Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Exp Mol Med ; 56(4): 809-819, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38658703

ABSTRACT

Chromosomal phase separation is involved in a broad spectrum of chromosome organization and functional processes. Nonetheless, the intricacy of this process has left its molecular mechanism unclear. Here, we introduce the principles governing phase separation and its connections to physiological roles in this context. Our primary focus is contrasting two phase separation mechanisms: self-association-induced phase separation (SIPS) and bridging-induced phase separation (BIPS). We provide a comprehensive discussion of the distinct features characterizing these mechanisms and offer illustrative examples that suggest their broad applicability. With a detailed understanding of these mechanisms, we explore their associations with nucleosomes and chromosomal biological functions. This comprehensive review contributes to the exploration of uncharted territory in the intricate interplay between chromosome architecture and function.


Subject(s)
Chromosomes , Nucleosomes , Humans , Animals , Nucleosomes/metabolism , Chromatin/metabolism , Chromatin/genetics , Chromatin/chemistry
2.
Biomed Pharmacother ; 167: 115441, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37696082

ABSTRACT

Telacebec is a new anti-tuberculosis agent with promising therapeutic activity and a favorable safety profile. This study aimed to characterize the pharmacokinetics of telacebec via interspecies scaling and population pharmacokinetic modeling for the prediction of human pharmacokinetics. Preclinical pharmacokinetic data were obtained from mice, rats, and dogs following intravenous and oral doses of telacebec. A population pharmacokinetic model was developed to describe the pharmacokinetic data from all three species. The disposition parameters were well correlated with the body weight for all species using an allometric equation. Thus, the allometric scaling was incorporated into the population pharmacokinetic model, which could simultaneously describe the plasma concentration vs. time data from all preclinical studies as well as the Phase 1A clinical study. The developed model was used to predict the pharmacokinetics of telacebec after IV injection, including the clearance (CL) of 168.58 [118.86 - 238.73] mL/min and volume of distribution (Vss) of 968.84 [396.87 - 2831.31] L for 80-kg human. The absolute bioavailability of telacebec in humans in the fed state was estimated as 70.34 ± 9.91%. Finally, the population pharmacokinetic model with allometric scaling was utilized to simulate the plasma concentration vs. time profiles of telacebec after multiple oral doses in humans. The model-predicted profiles well agreed with the observed data in Phase 1B clinical trial. The present pharmacokinetic model may help better understand the activity of telacebec, leading to the design of optimal dosing regimens and new formulation development.

3.
Antimicrob Agents Chemother ; 67(1): e0112322, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36507677

ABSTRACT

A phase 1b, randomized, placebo-controlled, double-blind, multiple ascending dose study (NCT02858973) was conducted to assess the safety, tolerability, and pharmacokinetics of the new antituberculosis agent telacebec (Q203). A total of 47 healthy adult subjects entered the study; 36 received telacebec, and 11 received placebo. Telacebec at doses of 20, 50, 100, 160, 250, and 320 mg was orally administered once daily with a standard meal for 14 days. Multiple oral doses of telacebec up to 320 mg daily for 14 days appeared to be safe and well tolerated by healthy adult subjects in this study. There were no deaths, serious adverse events, or subject discontinuations due to adverse events. Following oral doses of telacebec, the overall extent (AUCτ) and peak (Cmax) exposures of telacebec increased from 538.94 to 10,098.47 ng·h/mL and from 76.43 to 1502.33 ng/mL, respectively, with increasing telacebec doses from 20 mg to 320 mg. A steady state was achieved for plasma telacebec by day 12, and there was 1.9- to 3.1-fold accumulation in the extent of telacebec exposure after daily doses for 14 days. Analysis of plasma samples from the participants indicated that telacebec was the primary circulating entity with no significant metabolites. Three potential metabolites of telacebec have been identified, which may be relatively minimal compared to the parent drug. Consistent with findings from preclinical and previous single-dose clinical studies, these results also support the potential of telacebec for further development as a safe and effective agent for the treatment of tuberculosis.


Subject(s)
Tuberculosis , Adult , Humans , Area Under Curve , Tuberculosis/drug therapy , Double-Blind Method , Dose-Response Relationship, Drug , Administration, Oral
4.
Cancers (Basel) ; 14(19)2022 Oct 02.
Article in English | MEDLINE | ID: mdl-36230744

ABSTRACT

Although immune checkpoint blockade (ICB) represents a major breakthrough in cancer immunotherapy, only a limited number of patients with cancer benefit from ICB-based immunotherapy because most immune checkpoint inhibitors (ICIs) target only T cell activation. Therefore, targeting non-T cell components in the tumor microenvironment (TME) can help subvert resistance and increase the applications of ICB-based therapy. Axl and Mer are involved in the carcinogenesis of multiple types of cancer by modulating immune and biological behaviors within tumors. Colony stimulating factor 1 receptor (CSF1R) mediates tumorigenesis in the TME by enhancing tumor associated macrophage (TAM) and myeloid-derived suppressor cell (MDSC) infiltration, facilitating immune escape. Therefore, the simultaneous inhibition of Axl, Mer, and CSF1R kinases may improve therapeutic efficacy by targeting non-T cell components in the TME. Here, we present Q702, a selective, potent small molecule inhibitor targeting Axl, Mer, and CSF1R, for oral administration. Q702 induced antitumor activity in syngeneic tumor mouse models by: remodeling the TME toward immune stimulation; expanding M1 macrophage and CD8 T cell populations and decreasing M2 macrophage and MDSC populations in the TME; and increasing MHC class I and E-cadherin expression in tumor cells. Thus, Q702 may have great potential to broaden the coverage of populations benefiting from ICB-based immunotherapy.

5.
Antimicrob Agents Chemother ; 66(1): e0143621, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34694872

ABSTRACT

Telacebec (Q203) is a potent drug candidate under clinical development for the treatment of drug-naïve and drug-resistant tuberculosis. The first-in-human randomized, placebo-controlled, double-blind, dose-escalation Phase 1A trial (Q203-TB-PI-US001) was conducted to evaluate the safety, tolerability, and pharmacokinetics of telacebec. A total of 56 normal, healthy, male and female subjects (42 active and 14 placebo) were enrolled in the study. The doses of telacebec were 10 mg (Cohort 1), 30 mg (Cohort 2), 50 mg (Cohort 3), 100 mg (Cohort 4), 200 mg (Cohort 5), 400 mg (Cohort 6), and 800 mg (Cohort 7) in a fasted state. Subjects participating in Cohort 4 were also enrolled in Cohort 8 to investigate the food effect on the pharmacokinetics of telacebec after a high-fat meal. In all subjects dosed with telacebec (10 to 800 mg), telacebec was well tolerated and did not lead to any significant or serious adverse events. Following a single oral administration of telacebec (10 to 800 mg), telacebec plasma concentration reached the maximal plasma concentration (Cmax) in average 2.0 to 3.5 h and showed multi-exponential decline thereafter. The area under the plasma concentration versus time curve (AUC) was approximately dose-proportional. A significant increase in plasma concentrations was observed in the fed condition compared with the fasted condition with the geometric mean ratio of 3.93 for Cmax. Moderate delay in Tmax (4.5 h) was also observed in the fed condition. These results, combined with the demonstrated activity against drug-sensitive and multidrug-resistant Mycobacterium tuberculosis, support further investigation of telacebec for the treatment of tuberculosis.


Subject(s)
Piperidines , Pyridines , Administration, Oral , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Humans , Imidazoles , Male
6.
Article in English | MEDLINE | ID: mdl-32205344

ABSTRACT

Telacebec (Q203) is a new antitubercular drug with extremely potent activity against Mycobacterium ulcerans Here, we explored the treatment-shortening potential of Q203 alone or in combination with rifampin (RIF) in a mouse footpad infection model. The first study compared Q203 at 5 and 10 mg/kg doses alone and with rifampin. Q203 alone rendered most mouse footpads culture negative in 2 weeks. Combining Q203 with rifampin resulted in a relapse-free cure 24 weeks after completing 2 weeks of treatment, compared to a 25% relapse rate in mice receiving RIF with clarithromycin, the current standard of care, for 4 weeks. The second study explored the dose-ranging activity of Q203 alone and with RIF, including the extended activity of Q203 after treatment discontinuation. The bactericidal activity of Q203 persisted for ≥ 4 weeks beyond the last dose. All mice receiving just 1 week of Q203 at 2 to 10 mg/kg were culture negative 4 weeks after stopping treatment. Mice receiving 2 weeks of Q203 at 0.5, 2, and 10 mg/kg were culture negative 4 weeks after treatment. RIF did not increase the efficacy of Q203. A pharmacokinetics substudy revealed that Q203 doses of 2 to 10 mg/kg in mice produce plasma concentrations similar to those produced by 100 to 300 mg doses in humans, with no adverse effect of RIF on Q203 concentrations. These results indicate the extraordinary potential of Q203 to reduce the duration of treatment necessary for a cure to ≤ 1 week (or 5 doses of 2 to 10 mg/kg) in our mouse footpad infection model and warrant further evaluation of Q203 in clinical trials.


Subject(s)
Buruli Ulcer , Mycobacterium ulcerans , Animals , Anti-Bacterial Agents/therapeutic use , Buruli Ulcer/drug therapy , Drug Therapy, Combination , Imidazoles , Mice , Mice, Inbred BALB C , Piperidines , Pyridines
8.
J Nanosci Nanotechnol ; 19(10): 6285-6290, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31026949

ABSTRACT

Highly surface-roughened zirconia substrates were obtained from additive zirconia powder coating by room temperature spray processing. Homogeneous and dense zirconia coatings were deposited on sintered zirconia substrates with strong bonding by a powder spray coating method. The thickness and surface roughness of the coating layers on zirconia substrates increased with increasing coating cycles, which was confirmed from atomic force microscopy (AFM) and roughness analyses. The tetragonal phase and chemical composition of the zirconia coating layers were similar to those of the raw 3Y-TZP powder used as a raw material, indicating that no phase or composition changes occurred during the spray process.

9.
J Ethnopharmacol ; 225: 31-41, 2018 Oct 28.
Article in English | MEDLINE | ID: mdl-29958960

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The herbal composition Gyeongshingangjeehwan 18 (GGEx18), composed of Rheum palmatum L. (Polygonaceae), Laminaria japonica Aresch (Laminariaceae), and Ephedra sinica Stapf (Ephedraceae), is used as an antiobesity drug in Korean clinics. The constituents of GGEx18 have traditionally been reported to inhibit obesity and related metabolic diseases such as insulin resistance and dyslipidemia. OBJECTIVE: This study investigated the effects of GGEx18 on nonalcoholic fatty liver disease (NAFLD) in mice fed a high-fat diet (HFD) and the underlying cellular and molecular mechanisms involved. METHODS: C57BL/6 J mice were fed either a low-fat diet (LFD), an HFD, or an HFD supplemented with GGEx18 (125, 250, or 500 mg/kg of body weight/day). After 13 weeks, blood analyses, histology, immunohistochemistry, and real-time PCR were performed to assess NAFLD development in these mice. RESULTS: Mice fed an HFD had increases in body weight, epididymal adipose tissue mass, adipocyte size, and adipose expression of inflammation-related genes compared with those fed an LFD. These increases were ameliorated in mice treated with 500 mg/kg/day GGEx18 without affecting food consumption profiles. GGEx18 not only decreased serum levels of triglycerides, free fatty acids, and alanine aminotransferase, but also decreased hepatic lipid accumulation, numbers of mast cells and α-smooth muscle actin-positive cells, and collagen levels induced by an HFD. Consistent with the histological data, the hepatic expression of lipogenesis-, inflammation-, and fibrosis-related genes was lower, while hepatic fatty acid ß-oxidation-related gene expression was higher, in mice receiving GGEx18 compared to mice fed only the HFD. DISCUSSION AND CONCLUSION: These results indicate that GGEx18 attenuates visceral obesity and NAFLD, in part by altering the expression of genes involved in hepatic steatosis and fibroinflammation in HFD-induced obese mice. These findings suggest that GGEx18 may be effective for preventing and treating NAFLD associated with visceral obesity.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Anti-Obesity Agents/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity, Abdominal/drug therapy , Plant Preparations/therapeutic use , Animals , Diet, High-Fat , Ephedra sinica , Gene Expression Regulation/drug effects , Laminaria , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Obesity, Abdominal/genetics , Obesity, Abdominal/pathology , Phytotherapy , Plant Extracts , Rheum
10.
J Ethnopharmacol ; 206: 315-326, 2017 Jul 12.
Article in English | MEDLINE | ID: mdl-28602867

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Gangjihwan (DF), a polyherbal drug composed of Ephedra intermedia Schrenk et C. A. Mayer (Ephedraceae), Lithospermum erythrorhizon Siebold et Zuccarini (Borraginaceae), and Rheum palmatum L. (Polygonaceae), is used to treat obesity in local Korean clinics. The constituents of DF have traditionally been reported to exert anti-obesity and anti-nonalcoholic fatty liver disease (NAFLD) effects. Thus, we investigated the effects of DF on obesity and NAFLD and the underlying mechanisms. MATERIALS AND METHODS: DF was extracted with water (DF-FW), 30% ethyl alcohol (DF-GA30), or 70% ethyl alcohol (DF-GA70). The chemical profile of DF was monitored using high performance liquid chromatography (HPLC)-ultraviolet analysis. The effects of DF on indices of obesity and NAFLD in high fat diet (HFD)-fed C57BL/6J mice and HepG2 cells were examined using quantitative real-time polymerase chain reaction, Oil red O staining, hematoxylin-eosin staining, toluidine blue staining, and immunohistochemistry. RESULTS: The presence of ephedrine, pseudoephedrine, aloe-emodin, and emodin in DF was determined by 3D chromatography using HPLC. Administration of DF-GA70 to HFD-fed obese mice decreased body weight, epididymal adipose tissue mass, and epididymal adipocyte size. DF-GA70 reduced serum levels of free fatty acids and triglycerides. All three DF extracts lowered serum alanine transaminase levels, hepatic lipid accumulation, and infiltration of macrophages, with the largest effects observed for DF-GA70. DF-GA70 increased mRNA levels of fatty acid oxidation genes and decreased mRNA levels of genes for lipogenesis and inflammation in the liver of obese mice. Treatment of HepG2 cells with a mixture of oleic acid and palmitoleic acid induced significant lipid accumulation, whereas all three DF extracts inhibited lipid accumulation. DF-GA70 also altered the expression of lipolytic and lipogenic genes in HepG2 cells. CONCLUSIONS: These results indicate that DF inhibits obesity and obesity-induced severe hepatic steatosis and inflammation without any adverse effects and that these effects may be mediated by regulation of the hepatic expression of lipid metabolism and inflammatory genes. These findings suggest that DF is a safe and efficient anti-obesity and anti-nonalcoholic steatohepatosis drug.


Subject(s)
Diet, High-Fat , Fatty Liver/drug therapy , Inflammation/drug therapy , Plant Preparations/pharmacology , Animals , Chromatography, High Pressure Liquid , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Plant Preparations/therapeutic use
11.
Food Chem Toxicol ; 106(Pt A): 292-305, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28571771

ABSTRACT

Increasing evidence indicates that angiogenesis inhibitors regulate obesity. This study aimed to determine whether the lemon balm extract ALS-L1023 inhibits diet-induced obesity and nonalcoholic fatty liver disease (NAFLD) in female ovariectomized (OVX) mice. OVX mice received a low fat diet (LFD), a high fat diet (HFD) or HFD supplemented with ALS-L1023 (ALS-L1023) for 15 weeks. HFD mice exhibited increases in visceral adipose tissue (VAT) angiogenesis, body weight, VAT mass and VAT inflammation compared with LFD mice. In contrast, all of these effects were reduced in ALS-L1023 mice compared with HFD mice. Serum lipids and liver injury markers were improved in ALS-L1023 mice. Hepatic lipid accumulation, inflammatory cells and collagen levels were lower in ALS-L1023 mice than in HFD mice. ALS-L1023 mice exhibited a tendency to normalize hepatic expression of genes involved in lipid metabolism, inflammation and fibrosis to levels in LFD mice. ALS-L1023 also induced Akt phosphorylation and increased Nrf2 mRNA expression in livers of obese mice. Our results indicate that the angiogenesis inhibitor ALS-L1023 can regulate obesity, hepatic steatosis and fibro-inflammation, in part through improvement of VAT function, in obese OVX mice. These findings suggest that angiogenesis inhibitors may contribute to alleviation of NAFLD in post-menopausal women with obesity.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Melissa/chemistry , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/drug therapy , Plant Extracts/administration & dosage , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Female , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Ovariectomy , Plant Leaves/chemistry
12.
Int J Mol Sci ; 18(4)2017 Apr 17.
Article in English | MEDLINE | ID: mdl-28420164

ABSTRACT

Similar to neoplastic tissues, growth and development of adipose tissue are thought to be angiogenesis-dependent. Since visceral adipose tissue (VAT) is associated with development and progression of nonalcoholic fatty liver disease (NAFLD), we hypothesized that angiogenesis inhibition would attenuate obesity-induced NAFLD. We fed C57BL/6J mice a low-fat diet (LFD, chow 10% kcal fat), a high-fat diet (HFD, 45% kcal fat) or HFD supplemented with the lemon-balm extract ALS-L1023 (HFD-ALS) for 15 weeks. ALS-L1023 reduced endothelial cell-tube formation in vitro. HFD increased VAT angiogenesis and induced weight gains including body weight, VAT mass and visceral adipocyte size compared with LFD. However, HFD-ALS led to weight reductions without affecting calorie intake compared with HFD. HFD-ALS also reduced serum ALT and AST levels and improved lipid metabolism. HFD-ALS suppressed steatosis, infiltration of inflammatory cells, and accumulation of collagen in livers. HFD-ALS modulated hepatic expression of genes involved in lipid metabolism, inflammation, fibrosis, antioxidation, and apoptosis. Concomitantly, analysis of VAT function revealed that HFD-ALS led to fewer CD68-positive macrophage numbers and lower expression of inflammatory cytokines compared with HFD. Our findings show that the anti-angiogenic herbal extract ALS-L1023 attenuates NAFLD by targeting VAT during obesity, suggesting that angiogenesis inhibitors could aid in the treatment and prevention of obesity-induced human NAFLD.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Diet, High-Fat/adverse effects , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Plant Extracts/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Biomarkers , Disease Models, Animal , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipid Metabolism/drug effects , Lipids/blood , Male , Melissa/chemistry , Mice , Neovascularization, Physiologic/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Organ Size/drug effects , Plant Extracts/chemistry , Plant Leaves/chemistry
13.
J Ethnopharmacol ; 195: 204-213, 2017 Jan 04.
Article in English | MEDLINE | ID: mdl-27845265

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Hepatic steatosis has risen rapidly in parallel with a dramatic increase in obesity. The aim of this study was to determine whether the herbal composition Gambigyeongsinhwan (4) (GGH(4)), composed of Curcuma longa L. (Zingiberaceae), Alnus japonica (Thunb.) Steud. (Betulaceae), and the fermented traditional Korean medicine Massa Medicata Fermentata, regulates hepatic steatosis and inflammation. MATERIALS AND METHODS: The effects of GGH(4) on hepatic steatosis and inflammation in Otsuka Long-Evans Tokushima fatty (OLETF) rats and HepG2 cells were examined using Oil red O, hematoxylin and eosin, and toluidine blue staining, immunohistochemistry, quantitative real-time polymerase chain reaction, and peroxisome proliferator-activated receptor α (PPARα) transactivation assay. RESULTS: Administration of GGH(4) to OLETF rats improved hepatic steatosis and lowered serum levels of alanine transaminase, total cholesterol, triglycerides, and free fatty acids. GGH(4) increased mRNA levels of fatty acid oxidation enzymes (ACOX, HD, CPT-1, and MCAD) and decreased mRNA levels of lipogenesis genes (FAS, ACC1, C/EBPα, and SREBP-1c) in the liver of OLETF rats. In addition, infiltration of inflammatory cells and expression of inflammatory cytokines (CD68, TNFα, and MCP-1) in liver tissue were reduced by GGH(4). Treatment of HepG2 cells with a mixture of oleic acid and palmitoleic acid induced significant lipid accumulation, but GGH(4) inhibited lipid accumulation by regulating the expression of hepatic fatty acid oxidation and lipogenic genes. GGH(4) also increased PPARα reporter gene expression. These effects of GGH(4) were similar to those of the PPARα activator fenofibrate, whereas the PPARα antagonist GW6471 reversed the inhibitory effects of GGH(4) on lipid accumulation in HepG2 cells. CONCLUSIONS: These results suggest that GGH(4) inhibits obesity-induced hepatic steatosis and that this process may be mediated by regulation of the expression of PPARα target genes and lipogenic genes. GGH(4) also suppressed obesity-related hepatic inflammation. Thus, GGH(4) may be a promising drug for the treatment of obesity-related liver diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hepatitis/drug therapy , Hepatocytes/drug effects , Hypolipidemic Agents/pharmacology , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Plant Extracts/pharmacology , Alanine Transaminase/blood , Animals , Biomarkers/blood , Cytokines/metabolism , Disease Models, Animal , Fenofibrate/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hep G2 Cells , Hepatitis/blood , Hepatitis/genetics , Hepatocytes/enzymology , Humans , Inflammation Mediators/metabolism , Lipids/blood , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/enzymology , Male , Mice , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/genetics , Obesity/genetics , Oxazoles/pharmacology , PPAR alpha/genetics , PPAR alpha/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Inbred OLETF , Transfection , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
14.
Opt Express ; 24(19): 21399-406, 2016 Sep 19.
Article in English | MEDLINE | ID: mdl-27661881

ABSTRACT

High-sensitivity distributed measurement of hydrostatic pressure is experimentally demonstrated by optical time-domain analysis of Brillouin dynamic grating (BDG) in polarization maintaining fibers (PMF's). The spectral shift of the BDG in four different types of PMF's are investigated under hydrostatic pressure variation from 14.5 psi (1 bar) to 884.7 psi (61 bar) with less than 2 m spatial resolution. The pressure sensitivity of BDG frequency is measured to be ‒1.69, + 0.65, + 0.78, and + 0.85 MHz/psi for a PM photonic crystal fiber (PM-PCF), two Bow-tie fibers, and a PANDA fiber, respectively, which is about 65 to 169 times larger than that of Brillouin frequency-based pressure sensing.

15.
J Ethnopharmacol ; 178: 238-50, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26702505

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Melissa officinalis L. (Labiatae; lemon balm) has been used traditionally and contemporarily as an anti-stress herb. Current hypotheses suggest that not only chronic stress promotes angiogenesis, but angiogenesis also modulates adipogenesis and obesity. Because the herbal extract ALS-L1023 from M. officinalis L. (Labiatae; lemon balm) has an anti-angiogenic activity, we hypothesized that ALS-L1023 could inhibit adipogenesis and adipocyte hypertrophy. MATERIALS AND METHODS: ALS-L1023 was prepared by a two-step organic solvent fractionation from M. officinalis. The effects of ALS-L1023 on adipogenesis in 3T3-L1 adipocytes and adipocyte hypertrophy in high fat diet (HFD)-fed obese mice were measured using in vivo and in vitro approaches. RESULTS: ALS-L1023 inhibited angiogenesis in a dose-dependent manner in the HUVEC tube formation assay in vitro. Treatment of cells with ALS-L1023 inhibited lipid accumulation and adipocyte-specific gene expression caused by troglitazone or MDI differentiation mix. ALS-L1023 reduced mRNA expression of angiogenic factors (VEGF-A and FGF-2) and MMPs (MMP-2 and MMP-9) in differentiated cells. In contrast, mRNA levels of angiogenic inhibitors (TSP-1, TIMP-1, and TIMP-2) increased. Protease activity, as measured by zymography, showed that activity of MMP-2 and MMP-9 decreased in ALS-L1023-treated cells. ALS-L1023 also inhibited MMP-2 and MMP-9 reporter gene expression in the presence of the MMP inducer phorbol 12-myristate 13-acetate. An in vivo study showed that ALS-L1023 not only decreased adipose tissue mass and adipocyte size, but also reduced mRNA levels of adipose tissue angiogenic factors and MMPs in HFD-fed obese mice. CONCLUSIONS: These results suggest that the anti-angiogenic herbal extract ALS-L1023 suppresses adipogenesis and adipocyte hypertrophy, and this effect may be mediated by inhibiting angiogenesis and MMP activities. Thus, by curbing adipogenesis, anti-angiogenic ALS-L1023 yields a possible therapeutic choice for the prevention and treatment of human obesity and its associated conditions.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Adipose Tissue/drug effects , Diet, High-Fat/adverse effects , Hypertrophy/drug therapy , Melissa/chemistry , Plant Extracts/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Angiogenesis Inducing Agents/metabolism , Animals , Cell Line , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypertrophy/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/chemically induced , Obesity/drug therapy , Obesity/metabolism , Plant Extracts/chemistry , RNA, Messenger/metabolism
16.
PLoS One ; 10(11): e0141612, 2015.
Article in English | MEDLINE | ID: mdl-26599360

ABSTRACT

It has been suggested that angiogenesis modulates adipogenesis and obesity. This study was undertaken to determine whether ALS-L1023 (ALS) prepared by a two-step organic solvent fractionation from Melissa leaves, which exhibits antiangiogenic activity, can regulate adipose tissue growth. The effects of ALS on angiogenesis and extracellular matrix remodeling were measured using in vitro assays. The effects of ALS on adipose tissue growth were investigated in high fat diet-induced obese mice. ALS inhibited VEGF- and bFGF-induced endothelial cell proliferation and suppressed matrix metalloproteinase (MMP) activity in vitro. Compared to obese control mice, administration of ALS to obese mice reduced body weight gain, adipose tissue mass and adipocyte size without affecting appetite. ALS treatment decreased blood vessel density and MMP activity in adipose tissues. ALS reduced the mRNA levels of angiogenic factors (VEGF-A and FGF-2) and MMPs (MMP-2 and MMP-9), whereas ALS increased the mRNA levels of angiogenic inhibitors (TSP-1, TIMP-1, and TIMP-2) in adipose tissues. The protein levels of VEGF, MMP-2 and MMP-9 were also decreased by ALS in adipose tissue. Metabolic changes in plasma lipids, liver triglycerides, and hepatic expression of fatty acid oxidation genes occurred during ALS-induced weight loss. These results suggest that ALS, which has antiangiogenic and MMP inhibitory activities, reduces adipose tissue mass in nutritionally obese mice, demonstrating that adipose tissue growth can be regulated by angiogenesis inhibitors.


Subject(s)
Adipose Tissue/pathology , Angiogenesis Inhibitors/pharmacology , Melissa/chemistry , Plant Extracts/pharmacology , Adipocytes/drug effects , Adipose Tissue/blood supply , Adipose Tissue/drug effects , Adipose Tissue/enzymology , Animals , Body Weight/drug effects , Cell Size/drug effects , Diet, High-Fat , Human Umbilical Vein Endothelial Cells , Lipids/blood , Liver/drug effects , Liver/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Mice, Inbred C57BL , Mice, Obese , Organ Size/drug effects , Oxidation-Reduction/drug effects , PPAR alpha/genetics , PPAR alpha/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/metabolism , Weight Gain/drug effects
17.
J Ethnopharmacol ; 171: 287-94, 2015 Aug 02.
Article in English | MEDLINE | ID: mdl-26068433

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Adipocyte lipid accumulation due to impaired fatty acid oxidation causes adipocyte hypertrophy and adipose tissue increment, leading to obesity. The aim of this study was to determine the antiobesity effects of the herbal composition Gambigyeongsinhwan (4) (GGH(4)) composed of Curcuma longa L. (Zingiberaceae), Alnus japonica (Thunb.) Steud. (Betulaceae), and the fermented traditional Korean medicine Massa Medicata Fermentata. MATERIALS AND METHODS: The effects of GGH(4) and the individual components on lipid accumulation in 3T3-L1 adipocytes and body weight gain in Otsuka Long-Evans Tokushima Fatty (OLETF) rats were examined using Oil red O staining, hematoxylin and eosin staining, quantitative real-time PCR, and peroxisome proliferator-activated receptor α (PPARα) transactivation assay. RESULTS: GGH(4), individual components, and an active principle of Curcuma longa curcumin inhibited lipid accumulation and mRNA levels of adipocyte-specific genes (PPARγ, aP2, and C/EBPα) in 3T3-L1 adipocytes compared with control cells. Treatment with GGH(4), the individual components or curcmumin increased mRNA levels of mitochondrial (CPT-1, MCAD, and VLCAD) and peroxisomal (ACOX and thiolase) PPARα target genes. GGH(4) and the individual components also increased PPARα reporter gene expression compared with control cells. These effects were most prominent in GGH(4)-treated cells. However, the PPARα antagonist GW6471 reversed the inhibitory effects of GGH(4) on adipogenesis. An in vivo study showed that GGH(4) decreased body weight gain, adipose tissue mass, and visceral adipocyte size with increasing mRNA levels of adipose tissue PPARα target genes in OLETF rats. CONCLUSIONS: These results demonstrate that GGH(4) has an antiobesity effects through the inhibition of adipocyte lipid accumulation, and this process may be mediated in part through adipose PPARα activation.


Subject(s)
Anti-Obesity Agents/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Obesity/drug therapy , Plant Extracts/therapeutic use , Plant Preparations/therapeutic use , 3T3-L1 Cells , Alnus , Animals , Anti-Obesity Agents/pharmacology , CCAAT-Enhancer-Binding Proteins/genetics , Curcuma , Drugs, Chinese Herbal/pharmacology , Fatty Acid-Binding Proteins/genetics , Lipid Metabolism/drug effects , Male , Medicine, Korean Traditional , Mice , Obesity/metabolism , Oxazoles/pharmacology , PPAR alpha/antagonists & inhibitors , PPAR gamma/genetics , Phytotherapy , Plant Extracts/pharmacology , Plant Preparations/pharmacology , RNA, Messenger/metabolism , Rats, Inbred OLETF , Triglycerides/metabolism , Tyrosine/analogs & derivatives , Tyrosine/pharmacology , Weight Gain/drug effects
18.
Pharmacogn Mag ; 7(28): 314-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22262934

ABSTRACT

BACKGROUND: Cinnamomi cortex has wide varieties of pharmacological actions such as anti-inflammatory action, anti-platelet aggregation, and improving blood circulation. In this study, we tested to determine whether the Cinnamomi cortex extract has antioxidant activities. MATERIALS AND METHODS: Antioxidative actions were explored by measuring free radical scavenging activity, NO levels, and reducing power. The mechanism of antioxidative action of Cinnamomi cortex was determined by measuring iNOS and COX-II expression in lipopolysaccharide (LPS) stimulated Raw cells. RESULTS: Seventy percent methanolic extract of Cinnamomi cortex exerted significant 1,1-diphenyl--2--picrylhydrazyl (DPPH) free radicals and NO scavenging activities in a dose-dependent manner. More strikingly, the Cinnamomi cortex extract exerted dramatic reducing power activity (13-fold over control). Production of iNOS induced by LPS was significantly inhibited by the Cinnamomi cortex extract, suggesting that it inhibits NO production by suppressing iNOS expression. Additionally, COX-2 induced by LPS was dramatically inhibited by the Cinnamomi cortex extract. CONCLUSION: These results suggest that 70% methanolic extract of Cinnamomi cortex exerts significant antioxidant activity via inhibiting iNOS and COX-II induction.

SELECTION OF CITATIONS
SEARCH DETAIL
...