI 0.10-0.90), and beta-carotene (OR = 0.27, 95 % CI 0.09-0.85) showed significant inverse associations with cancer development. These associations were consistent with findings of inverse associations between the ingestion of green-yellow vegetables (OR = 0.44, 95 % CI 0.23-0.84), carrots and pumpkins (OR = 0.46, 95 % CI 0.25-0.86), and fruits (OR = 0.53, 95 % CI 0.30-0.94) and polyp in males, and between carrots and pumpkins (OR = 0.30, 95 % CI 0.09-0.99), legumes (OR = 0.14, 95 % CI 0.04-0.44), and seaweed (OR = 0.23, 95 % CI 0.07-0.75) and cancer development in females. CONCLUSIONS: These results provide further support for the protective effects of carotenoids contained in green-yellow vegetables and fruits against colorectal neoplasm in Japanese.


Subject(s)
Colorectal Neoplasms/blood , Polyps/blood , Xanthophylls/blood , Aged , Carotenoids/blood , Colorectal Neoplasms/diet therapy , Colorectal Neoplasms/pathology , Endoscopy , Female , Fruit , Humans , Japan , Lutein/blood , Male , Middle Aged , Polyps/pathology , Risk Factors , Vegetables , Zeaxanthins , beta Carotene/blood
11.
Biochem Biophys Res Commun ; 439(1): 54-9, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23958298

ABSTRACT

BACKGROUND: Nobiletin is a polymethoxylated flavone that is abundant in the peels of citrus fruits, such as Citrus unshiu (Satsuma mandarin) and Citrus sinensis. The dried peels of C. unshiu (chinpi) have been included in several formulae of Japanese Kampo medicines. Nobiletin may suppress the induction of inducible nitric oxide synthase (iNOS), which synthesizes the inflammatory mediator nitric oxide (NO) in hepatocytes. METHODS: A C. unshiu peel (CUP) extract was prepared. Primary cultured rat hepatocytes were treated with the CUP extract or nobiletin in the presence of interleukin 1ß (IL-1ß), which induces iNOS expression. NO production and iNOS gene expression were analyzed. RESULTS: High-performance liquid chromatography analyses revealed that the nobiletin content in the CUP extract was 0.14%. Nobiletin dose-dependently reduced the NO levels and decreased iNOS expression at the protein, mRNA and antisense transcript levels. Flavone, which does not contain any methoxy groups, also suppressed iNOS induction. Nobiletin reduced the transcriptional activity of iNOS promoter-luciferase constructs and the DNA-binding activity of nuclear factor κB (NF-κB) in the nuclei. CONCLUSIONS: The suppression of iNOS induction by nobiletin suggests that nobiletin may be responsible for the anti-inflammatory effects of citrus peels and have a therapeutic potential for liver diseases.


Subject(s)
Citrus/chemistry , Flavones/pharmacology , Gene Expression Regulation, Enzymologic , Hepatocytes/metabolism , Interleukin-1beta/pharmacology , Nitric Oxide Synthase Type II/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Hepatocytes/drug effects , L-Lactate Dehydrogenase/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Rats
12.
Biosci Biotechnol Biochem ; 77(3): 566-71, 2013.
Article in English | MEDLINE | ID: mdl-23470763

ABSTRACT

Lactoferrin (LF) is a multifunctional glycoprotein found in mammalian milk. We have shown in a previous clinical study that enteric-coated bovine LF tablets decreased visceral fat accumulation. To address the underlying mechanism, we conducted in vitro studies and revealed the anti-adipogenic action of LF in pre-adipocytes. The aim of this study was to assess whether LF could increase the lipolytic activity in mature adipocytes. Pre-adipocytes were prepared from rat mesenteric fat and differentiated into mature adipocytes for assays of lipolysis. The addition of LF significantly increased the glycerol concentration in the medium in a dose-dependent manner, whereas pepsin-degraded LF did not. A DNA microarray analysis demonstrated that LF decreased the expression of perilipin and affected the cAMP pathway. These findings are supported by the results of quantitative RT-PCR of perilipin and assays of cAMP. These data collectively indicate that visceral fat reduction by LF may result from the promotion of lipolysis and the additional anti-adipogenic activity of LF.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Cell Differentiation , Lactoferrin/pharmacology , Lipolysis/drug effects , Adipocytes/cytology , Animals , Cattle , Lactoferrin/metabolism , Lipolysis/genetics , Male , Oligonucleotide Array Sequence Analysis , Proteolysis , Rats , Rats, Sprague-Dawley , Transcriptome/drug effects
13.
Gen Physiol Biophys ; 32(2): 261-75, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23682023

ABSTRACT

The effects of biomolecules on peripheral tissues and their responsive machinery are not well understood. We examined MDM2 level in the plasma membrane (PM) and total MDM2 level of 3T3-L1 adipocytes treated with biomolecular anandamide, epinephrine, and other agents for 15 min. We also examined biomolecular responses in cells treated with mithramycin A, a binding inhibitor, or cells exposed to cooling and cell viability. Immunoblotting revealed that PM MDM2 level increased and total MDM2 level was not altered following treatment with anandamide, epinephrine, capsaicin, CL316243, and aluminum fluoride. PM MDM2 distribution caused by a biomolecular concentration was maintained by treatment with mithramycin A and exposure of cells to 28°C or 32°C but not to 18°C, and PM MDM2 levels after treatment with high concentrations of biomolecules were altered upon exposure to the inhibitor and mild hypothermia. These conditions did not decrease cell viability. Our findings indicate that 3T3-L1 adipocytes possess molecular machinery that responds differentially to anandamide and epinephrine under the inhibitor treatment and cool temperature conditions and that is sensitive to other agents (which mimic biomolecular responses); these machineries can induce subcellular alterations in molecular interactions. We provide information helpful for clarifying biomolecular responsive machinery present in 3T3-L1 adipocytes.


Subject(s)
Arachidonic Acids/administration & dosage , Cell Membrane/drug effects , Cell Membrane/metabolism , Endocannabinoids/administration & dosage , Epinephrine/administration & dosage , Polyunsaturated Alkamides/administration & dosage , Proto-Oncogene Proteins c-mdm2/metabolism , 3T3-L1 Cells , Animals , Dose-Response Relationship, Drug , Mice , Tissue Distribution
14.
Cryobiology ; 65(1): 12-20, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22465333

ABSTRACT

Cooling induces several responses that are modulated by molecular inhibitors and activators and receptor signaling. Information regarding potential targets involved in cold response mechanisms is still insufficient. We examined levels of the receptor-signaling mediator ß-arrestin-1 and phospho-Ser-412 ß-arrestin-1 in 3T3-L1 adipocytes exposed to 4-37 °C or treated with some molecular agents at 37°C. We also cooled cells with or without modification and signal-modulating agents. These conditions did not decrease cell viability, and western blot analysis revealed that exposure to 4 °C for 1.5h and to 28 and 32 °C for 24 and 48 h increased phospho-ß-arrestin-1 and ß-arrestin-1 levels and that exposure to 4 and 18 °C for 3 and 4.5h increased ß-arrestin-1 level. Serum removal and rewarming abolished ß-arrestin-1 alterations induced by cooling. Mithramycin A (a transcription inhibitor) treatment for 4 and 24h increased the level of ß-arrestin-1 but not that of phospho-ß-arrestin-1. The level of phospho-ß-arrestin-1 was increased by okadaic acid (a phosphatase inhibitor), decreased by epinephrine and aluminum fluoride (receptor-signaling modulators), and unaffected by N-ethylmaleimide (an alkylating agent) at 37 °C. N-Ethylmaleimide and the receptor-signaling modulators did not alter ß-arrestin-1 expression at 37 °C but impaired the induction of phospho-ß-arrestin-1 at 28 and 32 °C without affecting the induction of ß-arrestin-1. We show that cold-induced ß-arrestin-1 alterations are partially mimicked by molecular agents and that the responsive machinery for ß-arrestin-1 requires serum factors and N-ethylmaleimide-sensitive sites and is linked to rewarming- and receptor signaling-responsive machinery. Our findings provide helpful information for clarifying the cold-responsive machinery for ß-arrestin-1 and elucidating low-temperature responses.


Subject(s)
Adipocytes/metabolism , Arrestins/biosynthesis , Phosphoproteins/biosynthesis , 3T3-L1 Cells , Aluminum Compounds/pharmacology , Animals , Arrestins/metabolism , Cell Line , Cell Survival/physiology , Cold Temperature , Culture Media, Serum-Free , Epinephrine/pharmacology , Ethylmaleimide/pharmacology , Fluorides/pharmacology , Mice , Okadaic Acid/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , beta-Arrestin 1 , beta-Arrestins
15.
J Biol Chem ; 285(12): 9282-91, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20081197

