Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Korean Circ J ; 54(1): 43-56, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37973973

ABSTRACT

BACKGROUND AND OBJECTIVES: The therapeutic strategy for inflammation and degenerative calcification is of utmost importance for bioprosthetic heart valve (BHV) implanted patients. The purpose of this study was to compare the anti-inflammatory and anti-calcification effects of Entelon150® (grape seed extract), losartan, and rosuvastatin, in a rabbit model of intravascular BHV leaflet implantation in bovine pericardium. METHODS: A total of 28 rabbits were implanted with BHV leaflet in the external jugular veins. The Entelon150® group was administered 7.7 mg/kg Entelon150® twice daily for 6 weeks after surgery. The losartan and rosuvastatin groups received 5.14 mg/kg and 1 mg/kg, respectively, once per day. The control group received 1 ml of saline once daily. And then, calcium concentration was measured in the implanted BHV, and histological and molecular analyses were performed on the surrounding tissues. RESULTS: The calcium content of the implanted tissue in the Entelon150® group (0.013±0.004 mg/g) was lower than that in the control group (0.066±0.039 mg/g) (p=0.008). The losartan (0.024±0.016 mg/g, p=0.032) and rosuvastatin (0.022±0.011 mg/g, p=0.032) groups had lower calcium content than the control group, and higher tendency than the Entelon150® group. Immunohistochemistry revealed that the expressions of bone morphogenic protein 2 (BMP2), S-100, and angiotensin II type 1 receptor in the Entelon150® group showed lower tendency than those in the control group. The protein expression levels of BMP2 were reduced in the Entelon150® group compared with those in the control group. CONCLUSIONS: Entelon150® exhibited a significant effect, similar to other drugs, in reducing calcification and inflammation in the intravascular bovine pericardium.

2.
Lab Anim Res ; 39(1): 34, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38102726

ABSTRACT

BACKGROUND: This study aimed to establish an image evaluation grading criteria for experimental stifle joint osteoarthritis (OA) in anterior cruciate ligament transection induced OA beagle dog models. The severity of OA was assessed using X-ray and computed tomography (CT) imaging. RESULTS: A total of 32 dogs (8 controls and 24 OA-induced dogs) were included in the study. The OA-induced group showed significantly higher manual joint palpation, gait analysis, and OA severity scores than the control group. Based on these two results, we calculated correlation coefficients. There was a strong positive correlation between manual joint palpation scores and OA severity on diagnostic imaging and between gait analysis scores and OA severity. CONCLUSIONS: The developed grading criteria based on radiographic evaluation correlated with clinical assessments. The study also employed CT imaging to enhance the accuracy and sensitivity of early-stage OA change detection in the stifle joint. However, further studies with larger sample sizes and multiple evaluators are recommended for the validation and generalizability of this grading system. These established image evaluation grading criteria can help evaluate and monitor the efficacy of interventions and changes in OA lesions in canine models.

3.
Animals (Basel) ; 13(13)2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37444013

ABSTRACT

Atopic dermatitis (AD) is a chronic inflammatory skin disease that is associated with systemic inflammation and immune modulation. Previously, we have shown that extracellular vesicles resulting from human adipose-tissue-derived mesenchymal stem cells (ASC-EVs) attenuated AD-like symptoms by reducing the levels of multiple inflammatory cytokines. Here, we aimed to investigate the improvement of canine AD upon using canine ASC-exosomes in a Biostir-induced AD mouse model. Additionally, we conducted in vivo toxicity studies to determine whether they targeted organs and their potential toxicity. Firstly, we isolated canine ASCs (cASCs) from the adipose tissue of a canine and characterized the cASCs-EVs. Interestingly, we found that cASC-EVs improved AD-like dermatitis and markedly decreased the levels of serum IgE, ear thickness, inflammatory cytokines, and chemokines such as IL-4 and IFN-γ in a dose-dependent manner. Moreover, there was no systemic toxicity in single- or repeat-dose toxicity studies using ICR mice. In addition, we analyzed miRNA arrays from cASC-EVs using next-generation sequencing (NGS) to investigate the role of miRNAs in improving inflammatory responses. Collectively, our results suggest that cASC-EVs effectively attenuate AD by transporting anti-inflammatory miRNAs to atopic lesions alongside no toxicological findings, resulting in a promising cell-free therapeutic option for treating canine AD.

4.
Molecules ; 28(9)2023 May 06.
Article in English | MEDLINE | ID: mdl-37175330

ABSTRACT

Prion protein peptide (PrP) has demonstrated neurotoxicity in brain cells, resulting in the progression of prion diseases with spongiform degenerative, amyloidogenic, and aggregative properties. Thymosin beta 4 (Tß4) plays a role in the nervous system and may be related to motility, axonal enlargement, differentiation, neurite outgrowth, and proliferation. However, no studies about the effects of Tß4 on prion disease have been performed yet. In the present study, we investigated the protective effect of Tß4 against synthetic PrP (106-126) and considered possible mechanisms. Hippocampal neuronal HT22 cells were treated with Tß4 and PrP (106-126) for 24 h. Tß4 significantly reversed cell viability and reactive oxidative species (ROS) affected by PrP (106-126). Apoptotic proteins induced by PrP (106-126) were reduced by Tß4. Interestingly, a balance of neurotrophic factors (nerve growth factor and brain-derived neurotrophic factor) and receptors (nerve growth factor receptor p75, tropomyosin related kinase A and B) were competitively maintained by Tß4 through receptors reacting to PrP (106-126). Our results demonstrate that Tß4 protects neuronal cells against PrP (106-126) neurotoxicity via the interaction of neurotrophic factors/receptors.


Subject(s)
Prion Diseases , Thymosin , Humans , Neurons/metabolism , Signal Transduction , Nerve Growth Factors/metabolism , Transforming Growth Factor beta/metabolism , Thymosin/pharmacology , Hippocampus/metabolism
5.
Adv Healthc Mater ; 12(22): e2202942, 2023 09.
Article in English | MEDLINE | ID: mdl-37256639

ABSTRACT

This study aims to investigate the bone regeneration effect of bovine hydroxyapatite-processed biomaterials Bone-XB and S1-XB in a beagle mandibular defect model. A total of four saddle-type critical sizes (15 mm × 10 mm) bone defects are created in each dog: two defects in the left mandible and two defects in the right mandible. The defect control (DC) group is kept unfilled, and the other three defects are filled with three different biomaterials as follows: positive control Bio-Oss (Bio-Oss group), Bone-XB (XB group), and S1-XB (S1-XB group). Bone regeneration is evaluated by radiography, micro-computed tomography, and histological analysis. It is revealed that Bone-XB and S1-XB significantly increase newly formed bone, defect filling percentage, and bone healing score compared to the DC group, which is confirmed by bone microstructure augmentation (bone volume/total volume, trabecular number, and trabecular thickness). Interestingly, no significant differences are observed between the Bone-XB, S1-XB, and Bio-Oss groups. It is suggested that Bone-XB or S1-XB stimulates bone regeneration demonstrated by the increase in newly formed bone and bone microstructure, thereby improving bone defect filling, which is equivalent to the Bio-Oss. Therefore, bovine hydroxyapatite-processed Bone-XB or S1-XB can be considered effective biomaterials for correcting critical-size bone defects or fractures.


Subject(s)
Bone Substitutes , Animals , Dogs , Cattle , Bone Substitutes/pharmacology , Bone Substitutes/therapeutic use , X-Ray Microtomography , Bone Regeneration , Biocompatible Materials/pharmacology , Minerals/pharmacology , Mandible/diagnostic imaging , Mandible/pathology , Durapatite/pharmacology
6.
Antiviral Res ; 212: 105576, 2023 04.
Article in English | MEDLINE | ID: mdl-36870394

ABSTRACT

Rapid emergence of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has prompted an urgent need for the development of broadly applicable and potently neutralizing antibody platform against the SARS-CoV-2, which can be used for combatting the coronavirus disease 2019 (COVID-19). In this study, based on a noncompeting pair of phage display-derived human monoclonal antibodies (mAbs) specific to the receptor-binding domain (RBD) of SARS-CoV-2 isolated from human synthetic antibody library, we generated K202.B, a novel engineered bispecific antibody with an immunoglobulin G4-single-chain variable fragment design, with sub- or low nanomolar antigen-binding avidity. Compared with the parental mAbs or mAb cocktail, the K202.B antibody showed superior neutralizing potential against a variety of SARS-CoV-2 variants in vitro. Furthermore, structural analysis of bispecific antibody-antigen complexes using cryo-electron microscopy revealed the mode of action of K202.B complexed with a fully open three-RBD-up conformation of SARS-CoV-2 trimeric spike proteins by simultaneously interconnecting two independent epitopes of the SARS-CoV-2 RBD via inter-protomer interactions. Intravenous monotherapy using K202.B exhibited potent neutralizing activity in SARS-CoV-2 wild-type- and B.1.617.2 variant-infected mouse models, without significant toxicity in vivo. The results indicate that this novel approach of development of immunoglobulin G4-based bispecific antibody from an established human recombinant antibody library is likely to be an effective strategy for the rapid development of bispecific antibodies, and timely management against fast-evolving SARS-CoV-2 variants.


Subject(s)
Antibodies, Bispecific , COVID-19 , Animals , Mice , Humans , SARS-CoV-2/metabolism , Antibodies, Viral , Antibodies, Bispecific/pharmacology , Cryoelectron Microscopy , Antibodies, Neutralizing , Spike Glycoprotein, Coronavirus
7.
J Dairy Sci ; 106(3): 1549-1561, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36631322

ABSTRACT

Respiratory virus infections are an escalating issue and have become common worldwide. Influenza and COVID-19 are typical infectious respiratory diseases, and they sometimes lead to various complications. In a situation in which no established drug or treatment exists, consumption of proper food might be beneficial in maintaining health against external infections. We studied the potential effects of mixtures of probiotic strains on various viral infections. The purpose of this study was to assess the ability of yogurt containing probiotics to reduce the risk of respiratory viruses such as influenza H1N1 and SARS-CoV-2 infection. First, we performed in vitro tests using infected Madin-Darby canine kidney (MDCK) and Vero E6 cells, to evaluate the potential effects of yogurt containing high-dose probiotics against influenza H1N1 and SARS-CoV-2 infection. The yogurt significantly reduced plaque formation in the virus-infected cells. We also performed in vivo tests using influenza H1N1-infected C57BL/6 mice and SARS-CoV-2-infected Syrian golden hamsters, to evaluate the potential effects of yogurt. Yogurt was administered orally once daily during the experimental period. Yogurt was also administered orally as pretreatment once daily for 3 wk before viral infection. Regarding influenza H1N1, it was found that yogurt caused an increase in the survival rate, body weight, and IFN-γ, IgG1, and IL-10 levels against viral infection and a decrease in the inflammatory cytokines TNF-α and IL-6. Although the SARS-CoV-2 copy number was not significantly reduced in the lungs of yogurt-treated SARS-CoV-2-infected hamsters, the body weights and histopathological findings of the lungs were improved in the yogurt-treated group. In conclusion, we suggest that consumption of yogurt containing probiotics can lead to beneficial effects to prevent respiratory viral infections.


Subject(s)
COVID-19 , Influenza A Virus, H1N1 Subtype , Influenza, Human , Probiotics , Animals , Dogs , Humans , Mice , COVID-19/veterinary , Mice, Inbred C57BL , SARS-CoV-2 , Yogurt , Disease Models, Animal , Cell Line
8.
BMC Vet Res ; 18(1): 237, 2022 Jun 22.
Article in English | MEDLINE | ID: mdl-35733159

ABSTRACT

BACKGROUND: The aim of this study was to investigate the anti-obesity effects of DWP16001, a sodium-glucose cotransporter-2 (SGLT2 inhibitor), in naturally obese dogs. A total of 20 dogs were divided into four equal groups: one obese control (OC group), and three treated groups; DWP0.2 group, DWP0.5 group, and DWP1 group. OC group fed with food for maintenance and treated groups were fed with food for maintenance with 0.2 mg/kg DWP16001, 0.5 mg/kg DWP16001 and 1 mg/kg DWP16001, respectively. The food for maintenance was provided to dogs as 2 RER (Resting energy requirement) in kcal and DWP16001-supplemented food was administered once a day for 8 weeks. RESULTS: Body condition score, body weight, and fat thickness were significantly reduced (P < 0.05) in the DWP0.2 group compared with the OC group, respectively without affecting the food consumption. At the 10th week the food consumption rate was 101.35 ± 2.56, 166.59 ± 4.72, 98.47 ± 1.44 and 123.15 ± 2.45% compared with initial food consumption rate. Body fat percentage, chest and waist circumference, blood glucose, and insulin were reduced compared to OC group but not significantly different from those of the OC group during experimental period. Serum alanine aminotransferase, alkaline phosphatase, creatine phosphokinase, and creatinine were significantly reduced in DWP0.2 group on 8 weeks. Serum cholesterol and triglycerides were reduced but not significantly. No specific adverse effects were observed throughout the experiment, and hematological parameters were unchanged. The results indicate that DWP16001 was not harmful to the dogs in our study and might have anti-obesity effects in naturally obese dogs. CONCLUSIONS: The above results and discussion suggest that DWP16001 is safe and might have anti-obesity effects in naturally obese dogs.


Subject(s)
Dog Diseases , Obesity , Animals , Blood Glucose , Body Weight , Dog Diseases/drug therapy , Dogs , Insulin , Obesity/drug therapy , Obesity/veterinary , Triglycerides
9.
PLoS One ; 16(3): e0235454, 2021.
Article in English | MEDLINE | ID: mdl-33661896

ABSTRACT

BACKGROUND AND AIMS: Inflammation and calcification are major factors responsible for degeneration of bioprosthetic valve and other substitute heart valve implantations. The objective of this study was to evaluate the anti-inflammatory and anti-calcification effects of Entelon150® (consisting of grape-seed extract) in a beagle dog model of intravascular bovine pericardium implantation. METHODS: In total, 8 healthy male beagle dogs were implanted with a bovine pericardium bilaterally in the external jugular veins and divided into two groups. Animals in the Entelon150® group (n = 4) were treated with 150 mg of Entelon150® twice daily for six weeks after surgery. The negative control (NC) group (n = 4) was treated with 5 ml of saline using the same method. After six weeks, we measured the calcium content, performed histological examination, and performed molecular analysis. RESULTS: The calcium content of implanted tissue in the Entelon150® group (0.56±0.14 mg/g) was significantly lower than that in the NC group (1.48±0.57 mg/g) (p < 0.05). Histopathological examination showed that infiltration of chronic inflammatory cells, such as fibroblasts and macrophages, occurred around the graft in all groups; however, the inflammation level of the implanted tissue in the Entelon150® group was s lower than that in the NC group. Both immunohistochemical and western blot analyses revealed that bone morphogenetic protein 2 expression was significantly attenuated in the Entelon150® group. CONCLUSIONS: Our results indicate that Entelon150® significantly attenuates post-implantation inflammation and degenerative calcification of the bovine pericardium in dogs. Therefore, Entelon150® may increase the longevity of the bovine pericardium after intravascular implantation.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Calcinosis/drug therapy , Grape Seed Extract/therapeutic use , Postoperative Complications/drug therapy , Transcatheter Aortic Valve Replacement/methods , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Bioprosthesis , Calcinosis/etiology , Cattle , Dogs , Fibroblasts/drug effects , Grape Seed Extract/administration & dosage , Grape Seed Extract/pharmacology , Heart Valve Prosthesis , Macrophages/drug effects , Male , Pericardium/transplantation , Transcatheter Aortic Valve Replacement/adverse effects
10.
Biomed Pharmacother ; 125: 110018, 2020 May.
Article in English | MEDLINE | ID: mdl-32092828

ABSTRACT

INTRODUCTION: In recent decades, fine-dust particulate matter (FM) has become a potential health hazard, causing various pathological respiratory disorders around the world. Inflammation induced by FM is regarded as a major cause of respiratory disorder in humans. The purpose of this study was to evaluate the therapeutic efficacy of Shibashin Misena®, a functional food composed of various bioactive ingredients, on FM-induced respiratory disorders in mice. MATERIALS AND METHODS: Briefly, 40 mice were divided equally into four groups: normal controls (NC); FM-induced control group (FC); FM group treated with Shibashin Misena® 0.1 mL/head/day (FM0.1); FM group treated with Shibashin Misena® 0.2 mL/head/day (FM0.2). RESULTS: FM significantly induced TNF-α, IL-17A, IL-1ß, and TGF-ß in bronchoalveolar lavage fluid (BALF) collected from the FM mice. Compared with FC, Shibashin Misena® decreased TNF-α, IL-17A, and IL-1ß levels in BALF, and histopathologic evaluations revealed that Shibashin Misena® treatment significantly reduced inflammatory-cell infiltration and fibrosis related collagen deposition in lung tissue. CONCLUSION: This study demonstrated that Shibashin Misena® decreased FM-induced inflammation and fibrosis in lung tissue. Thus, Shibashin Misena® could be an effective supplement to prevent or improve FM-induced pulmonary disorders.


Subject(s)
Dust , Functional Food , Lung Diseases/diet therapy , Lung Diseases/etiology , Particulate Matter/adverse effects , Animals , Anti-Inflammatory Agents/administration & dosage , Biomarkers , Cytokines/metabolism , Disease Management , Disease Models, Animal , Disease Susceptibility , Immunohistochemistry , Inflammation Mediators/metabolism , Lung Diseases/metabolism , Lung Diseases/pathology , Mice , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology
11.
Eur J Pharmacol ; 869: 172891, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31877278

ABSTRACT

The blood-brain barrier (BBB) is a highly selective permeability barrier that separates the circulating blood from the brain and extracellular fluid in the central nervous system (CNS). The BBB is formed by cerebral endothelial cells connected by tight junctions. Prion diseases are neurodegenerative pathologies characterized by the accumulation of altered forms of the prion protein (PrP), named PrPSc. Thymosin beta 4 (Tß4) is an actin-sequestering peptide known to bind monomeric actin and inhibit its polymerization, and it is known to have a neuroprotective effect. However, the effect of Tß4 on prion disease has not yet been investigated. Therefore, in this study, we investigated the effect of Tß4 on prion-induced BBB dysfunction in hCMEC/D3 human cerebral endothelial cells. We found that Tß4 increased the expression of tight junction protein, but reduced the ratio of F-actin to G-actin. Moreover, we showed that Tß4 significantly improved PrP (106-126)-induced vascular permeability dysfunction in hCMEC/D3 cells. Through human BBB in vitro model, we found that PrP (106-126) could disrupt tight junctions and cytoskeleton arrangement. These results suggest that Tß4 may play a critical role in barrier stabilization. Furthermore, Tß4 may prevent neurodegenerative diseases caused by prion-induced BBB dysfunction.


Subject(s)
Actin Cytoskeleton/metabolism , Endothelial Cells/metabolism , Peptide Fragments , Prions , Thymosin/metabolism , Capillary Permeability , Cell Line , Cell Survival , Humans , Occludin/metabolism , Prion Diseases , Thymosin/genetics , Zonula Occludens-1 Protein/metabolism
12.
Neurotox Res ; 36(1): 58-65, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30552633

ABSTRACT

Prion protein peptide (PrP) has been associated with neurotoxicity in brain cells and progression of prion diseases due to spongiform degeneration and accumulation of the infectious scrapie prion protein (PrPSc). Autophagy has been shown to provide protective functions for neurodegenerative diseases, including prion disease. Thymosin beta 4 (Tß4) plays a key role in the nervous system, providing a neuronal growth effect that includes motility, neurite outgrowth, and proliferation. However, the effect of Tß4 on autophagy in prion disease has not been investigated. In this study, we investigated the neuroprotective effects of Tß4, an activator of autophagy, in cholinergic signaling activation in PrP (106-126)-treated HT22 cells. We found that Tß4-induced autophagy markers, LC3A/B and Beclin1, were protective against PrP-induced neurotoxicity. Interestingly, a balance between autophagy markers and autophagy pathway factors (AKT, p-AKT, mTOR, and p-mTOR) was maintained by Tß4 competitively against each protein factors reacted to PrP (106-126). The cholinergic signaling markers ChTp and AChE, which play an important role in the brain, were maintained by Tß4 competitively against each protein factors reacted to PrP (106-126). However, these results were reversed by 3-MA, an autophagy inhibitor. Taken together, our results indicate that Tß4 has cholinergic signaling activities through the induction of autophagy. Thus, Tß4 may be to a potential therapeutic agent for preventing neurodegenerative diseases.


Subject(s)
Acetylcholine/metabolism , Autophagy , Peptide Fragments/metabolism , Prions/metabolism , Thymosin/metabolism , Animals , Cell Line , Mice , Signal Transduction
13.
Am J Pathol ; 188(11): 2574-2588, 2018 11.
Article in English | MEDLINE | ID: mdl-30125542

ABSTRACT

Toll-like receptor 7 (TLR7) signaling regulates the production of type 1 interferons (IFNs) and proinflammatory cytokines, such as tumor necrosis factor (TNF)-α, implicated in the control of regulatory T (Treg) cell activity. However, the mechanistic interplay between TLR7 signaling and Treg cells in nonalcoholic steatohepatitis (NASH) has not been elucidated. Our aim was to clarify the role of TLR7 signaling in the pathogenesis of NASH. Steatohepatitis was induced in wild-type (WT), TLR7-deficient, IFN-α/ß receptor 1-deficient, and Treg cell-depleted mice. TLR7-deficient and IFN-α/ß receptor 1-deficient mice were more protective to steatohepatitis than WT mice. Of interest, both TNF-α and type 1 IFN promoted apoptosis of Treg cells involved in the prevention of NASH. Indeed, Treg cell-depleted mice had aggravated steatohepatitis compared with WT mice. Finally, treatment with immunoregulatory sequence 661, an antagonist of TLR7, efficiently ameliorated NASH in vivo. These results demonstrate that TLR7 signaling can induce TNF-α production in Kupffer cells and type I IFN production in dendritic cells. These cytokines subsequently induce hepatocyte death and inhibit Treg cells activities, leading to the progression of NASH. Thus, manipulating the TLR7-Treg cell axis might be used as a novel therapeutic strategy to treat NASH.


Subject(s)
Dendritic Cells/immunology , Kupffer Cells/immunology , Membrane Glycoproteins/physiology , Non-alcoholic Fatty Liver Disease/etiology , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 7/physiology , Animals , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/metabolism , Dendritic Cells/pathology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/pathology , Interferon Type I/metabolism , Kupffer Cells/metabolism , Kupffer Cells/pathology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/metabolism
14.
Pharm Biol ; 56(1): 183-191, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29521146

ABSTRACT

CONTEXT: Salicornia europaea (Amaranthaceae) (SE) has been shown to reduce obesity, but it remains a problem as a food supplement because of its high salt content (25-35% NaCl). OBJECTIVES: This study investigated the anti-obesity effects and mechanism of action of desalted SE powder (DSP). MATERIALS AND METHODS: Sprague-Dawley rats (n = 50) were divided into a normal control group (NC), a high-fat diet (HFD)-induced obesity control group (HFD), and HFD groups co-administered DSP (250 and 500 mg/kg) or Garcinia cambogia (Clusiaceae) extract (GE, 200 mg/kg, standard control) orally each day for 12 weeks. RESULTS: The body weight was significantly reduced by co-administration of DSP (596.51 ± 19.84 kg, 4.60% and 562.08 ± 9.74 kg, 10.10%, respectively) and GE (576.00 ± 11.29 kg, 7.88%) relative to the HFD group (625.25 ± 14.02 kg) and was accompanied by reduced abdominal fat mass, and serum lipid levels, with no effects on feed intake. To find the underlying mechanism of the anti-obesity effects, trans-ferulic acid (TFA) was identified as the main ingredient and investigated with regard to whether it attenuated adipogenesity in 3T3L-1 cells. DSP-derived TFA suppressed adipocyte differentiation and accumulation of intracellular lipids. TFA also down-regulated the adipogenesis-related gene expression of sterol regulatory element-binding protein 1, peroxisome proliferator-activated receptor γ, CCAAT/enhancer binding protein-α and fatty acid synthase. CONCLUSIONS: These findings suggest that DSP may be considered for use as a food supplement intent of controlling obesity through its antiobesity and antiadipogenic properties.


Subject(s)
Adipogenesis/drug effects , Anti-Obesity Agents/therapeutic use , Chenopodiaceae , Coumaric Acids/therapeutic use , Obesity/drug therapy , Plant Extracts/therapeutic use , 3T3-L1 Cells , Adipogenesis/physiology , Animals , Anti-Obesity Agents/isolation & purification , Anti-Obesity Agents/pharmacology , Body Weight/drug effects , Body Weight/physiology , Coumaric Acids/isolation & purification , Coumaric Acids/pharmacology , Dose-Response Relationship, Drug , Male , Mice , Obesity/metabolism , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley
15.
Biochem Biophys Res Commun ; 484(4): 890-895, 2017 03 18.
Article in English | MEDLINE | ID: mdl-28188785

ABSTRACT

Sortilin-related receptor 1 (SORL1) is a neuronal sorting protein that reduces amyloid precursor protein (APP) trafficking to secretases that generate amyloid beta (Aß). Although 6-shogaol, a constituent of ginger, has been reported to have anti-inflammatory and anti-oxidant effects on neuronal cells, research regarding the activation of SORL1 has not yet been reported. Here, we aimed to investigate whether 6-shogaol contributes to the increases in SORL1 that are related to Alzheimer's disease (AD). To clarify the effect of 6-shogaol as a possible activator of SORL1, we used SORL1 siRNA as a blockade of SORL1 in hippocampal neuronal cells (HT22). We found that SORL1 siRNA treatment naturally inhibited SORL1 and led to increases in ß-secretase APP cleaving enzyme (BACE), secreted APP-ß (sAPPß) and Aß. In contrast, 6-shogaol-mediated activation of SORL1 significantly downregulated BACE, sAPPß, and Aß in both in vitro HT22 cells and in vivo APPSw/PS1-dE9 Tg mice. Therefore, SORL1 activation by 6-shogaol provides neuronal cell survival through the inhibition of Aß production. These results indicate that 6-shogaol should be regarded as an SORL1 activator and a potential preventive agent for the treatment of AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Amyloidogenic Proteins/metabolism , Catechols/administration & dosage , Membrane Transport Proteins/metabolism , Neurons/metabolism , Receptors, LDL/metabolism , Signal Transduction/physiology , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Mice , Mutagens/administration & dosage , Neurons/drug effects , Signal Transduction/drug effects
16.
J Tissue Eng Regen Med ; 11(4): 1262-1273, 2017 04.
Article in English | MEDLINE | ID: mdl-25921810

ABSTRACT

Rapid vascular remodelling of damaged dermal tissue is required to heal burn wounds. Thymosin ß4 (Tß4) is a growth factor that has been shown to promote angiogenesis and dermal wound repair. However, the underlying mechanisms based on Tß4 function have not yet been fully investigated. In the present study, we investigated how Tß4 improves dermal burn wound healing via actin cytoskeletal remodelling and the action of heat-shock proteins (HSPs), which are a vital set of chaperone proteins that respond to heat shock. Our in vitro results achieved with the use of human umbilical vein endothelial cells (HUVECs) revealed a possible signal between Tß4 and HSP70. Moreover, we confirmed that remodelling of filamentous actin (F-actin) was regulated by Tß4-induced HSP70 in HUVECs. Based on these in vitro results, we confirmed the healing effects of Tß4 in an adapted dermal burn wound in vivo model. Tß4 improved wound-healing markers, such as wound closure and vascularization. Moreover, Tß4 maintained the long-term expression of HSP70, which is associated with F-actin regulation during the wound-healing period. These results suggest that an association between Tß4 and HSP70 is responsible for the healing of burn wounds, and that this association may regulate F-actin remodelling. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Actin Cytoskeleton/metabolism , Burns/metabolism , Burns/pathology , HSP70 Heat-Shock Proteins/metabolism , Thymosin/pharmacology , Wound Healing/drug effects , Animals , Biomarkers/metabolism , Cell Survival/drug effects , Dermis/pathology , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Inbred C57BL , Models, Biological , Neovascularization, Physiologic/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism
17.
Exp Toxicol Pathol ; 68(10): 553-558, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27575989

ABSTRACT

Phosphatidylcholine (PC) from egg yolk is a bioactive substance with various beneficial effects, including anti-inflammatory and anti-oxidant effects. Recently, this substance has been reported to prevent acute hepatotoxicity. In the present study, we aimed to evaluate the putative protective effect of PC on carbon tetrachloride (CCl4)-induced nephrotoxicity in ICR mice. Many previous studies demonstrated that CCl4 induces nephrotoxicity resulting in renal oxidative damage. CCl4 in corn oil (0.1ml, 1.2g/kg) was intra-peritoneally injected into 7-week-old ICR mice twice a week. PC in corn oil (0.1ml, 100mg/kg) was then orally injected daily for a week. In 7 days, blood urea nitrogen (BUN) and creatinine concentrations had significantly increased in the CCl4 group compared to the control group, whereas the PC and CCl4 co-injected group had significantly decreased BUN and creatinine concentrations compared to the CCl4 group. Comparative analysis of histopathological injuries revealed that PC abrogated the nephrotoxicity of CCl4 at 7 days. Accordingly, PC also improved renal fibrosis induced by CCl4. Various biomarkers associated with oxidative damage appeared to be up-regulated in the CCl4 group, whereas in the PC and CCl4 co-injected group, levels of oxidative damage significantly decreased. Aquaporin1 (AQP1), an important water transport protein in the kidney, was down regulated in the CCl4 group compared to the control group. PC counteracted this effect. These results strongly suggest that PC can protect against oxidative damage induced by CCl4 in the kidney and enhance recovery from renal disorders.


Subject(s)
Antioxidants/pharmacology , Carbon Tetrachloride Poisoning/prevention & control , Kidney/drug effects , Phosphatidylcholines/pharmacology , Animals , Disease Models, Animal , Immunoblotting , Kidney/pathology , Male , Mice , Mice, Inbred ICR , Oxidative Stress/drug effects , Real-Time Polymerase Chain Reaction
18.
Biochem Biophys Res Commun ; 477(1): 96-102, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27286707

ABSTRACT

Although 6-shogaol, a constituent of ginger, has been reported to have anti-inflammatory and anti-oxidant effects on neuronal cells, the effects of 6-shogaol on Alzheimer's disease (AD) have not yet been investigated. Here we aimed to determine whether 6-shogaol exerts neuroprotective effects against AD. Specifically, we investigated the effects of 6-shogaol on the cysteinyl leukotriene 1 receptor (CysLT1R), a major factor in AD pathogenesis. Moreover, we clarified the relationship between CysLT1R and cathepsin B, a cysteine protease. We used in vitro and in vivo models to determine whether 6-shogaol inhibits CysLT1R/cathepsin B in an amyloid-beta (Aß; 1-42)-induced model of neurotoxicity. We first confirmed that CysLT1R and cathepsin B are upregulated by Aß (1-42) and that CysLT1R activation induces cathepsin B. In contrast, we found that 6-shogaol-mediated inhibition of CysLT1R downregulates cathepsin B in both in vitro and in vivo models. Furthermore, we found that 6-shogaol-mediated inhibition of CysLT1R/cathepsin B reduces Aß deposition in the brain and ameliorates behavioral deficits in APPSw/PS1-dE9 Tg mice. Our results indicate that 6-shogaol is a CysLT1R/cathepsin B inhibitor and is a novel potential therapeutic agent for the treatment of various neurodegenerative diseases, including AD.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/antagonists & inhibitors , Catechols/pharmacology , Cathepsin B/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Receptors, Leukotriene/physiology , Animals , Avoidance Learning/drug effects , Brain/drug effects , Brain/enzymology , Brain/metabolism , Cell Line , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic
19.
Sci Rep ; 6: 27849, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27279075

ABSTRACT

Recently, a possible link between toll-like receptor 7 (TLR7) and liver disease was suggested, although it was limited to fibrosis. Based on this report, we investigated whether TLR7 has a pivotal role in non-alcoholic fatty liver disease (NAFLD). The TLR7 signaling pathway, which is activated by imiquimod (TLR7 ligand) naturally, induced autophagy and released insulin-like growth factor 1 (IGF-1) into medium from hepatocytes. Lipid accumulation induced by unsaturated fatty acid (UFA; arachidonic acid:oleic acid = 1:1) in hepatocytes, was attenuated in TLR7 and autophagy activation. Interestingly, TLR7 activation attenuated UFA-induced lipid peroxidation products, such as malondialdehyde (MDA) and 4-Hydroxy-2-Nonenal (4-HNE). To clarify a possible pathway between TLR7 and lipid peroxidation, we treated hepatocytes with MDA and 4-HNE. MDA and 4-HNE induced 2-folds lipid accumulation in UFA-treated hepatocytes via blockade of the TLR7 signaling pathway's IGF-1 release compared to only UFA-treated hepatocytes. In vivo experiments carried out with TLR7 knockout mice produced results consistent with in vitro experiments. In conclusion, TLR7 prevents progression of NAFLD via induced autophagy and released IGF-1 from liver. These findings suggest a new therapeutic strategy for the treatment of NAFLD.


Subject(s)
Fatty Acids, Unsaturated/metabolism , Hepatocytes/metabolism , Insulin-Like Growth Factor I/metabolism , Membrane Glycoproteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Toll-Like Receptor 7/metabolism , Aminoquinolines/pharmacology , Animals , Hepatocytes/pathology , Imiquimod , Insulin-Like Growth Factor I/genetics , Lipid Peroxidation/drug effects , Lipid Peroxidation/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Toll-Like Receptor 7/genetics
20.
Eur J Pharmacol ; 788: 241-247, 2016 Oct 05.
Article in English | MEDLINE | ID: mdl-27346834

ABSTRACT

6-Shogaol can be extracted from ginger and has been shown to exert anti-inflammatory and antioxidant activities, which are potentially relevant to the treatment of central nervous system disorders. Oxidative stress and inflammation are closely associated with ischemic injury and can eventually result in neuronal death. The aim of this study was to evaluate if 6-shogaol exerts neuroprotective activity. To this end, we determined its effects on oxidative stress and inflammation in a mouse model of middle cerebral artery occlusion (MCAO)-induced brain damage. In this model, MCAO was induced in C57BL/6 mice (30-35g, 9 weeks) for 1h, followed by 24h reperfusion. Mice were treated orally with 6-shogaol (0.1ml, 5 or 20mg/kg) once daily for 7 consecutive days prior to MCAO. We found that 6-shogaol significantly reduced neurological deficit scores and the mean infarct area. Moreover, 6-shogaol improved the behavioral deficits in the MCAO group. In addition, 6-shogaol pretreatment dampened MCAO-mediated production of reactive oxygen species and inflammatory cytokines. Mechanistic studies revealed that 6-shogaol inhibits the cysteinyl leukotriene 1 receptor (CysLT1R) and mitogen-activated protein kinase (MAPK) signaling proteins, thus providing a potential pharmacological mechanism for our observations. These results suggest that 6-shogaol can ameliorate the outcomes of MCAO and could thus be used as a potential preventive of stroke.


Subject(s)
Catechols/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Oxidative Stress/drug effects , Animals , Behavior, Animal/drug effects , Catechols/therapeutic use , Cytokines/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Inflammation/complications , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Receptors, Leukotriene/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...