Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Curr Oncol ; 30(4): 4197-4207, 2023 04 17.
Article in English | MEDLINE | ID: mdl-37185433

ABSTRACT

Emerging evidence highlights the important impact of early-life exposures on cancer development later in life. The present study aimed to investigate the impacts of a high-fat diet in early life on the mammary microenvironment in relation to breast tumorigenesis. Forty-four female C57BL/6 mice were fed a low-fat diet (LF, 10 kcal% fat) or a high-fat diet (HF, 60 kcal% fat) for 8 weeks starting at ~4 weeks of age. Twenty-two mice were sacrificed immediately after an 8 week feeding, and the rest of mice were switched to a normal diet for maintenance (Lab Diet, #5P76) for additional 12 weeks. A panel of metabolic parameters, inflammatory cytokines, as well as tumorigenic Wnt-signaling target genes were analyzed. The HF diet increased body weight and exacerbated mammary metabolic and inflammatory status. The disrupted microenvironment remains significant to the later life equivalent to young adulthood (p < 0.05). Mammary Wnt-signaling was elevated right after the HF diet as indicated by the upregulated expression of its downstream genes, whereas it was surprisingly suppressed after switching diets (p < 0.05). In summary, HF-induced overweight/obesity in early life altered the mammary metabolic and inflammatory microenvironments in favor of breast tumorigenesis, although its overall impact to breast cancer later in life warrants further investigation.


Subject(s)
Diet, High-Fat , Obesity , Mice , Female , Humans , Animals , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Body Weight , Carcinogenesis/metabolism , Tumor Microenvironment
2.
J Nutr ; 151(10): 2967-2975, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34383942

ABSTRACT

BACKGROUND: Adaptive thermogenesis is an iron-demanding pathway, significantly contributing to whole-body energy expenditure. However, the effects of iron-deficient diets on adaptive thermogenesis and obesity remain unknown. OBJECTIVES: We aimed to determine the impact of dietary iron deficiency on iron homeostasis in adipocytes, adaptive thermogenic capacity, and metabolic consequences in obesity. METHODS: C57BL/6 male mice were assigned to either the iron-adequate (IA, 35 ppm) or the iron-deficient group (ID, 3 ppm) at weaning. Upon 8 wk of age, both IA and ID groups received an isocaloric high-fat diet (45% kcal from fat) for 10 wk, maintaining the same iron content. Mice (n = 8) were used to determine the iron status at the systemic and tissue levels and lipid metabolism and inflammatory signaling in adipose tissue. The same mice were used to evaluate cold tolerance (4°C) for 3 h. For assessing adaptive thermogenesis, mice (n = 5) received an intraperitoneal injection of ß3-adrenoceptor agonist CL316243 (CL) for 5 d. RESULTS: Compared with the IA group, the ID group had nonanemic iron deficiency, lower serum ferritin (42.8%, P < 0.01), and greater weight gain (8.67%, P < 0.05) and insulin resistance (159%, P < 0.01), partly due to reduced AMP-activated protein kinase activation (61.0%, P < 0.05). Upon cold exposure, the ID group maintained a core body temperature 2°C lower than the IA group. The ID group had lower iron content (47.0%, P < 0.01) in the inguinal adipose tissue (iWAT) than the IA group, which was associated with impaired adaptive thermogenesis. In response to CL, ID mice showed decreased heat production (P < 0.01) and defective upregulation of beige adipocyte-specific markers, including uncoupling protein 1 (41.1%, P < 0.001), transferrin receptor 1 (47.5%, P < 0.001), and mitochondrial respiratory chain complexes (P < 0.05) compared with IA mice. CONCLUSIONS: Dietary iron deficiency deregulates iron balance in the iWAT and impairs adaptive thermogenesis, thereby escalating the diet-induced weight gain in C57BL/6 mice.


Subject(s)
Adipose Tissue, White , Iron Deficiencies , Adipocytes , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat/adverse effects , Energy Metabolism , Homeostasis , Iron/metabolism , Iron, Dietary/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Thermogenesis
3.
J Med Food ; 24(6): 586-594, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33751905

ABSTRACT

Obesity-induced inflammation in adipose tissue (AT) promotes the development of metabolic dysregulations by increasing macrophage recruitment in the stromal vascular fraction (SVF). The activation of nuclear factor-κB (NF-κB) signaling in macrophages serves as a pivotal mediator of AT inflammatory responses by increasing the expression of proinflammatory genes in obesity. Given the purported anti-inflammatory effects of berry consumption in humans, we evaluated if anthocyanin-rich aronia berry extract (ARN) can prevent obesity-induced AT inflammation in vivo. We also examined whether ARN suppresses lipopolysaccharide (LPS)-induced NF-κB activation in RAW 264.7 macrophages and mouse bone marrow-derived macrophages (BMDMs). Male C57BL/6J mice were fed a low-fat diet, a high-fat (HF), and high-sucrose (HS) diet or HF/HS diet supplemented with 0.2% ARN (HF/HS + ARN) for 14 weeks. Compared to HF-/HS-fed mice, ARN supplementation tended to decrease fasting serum glucose (P = .07). Furthermore, ARN supplementation significantly inhibited the phosphorylation of NF-κB p65 in epididymal AT with a concomitant decrease in the expression of Cd11b and Tnfα mRNAs in epididymal SVF isolated, compared with those from HF-/HS-fed mice. Consistent with these in vivo findings, ARN treatment significantly decreased the phosphorylation of p65 in LPS-stimulated RAW 264.7 macrophages and BMDMs. Moreover, ARN suppressed LPS-induced mRNA expression of inflammation mediators (iNos, Cox-2, Tnfα, Mcp-1, and Il-6) and glycolysis markers (Glut1, G6pdh, and Hk1) in both cell types. Taken together, our in vivo and in vitro results suggest that ARN supplementation may attenuate obesity-induced AT inflammation by inhibiting NF-κB signaling and glycolytic pathway in macrophages.


Subject(s)
NF-kappa B , Photinia , Adipose Tissue , Animals , Anthocyanins , Diet, High-Fat/adverse effects , Inflammation/drug therapy , Inflammation/genetics , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Plant Extracts/pharmacology , Sucrose
4.
J Biol Chem ; 296: 100452, 2021.
Article in English | MEDLINE | ID: mdl-33631196

ABSTRACT

The development of thermogenic adipocytes concurs with mitochondrial biogenesis, an iron-dependent pathway. Iron regulatory proteins (IRP) 1 and 2 are RNA-binding proteins that regulate intracellular iron homeostasis. IRPs bind to the iron-response element (IRE) of their target mRNAs, balancing iron uptake and deposition at the posttranscriptional levels. However, IRP/IRE-dependent iron regulation in adipocytes is largely unknown. We hypothesized that iron demands are higher in brown/beige adipocytes than white adipocytes to maintain the thermogenic mitochondrial capacity. To test this hypothesis, we investigated the IRP/IRE regulatory system in different depots of adipose tissue. Our results revealed that 1) IRP/IRE interaction was increased in proportional to the thermogenic function of the adipose depot, 2) adipose iron content was increased in adipose tissue browning upon ß3-adrenoceptor stimulation, while decreased in thermoneutral conditions, and 3) modulation of iron content was linked with mitochondrial biogenesis. Moreover, the iron requirement was higher in HIB1B brown adipocytes than 3T3-L1 white adipocytes during differentiation. The reduction of the labile iron pool (LIP) suppressed the differentiation of brown/beige adipocytes and mitochondrial biogenesis. Using the 59Fe-Tf, we also demonstrated that thermogenic stimuli triggered cell-autonomous iron uptake and mitochondrial compartmentalization as well as enhanced mitochondrial respiration. Collectively, our work demonstrated that IRP/IRE signaling and subsequent adaptation in iron metabolism are a critical determinant for the thermogenic function of adipocytes.


Subject(s)
Aconitate Hydratase/metabolism , Adipocytes/metabolism , Iron/metabolism , Thermogenesis/physiology , 3T3-L1 Cells , Acclimatization , Adipocytes, Beige/metabolism , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Animals , Body Temperature Regulation/physiology , Cell Differentiation , Homeostasis , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 2/genetics , Iron Regulatory Protein 2/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Organelle Biogenesis , RNA, Messenger/metabolism , Signal Transduction
5.
Molecules ; 26(2)2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33440605

ABSTRACT

Excess body weight is a major risk factor for type 2 diabetes (T2D) and associated metabolic complications, and weight loss has been shown to improve glycemic control and decrease morbidity and mortality in T2D patients. Weight-loss strategies using dietary interventions produce a significant decrease in diabetes-related metabolic disturbance. We have previously reported that the supplementation of low molecular chitosan oligosaccharide (GO2KA1) significantly inhibited blood glucose levels in both animals and humans. However, the effect of GO2KA1 on obesity still remains unclear. The aim of the study was to evaluate the anti-obesity effect of GO2KA1 on lipid accumulation and adipogenic gene expression using 3T3-L1 adipocytes in vitro and plasma lipid profiles using a Sprague-Dawley (SD) rat model. Murine 3T3-L1 preadipocytes were stimulated to differentiate under the adipogenic stimulation in the presence and absence of varying concentrations of GO2KA1. Adipocyte differentiation was confirmed by Oil Red O staining of lipids and the expression of adipogenic gene expression. Compared to control group, the cells treated with GO2KA1 significantly decreased in intracellular lipid accumulation with concomitant decreases in the expression of key transcription factors, peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT/enhancer-binding protein alpha (CEBP/α). Consistently, the mRNA expression of downstream adipogenic target genes such as fatty acid binding protein 4 (FABP4), fatty acid synthase (FAS), were significantly lower in the GO2KA1-treated group than in the control group. In vivo, male SD rats were fed a high fat diet (HFD) for 6 weeks to induced obesity, followed by oral administration of GO2KA1 at 0.1 g/kg/body weight or vehicle control in HFD. We assessed body weight, food intake, plasma lipids, levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) for liver function, and serum level of adiponectin, a marker for obesity-mediated metabolic syndrome. Compared to control group GO2KA1 significantly suppressed body weight gain (185.8 ± 8.8 g vs. 211.6 ± 20.1 g, p < 0.05) with no significant difference in food intake. The serum total cholesterol, triglyceride, and low-density lipoprotein (LDL) levels were significantly lower in the GO2KA1-treated group than in the control group, whereas the high-density lipoprotein (HDL) level was higher in the GO2KA1 group. The GO2KA1-treated group also showed a significant reduction in ALT and AST levels compared to the control. Moreover, serum adiponectin levels were significantly 1.5-folder higher than the control group. These in vivo and in vitro findings suggest that dietary supplementation of GO2KA1 may prevent diet-induced weight gain and the anti-obesity effect is mediated in part by inhibiting adipogenesis and increasing adiponectin level.


Subject(s)
Adipogenesis/drug effects , Anti-Obesity Agents/therapeutic use , Chitosan/analogs & derivatives , Obesity/drug therapy , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Anti-Obesity Agents/pharmacology , Chitosan/pharmacology , Chitosan/therapeutic use , Lipid Metabolism/drug effects , Lipids/blood , Male , Mice , Obesity/blood , Obesity/metabolism , Rats, Sprague-Dawley
6.
Antioxidants (Basel) ; 9(2)2020 Jan 28.
Article in English | MEDLINE | ID: mdl-32012803

ABSTRACT

Endocrine disrupting chemicals (EDCs) disrupt the physiological metabolism, thus playing an important role in the development of obesity. EDCs, the so-called 'obesogens', might predispose some individuals to gain weight. This study investigated the effects of bisphenol A (BPA) and its alternatives (BPS and BPF) on adipocyte differentiation and the effects of the leaves of Potentilla rugulosa Nakai extract (LPE) as a functional food ingredient on obesogen-induced lipid production and adipogenesis in 3T3-L1 cells. The results showed that LPE has high total phenolic and flavonoid contents (77.58 ± 0.57 mg gallic acid equivalents (GAE)/g and 57.31 ± 1.72 mg quercetin equivalents (QE)/g, respectively). In addition, LPE exerted significant antioxidant effects in terms of DPPH radical scavenging activity, reducing power, ferric-ion reducing antioxidant power, and oxygen radical absorbance capacity. BPA, BPS, and BPF increased lipid accumulation, protein expressions of adipogenic transcription factors (PPAR-γ, C/EBP-α, and aP2), and reactive oxygen species (ROS) production in 3T3-L1 cells. However, LPE suppressed the BPA-, BPS-, and BPF-induced effects on adipogenesis. Therefore, LPE has potential as a functional food supplement that can prevent bisphenol-induced lipid metabolism disorders.

7.
Int J Mol Med ; 44(2): 559-568, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31198982

ABSTRACT

Processed products from agricultural produce generate a large number of agricultural by­products that contain a number of functional substances. These are often discarded owing to the lack of suitable processing methods. The present study investigated the anti­photoaging properties of fermented rice bran (FRB), soybean cake (FSB) and sesame seed cake (FSC) on ultraviolet B (UVB)­irradiated hairless mouse skin. Results indicated that the oral administration of FRB, FSB and FSC effectively inhibited the UVB irradiation­induced expression of matrix metalloproteinase (MMP)­2, MMP­9, MMP­3 and MMP­13. Reverse transcription­quantitative polymerase chain reaction results also demonstrated that FRB, FSB and FSC significantly inhibited the UVB­induced expression of the genes encoding tumor necrosis factor­α, inducible nitric oxide synthase, interleukin (IL)­6 and IL­1ß when compared with the UVB­vehicle group (P<0.05). Additionally, collagen degradation and mast cell infiltration were reduced in hairless mouse skin. Furthermore, UVB­induced wrinkle formation was also significantly reduced in mouse skin compared with the UVB­vehicle group (P<0.05). These results reveal that fermented agricultural by­products may serve as potential functional materials with anti­photoaging activities.


Subject(s)
Plant Extracts/pharmacology , Skin Aging/drug effects , Skin Aging/radiation effects , Sunscreening Agents/pharmacology , Ultraviolet Rays/adverse effects , Animals , Crops, Agricultural/chemistry , Female , Fermentation , Mice, Hairless , Oryza/chemistry , Plant Extracts/chemistry , Sesamum/chemistry , Skin/drug effects , Skin/radiation effects , Skin/ultrastructure , Glycine max/chemistry , Sunscreening Agents/chemistry
8.
Food Sci Biotechnol ; 28(3): 907-911, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31093449

ABSTRACT

Postprandial blood glucose lowering effect of vitamin B6 (pyridoxine) was evaluated in healthy individuals with normal blood glucose levels. Blood glucose levels were measured every 30 min for 2 h after oral sugar administration with or without 50 mg of pyridoxine. Pyridoxine significantly lowered the postprandial blood glucose levels at 30 min (from 165.95 ± 17.19 to 138.36 ± 20.43, p < 0.01) and 60 min (from 131.40 ± 17.20 to 118.50 ± 15.95) after administration. In addition, the area under the concentration-time curve (AUCt) was reduced by about 8.3% (from 257.08 ± 22.38 to 235.71 ± 12.33, p < 0.05) and the maximum concentration of blood glucose (Cmax) was reduced by about 13.8% (from 165.95 ± 17.19 to 143.07 ± 11.34, p < 0.01) when compared with those of the control group. Our findings suggest that pyridoxine supplementation may be beneficial for controlling postprandial hyperglycemia.

9.
J Med Food ; 22(8): 841-850, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31094612

ABSTRACT

Skin aging is associated with increased reactive oxygen species (ROS) produced by human cells. These radicals are the main causes of skin aging, and skin cells have developed antioxidant enzymes for protection against ROS-induced damage. Antioxidants play critical roles to prevent ROS-induced aging symptoms. In this study, the antioxidant properties of Pourthiaea villosa (Thunb.) Decne. extract (PVDE) were studied. Human dermal fibroblast (HDF) cells were treated with PVDE to evaluate its antioxidant and antiaging activities and to investigate the underlying mechanisms. The identified compounds were polyols, and phenolic and flavonoid compounds from PVDE by UHPLC-LTQ-IT-MS/MS. PVDE exhibited significant antioxidant effects, as evaluated with reducing power, and ABTS and DPPH radical scavenging activity. Furthermore, PVDE treatment significantly increased antioxidant enzyme expressions and effectively blocked H2O2-induced matrix metalloproteinase activity through MAPK signaling pathways in HDFs. Therefore, these results showed that PVDE affords an advantage of being a functional natural material with antioxidant and antiaging effects for the skin.


Subject(s)
Fibroblasts/drug effects , Hydrogen Peroxide/adverse effects , Photinia/chemistry , Plant Extracts/pharmacology , Skin Aging/drug effects , Sunscreening Agents/pharmacology , Antioxidants/pharmacology , Fibroblasts/metabolism , Humans , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Plant Extracts/chemistry , Reactive Oxygen Species/metabolism , Skin Aging/radiation effects , Sunscreening Agents/chemistry , Ultraviolet Rays/adverse effects
10.
Food Sci Biotechnol ; 27(6): 1801-1809, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30483445

ABSTRACT

In the current study investigated the protective effects of citrus based mixture drinks (CBMDs) using oxidative stress in human dermal fibroblast (HDF) cells and restraint-stressed rats. The CBMDs contained citrus bioflavonoids including narirutin and hesperidin. The cell viability of HDF cells treated with H2O2 was observed at 53.9% but treated with CBMD-1 and CBMD-2 (500 µg/mL) on H2O2 exposed HDF cells significantly increased the relative cell viability at 65.0 and 72.2%, respectively. In the treadmill test, the time spent on the electrode plate in the restraint-stressed group was analyzed 24.1 s, but restraint-stressed rats with administered CBMDs (300 mg/kg) had significantly decreased the time at 2.4 (CBMD-1) and 4.7 (CBMD-2) s, respectively. In addition, number of touches the electrode plate in restraint-stressed group was observed at 42.4 ea, but, restraint-stressed rats with administered CBMD-1 and CBMD-2 (300 mg/kg) were significantly decreased at 7.0 and 10.2 ea, respectively.

11.
Nutrients ; 10(2)2018 Feb 14.
Article in English | MEDLINE | ID: mdl-29443928

ABSTRACT

Sesame is an important oilseed crop, which has been used as a traditional health food to ameliorate the prevention of various diseases. We evaluated the changes in the anti-allergic activities of sesame by bioconversion. SDS-PAGE of non-fermented sesame proteins showed major allergen bands, while that of fermented sesame showed only a few protein bands. Additionally, we investigated the effectiveness of fermented sesame by bioconversion in tumor necrosis factor-α (TNF-α)- and interferon-γ (IFN-γ)-induced HaCaT cells. In HaCaT cells, fermented sesame inhibited the mRNA expression of interleukin-6 (IL-6) and interleukin-1ß (IL-1ß), thymus and macrophage-derived chemokine (MDC/CCL22), activation-regulated chemokine (TARC/CCL17), and intercellular adhesion molecule-1 (ICAM-1). Moreover, fermented sesame inhibited the activation of nuclear factor-κB (NF-κB) and signal transducer and activator of transcription 1 (STAT1). Fermented sesame exerts anti-allergic effects by suppressing the expression of chemokines and cytokines via blockade of NF-κB and STAT1 activation.


Subject(s)
Allergens/adverse effects , Cytokines/antagonists & inhibitors , Fermented Foods/analysis , Keratinocytes/metabolism , Plant Proteins, Dietary/adverse effects , Seeds/chemistry , Sesamum/chemistry , Agaricales , Allergens/analysis , Allergens/metabolism , Cell Line , Chemokines/antagonists & inhibitors , Chemokines/genetics , Chemokines/metabolism , Crops, Agricultural/adverse effects , Crops, Agricultural/chemistry , Crops, Agricultural/growth & development , Crops, Agricultural/microbiology , Cytokines/genetics , Cytokines/metabolism , Dermatitis, Atopic/etiology , Dermatitis, Atopic/immunology , Dermatitis, Atopic/prevention & control , Fermentation , Fermented Foods/adverse effects , Fermented Foods/microbiology , Food Handling , Food Hypersensitivity/etiology , Food Hypersensitivity/immunology , Food Hypersensitivity/prevention & control , Fruiting Bodies, Fungal , Gene Expression Regulation, Neoplastic , Humans , Keratinocytes/immunology , Plant Proteins, Dietary/analysis , Plant Proteins, Dietary/metabolism , Republic of Korea , Seeds/adverse effects , Seeds/growth & development , Seeds/microbiology , Sesamum/adverse effects , Sesamum/growth & development , Sesamum/microbiology , Shiitake Mushrooms/isolation & purification , Shiitake Mushrooms/metabolism
12.
Biofactors ; 43(1): 90-99, 2017 Jan 02.
Article in English | MEDLINE | ID: mdl-27388525

ABSTRACT

We have previously reported that administration of low molecular weight chitosan oligosaccharide (GO2KA1) significantly suppressed postprandial blood glucose rise with increased plasma adiponectin and HbA1c levels in animals and humans. However, the cellular mechanisms whereby GO2KA1 exerts antihyperglycemic effects still remain to be determined. Using intestinal Caco-2 cells and 3T3-L1 cells, here we show that GO2KA1 has dual modes of antidiabetic action by (1) inhibiting intestinal α-glucosidase as well as glucose transporters SGLT1 and GLUT2 that were distinct from the acarbose effect; (2) enhancing adipocyte differentiation, PPARγ expression and its target genes, such as FABP4, adiponectin, and GLUT4, whereas the effects were abolished by co-treatment with BADGE, a PPARγ antagonist. Moreover, GO2KA1 significantly increased glucose uptake, which was reduced in the presence of BADGE. Our data show that GO2KA1 may prevent hyperglycemia by inhibiting intestinal glucose digestion and transport and also enhance glucose uptake, at least in part, by upregulating adiponectin expression through PPARγ in adipocytes. These findings may provide potential molecular modes of action for the antidiabetic effects of chitosan oligosaccharide observed in clinical and animal studies. © 2016 BioFactors, 43(1):90-99, 2017.


Subject(s)
Chitosan/analogs & derivatives , Glucose Transporter Type 4/metabolism , Glycoside Hydrolase Inhibitors/pharmacology , PPAR gamma/metabolism , 3T3-L1 Cells , Adiponectin/metabolism , Animals , Caco-2 Cells , Cell Differentiation/drug effects , Chitosan/pharmacology , Drug Evaluation, Preclinical , Fatty Acid-Binding Proteins/metabolism , Gene Expression/drug effects , Glucose/metabolism , Glucose Transporter Type 4/antagonists & inhibitors , Humans , Mice , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , alpha-Glucosidases/metabolism
13.
Food Sci Biotechnol ; 25(3): 911-914, 2016.
Article in English | MEDLINE | ID: mdl-30263353

ABSTRACT

The effect of chitosan oligosaccharide (GO2KA1) administration on postprandial blood glucose levels of subjects with normal blood glucose levels was evaluated following bread consumption. Postprandial blood glucose levels were determined for 2 h after bread ingestion with or without 500 mg of GO2KA1. GO2KA1 significantly lowered the mean, maximum, and minimum levels of postprandial blood glucose at 30 min after the meal. Postprandial blood glucose levels were decreased by about 25% (from 155.11±13.06 to 138.50±13.59, p<0.01) at 30 min when compared to control. Furthermore, we observed that the area under the concentration-time curve (AUCt) was decreased by about 6% (from 255.46±15.43 to 240.15±14.22, p<0.05) and the peak concentration of blood glucose (C max) was decreased by about 11% (from 157.94±10.90 to 140.61±12.52, p<0.01) when compared to control. However, postprandial the time to reach C max (Tmax) levels were the same as those found in control. Our findings suggest that GO2KA1 limits the increase in postprandial blood glucose levels following bread consumption.

14.
Int J Mol Sci ; 16(4): 8811-25, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25906471

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a metabolic disorder characterized by postprandial hyperglycemia, which is an early defect of T2DM and thus a primary target for anti-diabetic drugs. A therapeutic approach is to inhibit intestinal α-glucosidase, the key enzyme for dietary carbohydrate digestion, resulting in delayed rate of glucose absorption. Although tea extracts have been reported to have anti-diabetic effects, the potential bioactivity of tea pomace, the main bio waste of tea beverage processing, is largely unknown. We evaluated the anti-diabetic effects of three selected tea water extracts (TWE) and tea pomace extracts (TPE) by determining the relative potency of extracts on rat intestinal α-glucosidase activity in vitro as well as hypoglycemic effects in vivo. Green, oolong, and black tea bags were extracted in hot water and the remaining tea pomace were dried and further extracted in 70% ethanol. The extracts were determined for intestinal rat α-glucosidases activity, radical scavenging activity, and total phenolic content. The postprandial glucose-lowering effects of TWE and TPE of green and black tea were assessed in male Sprague-Dawley (SD) rats and compared to acarbose, a known pharmacological α-glucosidase inhibitor. The IC50 values of all three tea extracts against mammalian α-glucosidase were lower or similar in TPE groups than those of TWE groups. TWE and TPE of green tea exhibited the highest inhibitory effects against α-glucosidase activity with the IC50 of 2.04 ± 0.31 and 1.95 ± 0.37 mg/mL respectively. Among the specific enzymes tested, the IC50 values for TWE (0.16 ± 0.01 mg/mL) and TPE (0.13 ± 0.01 mg/mL) of green tea against sucrase activity were the lowest compared to those on maltase and glucoamylase activities. In the animal study, the blood glucose level at 30 min after oral intake (0.5 g/kg body wt) of TPE and TWE of both green and black tea was significantly reduced compared to the control in sucrose-loaded SD rats. The TPE of all three teas had significantly higher phenolic content than those of the TWE groups, which correlated strongly with the DPPH radical scavenging activity. This is the first report of tea pomace extract significantly inhibits intestinal α-glucosidase, resulting in delayed glucose absorption and thereby suppressed postprandial hyperglycemia. Our data suggest that tea pomace-derived bioactives may have great potential for further development as nutraceutical products and the reuse of otherwise biowaste as valuable bioresources for the industry.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glycoside Hydrolase Inhibitors/pharmacology , Hyperglycemia/drug therapy , Plant Extracts/pharmacology , alpha-Glucosidases/chemistry , Animals , Blood Glucose , Camellia sinensis/chemistry , Drug Evaluation, Preclinical , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Glycoside Hydrolase Inhibitors/chemistry , Intestines/drug effects , Intestines/enzymology , Male , Plant Extracts/chemistry , Rats, Sprague-Dawley , Tea/chemistry
15.
BMC Complement Altern Med ; 14: 272, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25074485

ABSTRACT

BACKGROUND: Type 2 diabetes is a serious problem for developed countries. Prevention of prediabetes progression to type 2 diabetes with the use of natural products appears to a cost-effective solution. Previously we showed that enzymatically digested low molecular weight chitosan-oligosaccharide with molecular weight (MW) below 1,000 Da (GO2KA1) has potential for hyperglycemia management. METHODS: In this study we evaluated the effect of long-term supplementation of GO2KA1 on hyperglycemia using a db/db mice model. Additionally, we evaluated the effect of GO2KA1 on sucrase and glucoamylase activities and expression, using the same db/db mice model. RESULTS: After 42 days we observed that GO2KA1 supplementation reduced both the blood glucose level and HbA1c in a similar manner with a known anti-diabetic drug, acarbose. When the sucrase and glucoamylase activities of GO2KA1 and control mice were evaluated using enzymatic assay, we observed that GO2KA1 significantly inhibited sucrase in all 3 parts of the intestine, while glucoamylase activity was significantly reduced only in the middle and lower part. When the sucrase-isomaltase (SI) complex expression on mRNA level was evaluated, we observed that GO2KA1 had minimal inhibitory effect on the upper part, more pronounced inhibitory effect on the middle part, while the highest inhibition was observed on the lower part. Our findings suggest that long-term GO2KA1 supplementation in db/db mice results to significant blood glucose and HbA1c reduction, to levels similar with those of acarbose. Furthermore, our findings confirm previous in vitro observations that GO2KA1 has inhibitory effect on carbohydrate hydrolysis enzymes, namely sucrase, maltase and SI complex. CONCLUSIONS: Results from this study provide a strong rationale for the use of GO2KA1 for type 2 diabetes prevention, via inhibition of carbohydrate hydrolysis enzymes. Based on the findings of this animal trial, clinical trials will be designed and pursued.


Subject(s)
Blood Glucose/drug effects , Chitosan/analogs & derivatives , Chitosan/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Hyperglycemia/drug therapy , Oligosaccharides/pharmacology , Prediabetic State/drug therapy , Animals , Body Weight/drug effects , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/prevention & control , Eating/drug effects , Glycated Hemoglobin/metabolism , Glycoside Hydrolases/metabolism , Hyperglycemia/blood , Hyperglycemia/metabolism , Intestines/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prediabetic State/blood , Prediabetic State/metabolism
16.
Int J Mol Sci ; 15(5): 8352-9, 2014 May 12.
Article in English | MEDLINE | ID: mdl-24823880

ABSTRACT

We have previously reported that Amadori compounds exert anti-diabetic effects by lowering sucrose-induced hyperglycemia in normal Sprague-Dawley rats. In the present study we extended our recent findings to evaluate whether α-glucosidase inhibitor arginyl-fructose (AF) lowers blood glucose level in diabetic db/db mice, a genetic model for type 2 diabetes. The db/db mice were randomly assigned to high-carbohydrate diets (66.1% corn starch) with and without AF (4% in the diet) for 6 weeks. Changes in body weight, blood glucose level, and food intake were measured daily for 42 days. Dietary supplementation of AF resulted in a significant decrease of blood glucose level (p < 0.001) and body weight (p < 0.001). The level of HbA1c, a better indicator of plasma glucose concentration over prolonged periods of time, was also significantly decreased for 6-week period (p < 0.001). Dietary treatment of acarbose® (0.04% in diet), a positive control, also significantly alleviated the level of blood glucose, HbA1c, and body weight. These results indicate that AF Maillard reaction product improves postprandial hyperglycemia by suppressing glucose absorption as well as decreasing HbA1c level.


Subject(s)
Diabetes Mellitus, Type 2/diet therapy , Fructose/analogs & derivatives , Fructose/therapeutic use , Glycated Hemoglobin/analysis , Glycoside Hydrolase Inhibitors/therapeutic use , Hyperglycemia/diet therapy , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Dietary Supplements/analysis , Glycoside Hydrolase Inhibitors/chemistry , Hyperglycemia/blood , Hyperglycemia/complications , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL
17.
Int J Mol Sci ; 14(7): 14214-24, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23839092

ABSTRACT

This research investigated the effect of enzymatically digested low molecular weight (MW) chitosan oligosaccharide on type 2 diabetes prevention. Three different chitosan oligosaccharide samples with varying MW were evaluated in vitro for inhibition of rat small intestinal α-glucosidase and porcine pancreatic α-amylase (GO2KA1; <1000 Da, GO2KA2; 1000-10,000 Da, GO2KA3; MW > 10,000 Da). The in vitro results showed that all tested samples had similar rat α-glucosidase inhibitory and porcine α-amylase inhibitory activity. Based on these observations, we decided to further investigate the effect of all three samples at a dose of 0.1 g/kg, on reducing postprandial blood glucose levels in Sprague-Dawley (SD) rat model after sucrose loading test. In the animal trial, all tested samples had postprandial blood glucose reduction effect, when compared to control, however GO2KA1 supplementation had the strongest effect. The glucose peak (Cmax) for GO2KA1 and control was 152 mg/dL and 193 mg/dL, respectively. The area under the blood glucose-time curve (AUC) for GO2KA1 and control was 262 h mg/dL and 305 h mg/dL, respectively. Furthermore, the time of peak plasma concentration of blood glucose (Tmax) for GO2KA1 was significantly delayed (0.9 h) compared to control (0.5 h). These results suggest that GO2KA1 could have a beneficial effect for blood glucose management relevant to diabetes prevention in normal and pre-diabetic individuals. The suggested mechanism of action is via inhibition of the carbohydrate hydrolysis enzyme α-glucosidase and since GO2KA1 (MW < 1000 Da) had higher in vivo effect, we hypothesize that it is more readily absorbed and might exert further biological effect once it is absorbed in the blood stream, relevant to blood glucose management.


Subject(s)
Blood Glucose/metabolism , Glycoside Hydrolase Inhibitors/pharmacology , Hyperglycemia , Oligosaccharides/pharmacology , Animals , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/prevention & control , Disease Models, Animal , Glycoside Hydrolase Inhibitors/chemistry , Hyperglycemia/blood , Hyperglycemia/drug therapy , Intestine, Small/metabolism , Oligosaccharides/chemistry , Rats , Rats, Sprague-Dawley , Swine , alpha-Amylases/antagonists & inhibitors , alpha-Amylases/metabolism , alpha-Glucosidases/metabolism
18.
Eur J Nutr ; 50(2): 81-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20567977

ABSTRACT

BACKGROUND: Naringenin, a flavonoid present in grapefruit, has recently been shown to exert hypolipidemic and hypocholesterolemic effects, which has a particular importance for protecting against chronic diseases. However, the lipid-lowering potential of naringenin at the concentrations in the dietary range and its underlying mechanisms have yet to be fully elucidated. AIM: The aim of the present study was (1) to investigate the effects of dietary naringenin on plasma and hepatic triglyceride and cholesterol levels and on adipose deposition in rat and (2) to determine the contribution of hepatic peroxisome proliferators-activated receptor α (PPARα) expression to fatty acid oxidation. METHODS: Male Long-Evans hooded rats were fed a diet supplemented with naringenin (0.003, 0.006, and 0.012%) for 6 weeks. We analyzed plasma and hepatic lipid contents and determined the protein expression of PPARα, carnitine-palmitoyl transferase 1L (CPT-1), and uncoupling protein 2 (UCP2), all of which are critical genes for fatty acid oxidation. RESULTS: Naringenin supplementation caused a significant reduction in the amount of total triglyceride and cholesterol in plasma and liver. In addition, naringenin supplementation lowered adiposity and triglyceride contents in parametrial adipose tissue. Naringenin-fed animals showed a significant increase in PPARα protein expression in the liver. Furthermore, expression of CPT-1 and UCP2, both of which are known to be regulated by PPARα, was markedly enhanced by naringenin treatment. CONCLUSIONS: Our results indicate that the activation of PPARα transcription factor and upregulation of its fatty acid oxidation target genes by dietary naringenin may contribute to the hypolipidemic and anti-adiposity effects in vivo.


Subject(s)
Adiposity , Flavanones/pharmacology , Hypolipidemic Agents/pharmacology , PPAR alpha/metabolism , Triglycerides/blood , Adipose Tissue/metabolism , Animals , Carnitine O-Palmitoyltransferase/metabolism , Cholesterol/blood , Citrus paradisi/chemistry , Diet , Ion Channels/metabolism , Lipid Metabolism , Liver/metabolism , Male , Mitochondrial Proteins/metabolism , PPAR alpha/genetics , Rats , Rats, Long-Evans , Transcription Factors/metabolism , Uncoupling Protein 2
19.
Phytother Res ; 25(1): 106-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20623718

ABSTRACT

The aim of this study was to investigate whether Rhodiola crenulata extract and tyrosol, a major bioactive phenolic compound present in Rhodiola, change the activities of endogenous antioxidant enzyme response (AER) and energy pathways linked to proline-mediated pentose phosphate pathway (PPP) during adipogenesis. Treatment with Rhodiola extracts inhibited the activities of proline dehydrogenase (PDH) and glucose-6-phosphate dehydrogenase (G6PDH) as well as lipid accumulation and reactive oxygen species (ROS) production. The inhibition of PDH and G6PDH activities by Rhodiola likely prevented proline oxidation required for critical ATP generation that is coupled to AER via the PPP, leading to inhibition of adipogenesis. Rhodiola extracts dose-dependently increased superoxide dismutase (SOD) activity, resulting in a reduced ROS level during adipogenesis. Moreover, the effects of tyrosol, a major bioactive compound in Rhodiola species, were directly correlated with all observed effects by Rhodiola extracts. These results indicate that the antiadipogenic effects of Rhodiola extracts can be attributed to a phenolic tyrosol that may potentially disrupt proline-mediated energy generation and AER via PPP, resulting in the suppression of adipogenesis and lipid accumulation. This further provides a biochemical rationale to identify the roles of phenolics that modulate the cellular redox environment and therefore have relevance for obesity management.


Subject(s)
Adipogenesis/drug effects , Anti-Obesity Agents/pharmacology , Pentose Phosphate Pathway , Rhodiola/chemistry , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/isolation & purification , Antioxidants/metabolism , Catalase/metabolism , Cell Differentiation/drug effects , Glucosephosphate Dehydrogenase/metabolism , Lipid Metabolism , Mice , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Proline Oxidase/metabolism , Reactive Oxygen Species/metabolism , Succinate Dehydrogenase/metabolism , Superoxide Dismutase/metabolism
20.
Biofactors ; 36(6): 459-67, 2010.
Article in English | MEDLINE | ID: mdl-20806284

ABSTRACT

Puerarin, a major isoflavone glycoside from Kudzu root (Pueraria lobata), has been reported to exert antihyperglycemic and antioxidant effects and thus have pharmacological actions in the treatment of diabetes and cardiovascular diseases. We investigated the effects of puerarin on the changes of key gene expression associated with adipocyte differentiation and insulin sensitivity and link to cellular antioxidant response pathways. Puerarin treatment significantly enhanced differentiation of 3T3-L1 preadipocytes accompanying increased lipid accumulation and glucose-6-phosphate dehydrogenase (G6PDH) activity. At a molecular level, puerarin upregulated mRNA expression of peroxisome proliferator-activated receptor γ (PPARγ) and its target genes, an adipocyte-specific fatty acid binding protein (aP2) and GLUT4. Puerarin also caused a significant increase in mRNA level of adiponectin, an important insulin-sensitizing adipocytokine that is downregulated in insulin-resistant and diabetic states. In addition, treatment with puerarin was found to upregulate mRNA levels of G6PDH, glutathione reductase, and catalase, all of which are important for endogenous antioxidant responses. These data suggest that the hypoglycemic effects of puerarin can be attributed to the upregulation of PPARγ and its downstream target genes, GLUT4 and adiponectin expression, leading to increased glucose utilization. Puerarin may also be effective in preventing the rise of oxidative stress during adipocyte differentiation by increasing endogenous antioxidant responses.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Adiponectin , Cell Differentiation/drug effects , Fatty Acid-Binding Proteins/metabolism , Glucose Transporter Type 4/metabolism , Glucosephosphate Dehydrogenase/metabolism , Insulin/metabolism , Insulin/pharmacology , Isoflavones , PPAR gamma/metabolism , Vasodilator Agents , 3T3-L1 Cells , Adiponectin/genetics , Adiponectin/metabolism , Animals , Cell Line , Fatty Acid-Binding Proteins/genetics , Gene Expression/drug effects , Glucose/metabolism , Glucose Transporter Type 4/genetics , Glucosephosphate Dehydrogenase/genetics , Isoflavones/metabolism , Isoflavones/pharmacology , Mice , Oxidation-Reduction/drug effects , PPAR gamma/genetics , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...