ABSTRACT

Myeloperoxidase (MPO) generates reactive halogenating species that can modify DNA. The aim of this study was to investigate the formation of 8-halogenated 2'-deoxyguanosines (8- halo-dGs) during inflammatory events. 8-Bromo-2'-dG (8-BrdG) and 8-chloro-2'-dG (8-CldG) were generated by treatment of MPO with hydrogen peroxide at physiological concentrations of Cl(-) and Br(-). The formation of 8-halo-dGs with other oxidative stress biomarkers in lipopolysaccharide-treated rats was assessed by liquid chromatography tandem mass spectrometry and immunohistochemistry using a novel monoclonal antibody (mAb8B3) to 8-BrdG-conjugated keyhole limpet hemocyanin. The antibody recognized both 8-BrdG and 8-CldG. In the liver of lipopolysaccharide-treated rats, immunostaining for 8-halo-dGs, halogenated tyrosines, and MPO were increased at 8 h, whereas those of 8-oxo-2'-dG (8-OxodG) and 3-nitrotyrosine were increased at 24 h. Urinary excretion of both 8-CldG and 8-BrdG was also observed earlier than those of 8-OxodG and modified tyrosines (3-nitrotyrosine, 3-chlorotyrosine, and 3- bromotyrosine). Moreover, the levels of the 8-halo-dGs in urine from human diabetic patients were 8-fold higher than in healthy subjects (n = 10, healthy and diabetic, p < 0.0001), whereas there was a moderate difference in 8-OxodG between the two groups (p < 0.001). Interestingly, positive mAb8B3 antibody staining was observed in liver tissue from hepatocellular carcinoma patients but not in liver tissue from human cirrhosis patients. These data suggest that 8-halo-dGs may be potential biomarkers of early inflammation.


Subject(s)
Deoxyguanosine/chemistry , Inflammation/pathology , 8-Hydroxy-2'-Deoxyguanosine , Animals , Antibodies, Monoclonal/chemistry , Chromatography, Liquid/methods , Deoxyguanosine/analogs & derivatives , Female , Halogens/chemistry , Lipopolysaccharides/chemistry , Liver/metabolism , Mass Spectrometry/methods , Mice , Mice, Inbred C57BL , Peroxidase/metabolism , Rats , Rats, Wistar , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/chemistry
16.
Br J Nutr ; 105(2): 200-11, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20854698

ABSTRACT

Lactoferrin (LF) is a multifunctional glycoprotein in mammalian milk. In a previous report, we showed that enteric-coated bovine LF tablets can decrease visceral fat accumulation, hypothesising that the enteric coating is critical to the functional peptides reaching the visceral fat tissue and exerting their anti-adipogenic activity. The aim of the present study was to assess whether ingested LF can retain its anti-adipogenic activity. We therefore investigated the effects of LF and LF treated with digestive enzymes (the stomach enzyme pepsin and the small intestine enzyme trypsin) on lipid accumulation in pre-adipocytes derived from the mesenteric fat tissue of male Sprague-Dawley rats. Lipid accumulation in pre-adipocytes was significantly reduced by LF in a dose-dependent manner and was associated with reduction in gene expression of CCAAT/enhancer binding protein delta, CCAAT/enhancer binding protein alpha and PPARγ as revealed by DNA microarray analysis. Trypsin-treated LF continued to show anti-adipogenic action, whereas pepsin-treated LF abrogated the activity. When an LF solution (1000 mg bovine LF) was administered by gastric intubation to Sprague-Dawley rats, immunoreactive LF determined by ELISA could be detected in mesenteric fat tissue at a concentration of 14·4 µg/g fat after 15 min. The overall results point to the importance of enteric coating for action of LF as a visceral fat-reducing agent when administered in oral form.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Lactoferrin/pharmacology , Pepsin A/pharmacology , Trypsin/pharmacology , Adipocytes/cytology , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Animals , Cattle , Female , Humans , In Vitro Techniques , Intra-Abdominal Fat/cytology , Lactoferrin/administration & dosage , Lactoferrin/pharmacokinetics , Lipid Metabolism/drug effects , Male , Obesity, Abdominal/drug therapy , Rats , Rats, Sprague-Dawley , Tablets, Enteric-Coated , Tissue Distribution
17.
Nutrients ; 13(4)2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33810214

ABSTRACT

Fucoxanthin (Fx) has preventive effect against muscle atrophy and myotube loss in vitro, but it has not yet been examined in vivo. Therefore, we aimed to investigate the effect of Fx on dexamethasone (Dex)-induced muscle atrophy and fat mass in mice. ICR mice were fed with Fx diets from 2 weeks before Dex treatment to the end of the study. Muscle atrophy was induced in the mice by oral administration of Dex. Body weight was significantly lower by Dex treatment. Visceral fat mass in the Fx-treated group were significantly lower than those in the control group. The Dex-induced decrease in tibialis anterior muscle mass was ameliorated by Fx treatment. Fx treatment significantly attenuated muscle lipid peroxidation compared with the control and Dex-treated groups. The phosphorylation of AMPK was significantly higher in the Dex-treated group than in the control group. The expression of cytochrome c oxidase (COX) IV was significantly higher in the Fx-treated group than in the control group. These results suggest that Fx may be a beneficial material to prevent muscle atrophy in vivo, in addition to the effect of fat loss.


Subject(s)
Dexamethasone/toxicity , Muscle, Skeletal/drug effects , Muscular Atrophy/chemically induced , Muscular Atrophy/prevention & control , Xanthophylls/therapeutic use , Animals , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred ICR , Muscle Proteins/genetics , Muscle Proteins/metabolism
18.
Biochim Biophys Acta ; 1790(4): 260-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19714865

ABSTRACT

BACKGROUND: The precise mechanism of the anti-tumor action of fucoxanthin has yet to be elucidated. We previously reported that gadd45a and gadd45b might play a role in the G1 arrest induced by fucoxanthin. In the present study, we show that several MAPKs modulate the induction of gadd45 and G1 arrest METHODS: HepG2 and DU145 cells were used. The cell cycle was analyzed using flow cytometry. Expression of gadd45 was assayed by Northern blot and/or quantitative RT-PCR analyses. Activation of MAPK was assayed by Western blot analysis. RESULTS: Inhibition of p38 MAPK enhanced the induction ofgadd45a expression and G1 arrest by fucoxanthin in HepG2 cells. Inhibition of ERK enhanced gadd45b expression but had no effect on the induction of G1 arrest by fucoxanthin in HepG2 cells. Inhibition of SAPK/JNK suppressed the induction of gadd45a expression and G1 arrest by fucoxanthin in DU145 cells. These data suggest that gadd45a is closely related with the G1 arrest induced by fucoxanthin, and that the pattern of MAPK involvement in the induction of gadd45a and G1 arrest by fucoxanthin differs depending on the cell type. GENERAL SIGNIFICANCE: The implication of GADD45 and MAPK involvement in the anti-tumor action of carotenoids is first described.


Subject(s)
Cell Cycle Proteins/biosynthesis , Cell Cycle/drug effects , G1 Phase/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins/biosynthesis , Xanthophylls/pharmacology , Cell Cycle/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans
19.
Br J Nutr ; 104(11): 1688-95, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20691130

ABSTRACT

Lactoferrin (LF), a multifunctional glycoprotein in mammalian milk, is reported to exert a modulatory effect on lipid metabolism. The aim of the present study was to elucidate whether enteric-coated LF (eLF) might improve visceral fat-type obesity, an underlying cause of the metabolic syndrome. Using a double-blind, placebo-controlled design, Japanese men and women (n 26; aged 22-60 years) with abdominal obesity (BMI>25 kg/m2, and visceral fat area (VFA)>100 cm2) consumed eLF (300 mg/d as bovine LF) or placebo tablets for 8 weeks. Measurement of the total fat area, VFA and subcutaneous fat area from computed tomography images revealed a significant reduction in VFA ( - 14.6 cm2) in the eLF group, as compared with the placebo controls ( - 1.8 cm2; P = 0.009 by ANCOVA). Decreases in body weight, BMI and hip circumference in the eLF group ( - 1.5 kg, - 0.6 kg/m2, - 2.6 cm) were also found to be significantly greater than with the placebo (+1.0 kg, +0.3 kg/m2, - 0.2 cm; P = 0.032, 0.013, 0.041, respectively). There was also a tendency for a reduction in waist circumference in the eLF group ( - 4.4 cm) as compared with the placebo group ( - 0.9 cm; P = 0.073). No adverse effects of the eLF treatment were found with regard to blood lipid or biochemical parameters. From these results, eLF appears to be a promising agent for the control of visceral fat accumulation.


Subject(s)
Adiposity/drug effects , Anti-Obesity Agents/therapeutic use , Body Weight/drug effects , Intra-Abdominal Fat/drug effects , Lactoferrin/therapeutic use , Obesity, Abdominal/drug therapy , Adult , Anti-Obesity Agents/pharmacology , Asian People , Body Mass Index , Double-Blind Method , Female , Hip/anatomy & histology , Humans , Intra-Abdominal Fat/metabolism , Lactoferrin/pharmacology , Male , Middle Aged , Tablets, Enteric-Coated , Tomography, X-Ray Computed , Waist Circumference/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL