Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Heliyon ; 9(4): e14777, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37025807

ABSTRACT

The extracellular matrix (ECM) components present within all tissues and organs help to maintain the cytoskeletal architecture and tissue morphology. Although the ECM plays a role in cellular events and signaling pathways, it has not been well studied due its insolubility and complexity. Brain tissue has a higher cell density and weaker mechanical strength than other tissues in the body. When removing cells using a general decellularization method to produce scaffolds and obtain ECM proteins, various problems must be considered because tissues are easily damaged. To retain the brain shape and ECM components, we performed decellularization in combination with polymerization. We immersed mouse brains in oil for polymerization and decellularization via O-CASPER (Oil-based Clinically and Experimentally Applicable Acellular Tissue Scaffold Production for Tissue Engineering and Regenerative Medicine) and then isolated ECM components using sequential matrisome preparation reagents (SMPRs), namely, RIPA, PNGase F, and concanavalin A. Adult mouse brains were preserved with our decellularization method. Western blot and LC-MS/MS analyses revealed that ECM components, including collagen and laminin, were isolated efficiently from decellularized mouse brains using SMPRs. Our method will be useful to obtain matrisomal data and perform functional studies using adult mouse brains and other tissues.

2.
Int J Mol Sci ; 22(1)2021 Jan 02.
Article in English | MEDLINE | ID: mdl-33401721

ABSTRACT

Mitochondrial dysfunction contributes to neurodegenerative diseases and developmental disorders such as Fragile X syndrome (FXS). The cross-talk between mitochondria and extracellular vesicles (EVs) suggests that EVs may transfer mitochondrial components as intermediators for intracellular communication under physiological and pathological conditions. In the present study, the ability of EVs to transfer mitochondrial components and their role in mitochondrial dysfunction in astrocytes were examined in the brains of Fmr1 knockout (KO) mice, a model of FXS. The amounts of mitochondrial transcription factor NRF-1, ATP synthases ATP5A and ATPB, and the mitochondrial membrane protein VDAC1 in EVs were reduced in cerebral cortex samples and astrocytes from Fmr1 KO mice. These reductions correspond to decreased mitochondrial biogenesis and transcriptional activities in Fmr1 KO brain, along with decreased mitochondrial membrane potential (MMP) with abnormal localization of vimentin intermediate filament (VIF) in Fmr1 KO astrocytes. Our results suggest that mitochondrial dysfunction in astrocytes is associated with the pathogenesis of FXS and can be monitored by depletion of components in EVs. These findings may improve the ability to diagnose developmental diseases associated with mitochondrial dysfunction, such as FXS and autism spectrum disorders (ASD).


Subject(s)
Astrocytes/metabolism , Extracellular Vesicles/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Mitochondria/metabolism , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Disease Models, Animal , Extracellular Vesicles/genetics , Extracellular Vesicles/ultrastructure , Fragile X Mental Retardation Protein/genetics , Immunohistochemistry , Male , Membrane Potential, Mitochondrial/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/genetics
3.
Mar Drugs ; 17(11)2019 Oct 27.
Article in English | MEDLINE | ID: mdl-31717879

ABSTRACT

Metabolic disorders such as diabetes and obesity are serious global health issues. These diseases are accelerated by mineral deficiencies, emphasizing the importance of addressing these deficiencies in disease management plans. Lactate metabolism is fundamentally linked to glucose metabolism, and several clinical studies have reported that blood lactate levels are higher in obese and diabetic patients than in healthy subjects. Balanced deep-sea water contains various minerals and exhibits antiobesity and antidiabetic activities in mice; however, the impact of balanced deep-sea water on lactate metabolism is unclear. Thus, we evaluated the effects of balanced deep-sea water on lactate metabolism in C2C12 myotubes, and found that balanced deep-sea water mediated lactate metabolism by regulating the gene expression levels of lactate dehydrogenases A and B, a monocarboxylate transporter, and a mitochondrial pyruvate carrier. The activities of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and signaling molecules involved in PGC-1α activation were also upregulated by treatment with balanced deep-sea water. These results suggest that balanced deep-sea water, which can mediate lactate metabolism, may be used to prevent or treat obesity and diabetes mellitus.


Subject(s)
Lactates/metabolism , Minerals/administration & dosage , Muscle Fibers, Skeletal/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Cell Line , Gene Expression Regulation/drug effects , Isoenzymes/genetics , L-Lactate Dehydrogenase/genetics , Lactate Dehydrogenase 5/genetics , Metabolic Diseases/drug therapy , Metabolic Diseases/physiopathology , Mice , Minerals/pharmacology , Muscle Fibers, Skeletal/metabolism , Obesity/drug therapy , Obesity/physiopathology , Seawater/chemistry , Signal Transduction/drug effects
4.
Int J Oncol ; 51(3): 859-866, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28713989

ABSTRACT

Proton beam therapy has recently been used to improve local control of tumor growth and reduce side-effects by decreasing the global dose to normal tissue. However, the regulatory mechanisms underlying the physiological role of proton beam radiation are not well understood, and many studies are still being conducted regarding these mechanisms. To determine the effects of proton beams on mitochondrial biogenesis, we investigated: mitochondrial DNA (mtDNA) mass; the gene expression of mitochondrial transcription factors, functional regulators, and dynamic-related regulators; and the phosphorylation of the signaling molecules that participate in mitochondrial biogenesis. Both the mtDNA/nuclear DNA (nDNA) ratio and the mitochondria staining assays showed that proton beam irradiation increases mitochondrial biogenesis in 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced aggressive HT-29 cells. Simultaneously, proton beam irradiation increases the gene expression of the mitochondrial transcription factors PGC-1α, NRF1, ERRα, and mtTFA, the dynamic regulators DRP1, OPA1, TIMM44, and TOM40, and the functional regulators CytC, ATP5B and CPT1-α. Furthermore, proton beam irradiation increases the phosphorylation of AMPK, an important molecule involved in mitochondrial biogenesis that is an energy sensor and is regulated by the AMP/ATP ratio. Based on these findings, we suggest that proton beam irradiation inhibits metastatic potential by increasing mitochondrial biogenesis and function in TPA-induced aggressive HT-29 cells.


Subject(s)
Adenocarcinoma/radiotherapy , Colorectal Neoplasms/radiotherapy , DNA, Mitochondrial/radiation effects , Organelle Biogenesis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/radiation effects , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/radiation effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/radiation effects , HT29 Cells , Humans , Neoplasm Proteins/radiation effects , Phosphorylation/radiation effects , Proton Therapy , Tetradecanoylphorbol Acetate/toxicity
5.
Biomed Pharmacother ; 83: 477-484, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27434863

ABSTRACT

Recent studies showed that deficiencies of essential minerals including Mg, Ca, and K, and trace minerals including Se, Zn, and V, have implications for the development, prevention, and treatment of several chronic diseases including obesity and type 2 diabetes. Our previous studies revealed that balanced deep-sea water (BDSW), which is composed of desalinated water enriched with Mg and Ca, has potential as a treatment for diabetes and obesity. In this study, to determine whether BDSW regulates mitochondrial biogenesis and function, we investigated its effects on mitochondrial DNA (mtDNA) content, mitochondrial enzyme activity, expression of key transcription factors and mitochondria-specific genes, phosphorylation of signaling molecules associated with mitochondrial biogenesis, and mitochondrial function in 3T3-L1 preadipocytes. BDSW increased mitochondrial biogenesis in a dose-dependent manner. Quantitative real-time PCR revealed that BDSW enhances expression of PGC1-α, NRF1, and TFAM genes. Upregulation of these genes was supported by increased mitochondria staining, CytC oxidase activity, and AMPK phosphorylation. The stimulatory effect of BDSW on mitochondrial biogenesis and function suggests a novel mechanism for BDSW-induced anti-diabetic and anti-obesity action.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipocytes/metabolism , Calcium/pharmacology , Magnesium/pharmacology , Mitochondria/metabolism , Organelle Biogenesis , Seawater/chemistry , Signal Transduction/drug effects , 3T3-L1 Cells , Adipocytes/drug effects , Animals , DNA, Mitochondrial/metabolism , Gene Expression Regulation/drug effects , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Phosphorylation/drug effects
6.
Mar Biotechnol (NY) ; 18(4): 475-84, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27215753

ABSTRACT

Deep-sea water (DSW) and chitosan oligosaccharides (COS) have recently drawn much attention because of their potential medical and pharmaceutical applications. Balanced DSW (BDSW) was prepared by mixing DSW mineral extracts and desalinated water. This study investigated the effects of BDSW, COS, and BDSW containing COS on glucose uptake and their mode of action in mature C2C12 myotubes. BDSW and COS increased glucose uptake in a dose-dependent manner. BDSW containing COS synergistically increased glucose uptake; this was dependent on the activation of insulin receptor substrate 1 and protein kinase C in insulin-dependent signaling pathways as well as liver kinase B1, AMP-activated protein kinase, and mammalian target of rapamycin in insulin-independent signaling pathways. Quantitative real-time polymerase chain reaction revealed that the expressions of the following genes related to glucose uptake were elevated: glucose transporter 4 (GLUT4), insulin-responsive aminopeptidase, and vesicle-associated membrane protein 2 for abundant proteins of GLUT4 storage vesicles (GSVs); syntaxin 4 and soluble N-ethylmaleimide-sensitive factor attachment protein 23 for trafficking between the plasma membrane and GSVs; and syntaxin 6 and syntaxin 16 for trafficking between GSVs and the trans-Golgi network. Taken together, these results suggest BDSW containing COS has a greater stimulatory effect on glucose uptake than BDSW or COS alone. Moreover, this effect is mediated by the stimulation of diverse signaling pathways via the activation of main signaling molecules related to GSV trafficking.


Subject(s)
Chitosan/pharmacology , Gene Expression Regulation/drug effects , Glucose/metabolism , Muscle Fibers, Skeletal/drug effects , Oligosaccharides/pharmacology , Seawater/chemistry , Animals , Biological Transport/drug effects , Cell Line , Culture Media/chemistry , Culture Media/pharmacology , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Qb-SNARE Proteins/genetics , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/metabolism , Salinity , Signal Transduction
7.
PLoS One ; 10(6): e0129972, 2015.
Article in English | MEDLINE | ID: mdl-26068191

ABSTRACT

The worldwide prevalence of metabolic diseases, including obesity and diabetes, is increasing. Mitochondrial dysfunction is recognized as a core feature of these diseases. Emerging evidence also suggests that defects in mitochondrial biogenesis, number, morphology, fusion, and fission, contribute to the development and progression of metabolic diseases. Our previous studies revealed that balanced deep-sea water (BDSW) has potential as a treatment for diabetes and obesity. In this study, we aimed to investigate the mechanism by which BDSW regulates diabetes and obesity by studying its effects on mitochondrial metabolism. To determine whether BDSW regulates mitochondrial biogenesis and function, we investigated its effects on mitochondrial DNA (mtDNA) content, mitochondrial enzyme activity, and the expression of transcription factors and mitochondria specific genes, as well as on the phosphorylation of signaling molecules associated with mitochondria biogenesis and its function in C2C12 myotubes. BDSW increased mitochondrial biogenesis in a time and dose-dependent manner. Quantitative real-time PCR revealed that BDSW enhances gene expression of PGC-1α, NRF1, and TFAM for mitochondrial transcription; MFN1/2 and DRP1 for mitochondrial fusion; OPA1 for mitochondrial fission; TOMM40 and TIMM44 for mitochondrial protein import; CPT-1α and MCAD for fatty acid oxidation; CYTC for oxidative phosphorylation. Upregulation of these genes was validated by increased mitochondria staining, CS activity, CytC oxidase activity, NAD+ to NADH ratio, and the phosphorylation of signaling molecules such as AMPK and SIRT1. Moreover, drinking BDSW remarkably improved mtDNA content in the muscles of HFD-induced obese mice. Taken together, these results suggest that the stimulatory effect of BDSW on mitochondrial biogenesis and function may provide further insights into the regulatory mechanism of BDSW-induced anti-diabetic and anti-obesity action.


Subject(s)
Mitochondria, Muscle/drug effects , Organelle Biogenesis , Seawater , Water/pharmacology , Animals , Cell Line , Mice , Mice, Inbred C57BL , Mitochondria, Muscle/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Oxidative Phosphorylation , Up-Regulation , Water/chemistry
8.
Int J Oncol ; 46(6): 2621-8, 2015.
Article in English | MEDLINE | ID: mdl-25845382

ABSTRACT

Proton radiotherapy has been established as a highly effective modality used in the local control of tumor growth. Although proton radiotherapy is used worldwide to treat several types of cancer clinically with great success due to superior targeting and energy deposition, the detailed regulatory mechanisms underlying the functions of proton radiation are not yet well understood. Accordingly, in the present study, to assess the effects of proton beam on integrin-mediated signaling pathways, we investigated the expression of integrins related to tumor progression and integrin trafficking, and key molecules related to cell adhesion, as well as examining phosphorylation of signaling molecules involved in integrin-mediated signaling pathways. Proton beam irradiation inhibited the increase in 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced integrin ß1 protein expression and the gene expression of members of the integrin family, such as α5ß1, α6ß4, αvß3, and αvß6 in human colorectal adenocarcinoma HT-29 cells. Simultaneously, the gene expression of cell adhesion molecules, such as FAK and CDH1, and integrin trafficking regulators, such as RAB4, RAB11, and HAX1, was decreased by proton beam irradiation. Moreover, proton beam irradiation decreased the phosphorylation of key molecules involved in integrin signaling, such as FAK, Src, and p130Cas, as well as PKC and MAPK, which are known as promoters of cell migration, while increased the phosphorylation of AMPK and the gene expression of Rab IP4 involved in the inhibition of cell adhesion and cell spreading. Taken together, our findings suggest that proton beam irradiation can inhibit metastatic potential, including cell adhesion and migration, by modulating the gene expression of molecules involved in integrin trafficking and integrin-mediated signaling, which are necessary for tumor progression.


Subject(s)
Colonic Neoplasms/metabolism , Integrins/metabolism , Signal Transduction/radiation effects , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Cell Adhesion/radiation effects , Cell Movement/radiation effects , Colonic Neoplasms/genetics , Colonic Neoplasms/radiotherapy , Gene Expression Regulation, Neoplastic/radiation effects , HT29 Cells , Humans , Integrins/genetics , Proton Therapy , Tetradecanoylphorbol Acetate/toxicity
9.
Oncol Lett ; 9(1): 483-487, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25436013

ABSTRACT

Colorectal adenocarcinoma is the most common type of gastrointestinal cancer. Colon adenocarcinoma is a major health problem worldwide due to the high prevalence and mortality rates associated with the disease. The majority of colorectal carcinomas are adenocarcinomas, which originate from the epithelial cells of the colorectal mucosa. HT-29 cells, which originate from human colon adenocarcinoma, are used as an in vitro model to investigate the effect of malignant transformation on the expression of cellular constituents and functions of the intestinal epithelium. Nitric oxide (NO) is a signaling molecule, which is involved in inflammation and carcinogenesis. It has been reported that enhanced inducible NO synthase (iNOS) activity and the resulting NO concentrations in human colon carcinoma contribute to tumor progression and vascular invasion. The present study investigates the effect of pro-inflammatory cytokine-induced nitric oxide (NO) production and iNOS expression on the invasion of human colorectal adenocarcinoma HT-29 cells, and the effect of extract from Cnidii Rhizoma on NO production and the invasiveness of HT-29 cells. Treatment of HT-29 cells with cytokines, 100 U/ml interferon γ, 10 ng/ml interleukin-1 α and 25 ng/ml tumor necrosis factor α was found to increase NO production. Pretreatment of the cells with Cnidii Rhizoma (0.1-5 mg/ml) resulted in an inhibition of cytokine-induced NO production and iNOS expression. The invasiveness of HT-29 cells through Matrigel was significantly increased by treatment with cytokines. Cnidii Rhizoma inhibited the invasiveness of cytokine-treated HT-29 cells through the Matrigel-coated membrane in a concentration-dependent manner. Matrix metalloproteinase (MMP) activity in HT-29 cells increased following the treatment with cytokines, and pretreatment of the cells with Cnidii Rhizoma inhibited cytokine-induced MMP-2 activity. These results provide sufficient information for the further development of Cnidii Rhizoma as an antitumor metastatic agent for the treatment of colon cancer.

10.
PLoS One ; 9(7): e102095, 2014.
Article in English | MEDLINE | ID: mdl-25013896

ABSTRACT

The aim of this study was to determine the effects of balanced deep-sea water (BDSW) on hyperglycemia and glucose intolerance in streptozotocin (STZ)-induced diabetic mice. BDSW was prepared by mixing DSW mineral extracts and desalinated water to yield a final hardness of 1000-4000 ppm. Male ICR mice were assigned to 6 groups; mice in each group were given tap water (normal and STZ diabetic groups) or STZ with BDSW of varying hardness (0, 1000, 2000, and 4000 ppm) for 4 weeks. The STZ with BDSW group exhibited lowered fasting plasma glucose levels than the STZ-induced diabetic group. Oral glucose tolerance tests showed that BDSW improves impaired glucose tolerance in STZ-induced diabetic mice. Histopathological evaluation of the pancreas showed that BDSW restores the morphology of the pancreatic islets of Langerhans and increases the secretion of insulin in STZ-induced diabetic mice. Quantitative real-time PCR assay revealed that the expression of hepatic genes involved in gluconeogenesis, glucose oxidation, and glycogenolysis was suppressed, while the expression of the genes involved in glucose uptake, ß-oxidation, and glucose oxidation in muscle were increased in the STZ with BDSW group. BDSW stimulated PI3-K, AMPK, and mTOR pathway-mediated glucose uptake in C2C12 myotubes. BDSW increased AMPK phosphorylation in C2C12 myotubes and improved impaired AMPK phosphorylation in the muscles of STZ-induced diabetic mice. Taken together, these results suggest that BDSW is a potential anti-diabetic agent, owing to its ability to suppress hyperglycemia and improve glucose intolerance by modulating glucose metabolism, recovering pancreatic islets of Langerhans and increasing glucose uptake.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Hyperglycemia/metabolism , Seawater , Animals , Blood Glucose/drug effects , Glucose Tolerance Test , Hypoglycemic Agents/therapeutic use , Male , Mice , Mice, Inbred ICR
11.
Biofactors ; 40(4): 436-47, 2014.
Article in English | MEDLINE | ID: mdl-24756979

ABSTRACT

Many active components derived from edible natural resources such as plant extracts have recently attracted attention for their potential use as functional foods or drugs for preventing and treating metabolic diseases such as diabetes. To obtain a novel modulator of glucose metabolism, we conducted screening of a small compound library in cultured L6 myotubes. We identified nepodin that stimulated glucose uptake dose-dependently in differentiated L6 myotubes. The stimulatory effect of nepodin on glucose uptake was abrogated by a 5'-adenosine monophosphate-activated protein kinase (AMPK) inhibitor. In addition, nepodin stimulated the phosphorylation of AMPK. Nepodin also stimulated the translocation of GLUT4 to the plasma membrane in L6 myoblasts transfected with a Glut4 cDNA-coding vector and in differentiated L6 myotubes. In in vivo study, nepodin suppressed the increases in fasting blood glucose levels and improved the glucose intolerance of C57BL/KsJ-db/db mice, a type 2 diabetic animal model. Nepodin rescued the impaired phosphorylation of AMPK in the skeletal muscle of db/db mice. These results suggest that nepodin has an antidiabetic effect, which is at least partly mediated by stimulation of GLUT4 translocation via AMPK activation by nepodin.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Naphthalenes/pharmacology , Adenylate Kinase/metabolism , Animals , Blood Glucose , Cell Line , Diabetes Mellitus, Type 2/blood , Drug Evaluation, Preclinical , Gene Expression/drug effects , Glucose/metabolism , Glucose Intolerance , Glucose Transporter Type 4/metabolism , Hypoglycemic Agents/therapeutic use , Insulin/blood , Insulin Resistance , Lipid Metabolism/drug effects , Liver/drug effects , Liver/enzymology , Male , Mice, Inbred C57BL , Mice, Obese , Naphthalenes/therapeutic use , Plant Roots/chemistry , Protein Processing, Post-Translational/drug effects , Protein Transport , Rats , Rumex/chemistry , Signal Transduction/drug effects
12.
Obesity (Silver Spring) ; 22(7): 1669-78, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24634394

ABSTRACT

OBJECTIVE: To determine the effects of balanced deep-sea water (BDSW) on adipocyte hypertrophy and liver steatosis in high-fat diet (HFD)-induced obese C57BL/6J mice. METHODS: BDSW was prepared by mixing deep-sea water (DSW) mineral extracts and desalinated water. C57BL/6J mice were fed a normal diet or HFD with or without BDSW with different hardness (500, 1000, or 2000) for 20 weeks. RESULTS: BDSW suppressed body weight gain in HFD-fed mice. Histopathologic assays of the fat and liver revealed that BDSW inhibited the increase in adipocyte size and improved severe liver steatosis in HFD-fed mice. BDSW suppressed the expression of adipogenic, lipogenic, lipolytic, and pro-inflammatory cytokine genes and increased the expression of adipokines and ß-oxidation genes in fat. In the liver, BDSW suppressed the expression of genes involved in lipogenesis and cholesterol synthesis, and increased the expression of genes related to ß-oxidation. Furthermore, BDSW improved the impaired phosphorylation of IRS-1, LKB1, AMPK, and mTOR in fat and liver tissues of HFD-fed mice. CONCLUSIONS: These results suggest that BDSW has potential as an anti-lipidemic agent, given its ability to suppress body weight gain and liver steatosis through the regulation of lipid metabolism by signal molecule activation.


Subject(s)
Adipocytes/drug effects , Adipocytes/pathology , Diet, High-Fat , Fatty Liver/pathology , Seawater , Adipogenesis/drug effects , Animals , Cell Size/drug effects , Hypertrophy , Lipid Metabolism , Lipogenesis/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Water , Weight Gain/drug effects
13.
Mar Drugs ; 11(11): 4193-212, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24172214

ABSTRACT

In this study, we investigated the effects of balanced deep-sea water (BDSW) on hyperglycemia and glucose intolerance in high-fat diet (HFD)-induced diabetic C57BL/6J mice. BDSW was prepared by mixing deep-sea water (DSW) mineral extracts and desalinated water to give a final hardness of 500-2000. Mice given an HFD with BDSW showed lowered fasting plasma glucose levels compared to HFD-fed mice. Oral and intraperitoneal glucose tolerance tests showed that BDSW improves impaired glucose tolerance in HFD-fed mice. Histopathological evaluation of the pancreas showed that BDSW recovers the size of the pancreatic islets of Langerhans, and increases the secretion of insulin and glucagon in HFD-fed mice. Quantitative reverse transcription polymerase chain reaction results revealed that the expression of hepatic genes involved in glucogenesis, glycogenolysis and glucose oxidation were suppressed, while those in glucose uptake, ß-oxidation, and glucose oxidation in muscle were increased in mice fed HFD with BDSW. BDSW increased AMP-dependent kinase (AMPK) phosphorylation in 3T3-L1 pre- and mature adipocytes and improved impaired AMPK phosphorylation in the muscles and livers of HFD-induced diabetic mice. BDSW stimulated phosphoinositol-3-kinase and AMPK pathway-mediated glucose uptake in 3T3-L1 adipocytes. Taken together, these results suggest that BDSW has potential as an anti-diabetic agent, given its ability to suppress hyperglycemia and improve glucose intolerance by increasing glucose uptake.


Subject(s)
Diabetes Mellitus/metabolism , Diet, High-Fat/adverse effects , Hypoglycemic Agents/pharmacology , Seawater , Water/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Blood Glucose/drug effects , Cell Line , Glucagon/metabolism , Glucose/metabolism , Glucose Tolerance Test/methods , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Signal Transduction/drug effects , Water/metabolism
14.
J Immunoassay Immunochem ; 33(2): 166-79, 2012.
Article in English | MEDLINE | ID: mdl-22471607

ABSTRACT

We used a ProteoChip coated with a calixcrown derivative protein linker to measure adiponectin and monocyte chemoattractant protein-1 (MCP-1) levels and compared the results with commercial enzyme-linked immunosorbent assay (ELISA) kits. Adiponectin and MCP-1 levels in normal human serum and RAW264 cell supernatants, respectively, were measured. The ProteoChip quantification results correlated with those from the ELISA kits; however, the ProteoChip required less sample volume, exhibited higher sensitivity, and had a wider detection range. The ProteoChip was capable of detecting and quantifying small amounts of protein, possibly replacing ELISA kits in evaluating the levels of adiponectin and MCP-1.


Subject(s)
Adiponectin/analysis , Calixarenes/chemistry , Chemokine CCL2/analysis , Crown Ethers/chemistry , Protein Array Analysis/methods , Adiponectin/immunology , Animals , Antibodies, Monoclonal/immunology , Calixarenes/immunology , Cell Line , Chemokine CCL2/immunology , Crown Ethers/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Protein Array Analysis/instrumentation , Recombinant Proteins/analysis , Recombinant Proteins/immunology , Sensitivity and Specificity
15.
J Nutr Biochem ; 23(5): 501-9, 2012 May.
Article in English | MEDLINE | ID: mdl-21684132

ABSTRACT

Genistein, an isoflavone, is known to possess diverse biological functions such as antioxidative and anti-inflammatory actions. It also acts like estrogen and inhibits several tyrosine kinases. Genistein was reported to suppress insulin-mediated glucose uptake in adipocytes. In this study, we investigated the effects of genistein on glucose uptake in vitro and in vivo as well as the mechanisms associated with the glucose uptake. We found that genistein decreased nonfasting blood glucose levels in KK-Ay/Ta Jcl mice, a type 2 diabetic animal model. It also dose-dependently induced insulin secretion by Rin-5F cells. In L6 myotubes, it directly stimulated glucose uptake independently of insulin under normal and high glucose conditions in dose-dependent manners. It promoted the translocation of glucose transporter 4 to the cell membrane under both glucose conditions. Based on studies using inhibitors of signaling molecules related to glucose uptake, the stimulatory effect of genistein on glucose uptake appeared to be dependent on the phosphatidylinositol 3-kinase, mammalian target of rapamycin, protein kinase C and 5'-adenosine-monophosphate-activated protein kinase pathway under both glucose conditions. In addition, O-GlcNAcylation by O-(2-acetamido-2-deoxy-D-glucopyranosylidene) amino N-phenyl carbamate, an inhibitor of N-acetylglucosaminidase, reduced the stimulatory effect of genistein on glucose uptake under both glucose conditions. Taken together, genistein may regulate glucose uptake by increasing the phosphorylation and decreasing the O-GlcNAcylation of proteins related to glucose homeostasis.


Subject(s)
Genistein/pharmacology , Glucose/metabolism , Animals , Biological Transport , Cell Membrane/metabolism , Cells, Cultured , Glucose Transporter Type 4/metabolism , Male , Mice , Mice, Inbred BALB C
16.
J Nutr Biochem ; 22(1): 8-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20233653

ABSTRACT

Flavonoids, a group of polyphenolic compounds abundant in plants, are known to prevent bone loss in ovariectomized (OVX) animal models. Inhibition of osteoclast differentiation and bone resorption is considered as an effective therapeutic approach in the treatment of postmenopausal bone loss. Luteolin, a plant flavonoid, has potent anti-inflammatory properties both in vivo and vitro. In this study, we found that luteolin markedly decreased the differentiation of both bone marrow mononuclear cells and Raw264.7 cells into osteoclasts. Luteolin also inhibited the bone resorptive activity of differentiated osteoclasts. We further investigated the effects of luteolin on ovariectomy-induced bone loss using micro-computed tomography, biomechanical tests and serum markers assay for bone remodeling. Oral administration of luteolin (5 and 20 mg/kg per day) to OVX mice caused significant increase in bone mineral density and bone mineral content of trabecular and cortical bones in the femur as compared to those of OVX controls, and prevented decreases of bone strength indexes induced by OVX surgery. Serum biochemical markers assays revealed that luteolin prevents OVX-induced increases in bone turnover. These data strongly suggest that luteolin has the potential for prevention of bone loss in postmenopausal osteoporosis by reducing both osteoclast differentiation and function.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Resorption/prevention & control , Cell Differentiation/drug effects , Luteolin/pharmacology , Luteolin/therapeutic use , Osteoclasts/drug effects , Osteoporosis, Postmenopausal/prevention & control , Animals , Biomarkers/blood , Biomarkers/metabolism , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Bone and Bones/metabolism , Cell Line, Transformed , Cells, Cultured , Dose-Response Relationship, Drug , Female , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Luteolin/administration & dosage , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Osteoclasts/cytology , Osteoclasts/physiology , Osteoporosis, Postmenopausal/blood , Osteoporosis, Postmenopausal/diagnostic imaging , Osteoporosis, Postmenopausal/metabolism , Ovariectomy , Radiography , Random Allocation
17.
Proteomics ; 8(13): 2625-39, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18600791

ABSTRACT

Osteoclast formation and bone resorption are multiple processes that involve the participation of specialized membrane structures and their associated proteins. In this study, we used an MS to analyze the profile of proteins associated with osteoclast membranes and focused on the function of channel proteins in osteoclast differentiation and function. We filtered out with a SEQUEST score greater than 10 and a peptide hit number of more than 2, resulting in the identification of 499 proteins that were commonly found in both macrophages and osteoclasts, 96 proteins selectively found in osteoclasts, and 179 proteins selectively found in macrophages. The proteins that were selectively found in osteoclasts were classified based on their localizations: plasma membrane (17%), ER/Golgi and lysosome/endosome (15%), mitochondrion (18%), nucleus (13%), cytosol (19%), and unknown (18%). Proteins associated with osteoclast function such as v-ATPase, IGF2R, TRAP, and cathepsin K were found in osteoclasts as previously shown. We found several ion channel proteins such as Ank and Nhedc2 and signaling molecules such as Dock5 and RAB-10 in osteoclasts. Inhibition of the Na(+)/H(+) exchanger family by amiloride suppressed RANKL-induced osteoclast fusion and bone resorption. In addition, shRNA for Nhedc2 inhibited osteoclast differentiation. Our results provide a proteomic profile of osteoclast membrane proteins and identify Nhedc2, which is probably associated with proton transport in osteoclasts, as a regulator of osteoclast function.


Subject(s)
Cell Fusion , Membrane Proteins/analysis , Osteoclasts/cytology , Osteoclasts/physiology , Sodium-Hydrogen Exchangers/chemistry , Acid Phosphatase/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Fractionation , Cell Line , Chromatography, Liquid , Culture Media/pharmacology , Indicators and Reagents/metabolism , Isoenzymes/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Membrane Proteins/classification , Membrane Proteins/physiology , Mice , Mice, Inbred ICR , Protein Structure, Tertiary , RANK Ligand/pharmacology , Rosaniline Dyes/metabolism , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Tartrate-Resistant Acid Phosphatase , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL