Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Phytomedicine ; 128: 155403, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38564920

ABSTRACT

BACKGROUND: Cardiovascular disease is one of the main causes of global mortality, and there is an urgent need for effective treatment strategies. Gut microbiota-dependent metabolite trimethylamine-N-oxide (TMAO) promotes the development of cardiovascular diseases, and shizukaol C, a natural sesquiterpene isolated from Chloranthus multistachys with various biological activities, might exhibit beneficial role in preventing TMAO-induced vascular inflammation. PURPOSE: The purpose of this study was to investigate the anti-inflammatory effects and the underlying mechanisms of shizukaol C on TMAO-induced vascular inflammation. METHODS: The effect and underlying mechanism of shizukaol C on TMAO-induced adhesion molecules expression, bone marrow-derived macrophages (BMDM) adhesion to VSMC were evaluated by western blot, cell adhesion assay, co-immunoprecipitation, immunofluorescence assay, and quantitative Real-Time PCR, respectively. To verify the role of shizukaol C in vivo, TMAO-induced vascular inflammation model were established using guidewire-induced injury on mice carotid artery. Changes in the intima area and the expression of GSTpi, VCAM-1, CD68 were examined using haematoxylin-eosin staining, and immunofluorescence assay. RESULTS: Our data demonstrated that shizukaol C significantly suppressed TMAO-induced adhesion molecule expression and the bone marrow-derived macrophages (BMDM) adhesion in vascular smooth muscle cells (VSMC). Mechanically, shizukaol C inhibited TMAO-induced c-Jun N-terminal kinase (JNK)-nuclear factor-kappa B (NF-κB)/p65 activation, and the JNK inhibition was dependent on the shizukaol C-mediated glutathione-S-transferase pi (GSTpi) expression. By further molecular docking and protein-binding analysis, we demonstrated that shizukaol C directly binds to Keap1 to induce Nrf2 nuclear translocation and upregulated GSTpi expression. Consistently, our in vivo experiment showed that shizukaol C elevated the expression level of GSTpi in carotid arteries and alleviates TMAO-induced vascular inflammation. CONCLUSION: Shizukaol C exerts anti-inflammatory effects in TMAO-treated VSMC by targeting Keap1 and activating Nrf2-GSTpi signaling and resultantly inhibits the downstream JNK-NF-κB/p65 activation and VSMC adhesion, and alleviates TMAO-induced vascular inflammation in vivo, suggesting that shizukaol C may be a potential drug for treating TMAO-induced vascular diseases.


Subject(s)
Inflammation , Muscle, Smooth, Vascular , Sesquiterpenes , Animals , Male , Mice , Anti-Inflammatory Agents/pharmacology , Cell Adhesion/drug effects , Inflammation/chemically induced , Inflammation/drug therapy , Kelch-Like ECH-Associated Protein 1/drug effects , Kelch-Like ECH-Associated Protein 1/metabolism , Macrophages/drug effects , Macrophages/metabolism , Methylamines/pharmacology , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/metabolism , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , Glutathione S-Transferase pi/drug effects , Glutathione S-Transferase pi/metabolism
2.
Alcohol Clin Exp Res ; 45(6): 1188-1199, 2021 06.
Article in English | MEDLINE | ID: mdl-33885179

ABSTRACT

BACKGROUND: Recent studies have shown that human and experimental alcohol-related liver disease (ALD) is robustly associated with dysregulation of bile acid homeostasis, which may in turn modulate disease severity. Pharmacological agents targeting bile acid metabolism and signaling may be potential therapeutics for ALD. METHODS: The potential beneficial effects of a gut-restricted apical sodium-dependent bile acid transporter (ASBT) inhibitor were studied in a chronic-plus-binge ALD mouse model. RESULTS: Blocking intestinal bile acid reabsorption by the gut-restricted ASBT inhibitor GSK2330672 attenuated hepatic steatosis and liver injury in a chronic-plus-binge ALD mouse model. Alcohol feeding is associated with intestinal bile acid accumulation but paradoxically impaired ileal farnesoid × receptor (FXR) function, and repressed hepatic cholesterol 7α-hydrolase (CYP7A1) expression despite decreased hepatic small heterodimer partner (SHP) and ileal fibroblast growth factor 15 (FGF15) expression. ASBT inhibitor treatment decreased intestinal bile acid accumulation and increased hepatic CYP7A1 expression, but further decreased ileal FXR activity. Alcohol feeding induces serum bile acid concentration that strongly correlates with a liver injury marker. However, alcohol-induced serum bile acid elevation is not due to intrahepatic bile acid accumulation but is strongly and positively associated with hepatic multidrug resistance-associated protein 3 (MRP4) and MRP4 induction but poorly associated with sodium-taurocholate cotransporting peptide (NTCP) expression. ASBT inhibitor treatment decreases serum bile acid concentration without affecting hepatocyte basolateral bile acid uptake and efflux transporters. CONCLUSION: ASBT inhibitor treatment corrects alcohol-induced bile acid dysregulation and attenuates liver injury in experimental ALD.


Subject(s)
Lipid Metabolism/drug effects , Liver Diseases, Alcoholic/drug therapy , Liver/drug effects , Methylamines/therapeutic use , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Symporters/antagonists & inhibitors , Thiazepines/therapeutic use , Angiogenic Proteins/metabolism , Animals , Bile Acids and Salts/blood , Drug Evaluation, Preclinical , Liver/metabolism , Male , Methylamines/pharmacology , Mice, Inbred C57BL , Multidrug Resistance-Associated Proteins/metabolism , Thiazepines/pharmacology , Transaminases/blood
3.
Plant Signal Behav ; 16(4): 1879542, 2021 04 03.
Article in English | MEDLINE | ID: mdl-33586610

ABSTRACT

N,N-dimethyl-hexadecylamine (DMHDA) is a volatile organic compound (VOC) produced by some plant growth-promoting rhizobacteria (PGPR), which inhibits the growth of pathogenic fungi and induces iron uptake by roots. In this report, through the application of a wide range of concentrations, we found that DMHDA affects Arabidopsis primary root growth and lateral root formation in a dose-dependent manner where 1 and 2 µM promoted root growth and higher (4-32 µM) concentrations repressed growth. Cytokinin-inducible TCS::GFP and ARR5::uidA gene constructs showed an increased expression in columella cells and root meristem, respectively, at 2 µM DMHDA, but their expression domains strongly diminished at growth repressing treatments. To test if either primary root growth promotion or repression could involve members of the cytokinin receptor family, the growth of WT and double mutant combinations cre1-12 ahk2-2, cre1-12 ahk3-3, and ahk2-2 ahk3-3 was tested in control conditions or supplemented with 2 µM or 16 µM DMHDA. Noteworthy, the root growth promotion disappeared in cre1-12 ahk2-2 and ahk2-2 ahk3-3 combinations, whereas all double mutants had higher repression than the WT at high doses. We further show that DMHDA fails to mimic the effects of ethylene in Arabidopsis seedlings grown in darkness that include an exaggerated apical hook, stem and root shortening, and root hair elongation. Our data help unravel how Arabidopsis senses a growth-modulating bacterial volatile through changes in cytokinin responsiveness.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Cytokinins/metabolism , Histidine Kinase/metabolism , Methylamines/pharmacology , Plant Roots/growth & development , Signal Transduction , Volatile Organic Compounds/pharmacology , Arabidopsis/drug effects , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis Proteins/genetics , Gene Expression Regulation, Plant/drug effects , Genes, Reporter , Histidine Kinase/genetics , Mutation/genetics , Plant Roots/drug effects , Plants, Genetically Modified , Signal Transduction/drug effects
4.
Sci Rep ; 10(1): 11047, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32632088

ABSTRACT

Hypothalamic adult neurogenesis provides the basis for renewal of neurons involved in the regulation of whole-body energy status. In addition to hormones, cytokines and growth factors, components of the diet, particularly fatty acids, have been shown to stimulate hypothalamic neurogenesis; however, the mechanisms behind this action are unknown. Here, we hypothesized that GPR40 (FFAR1), the receptor for medium and long chain unsaturated fatty acids, could mediate at least part of the neurogenic activity in the hypothalamus. We show that a GPR40 ligand increased hypothalamic cell proliferation and survival in adult mice. In postnatal generated neurospheres, acting in synergy with brain-derived neurotrophic factor (BDNF) and interleukin 6, GPR40 activation increased the expression of doublecortin during the early differentiation phase and of the mature neuronal marker, microtubule-associated protein 2 (MAP2), during the late differentiation phase. In Neuro-2a proliferative cell-line GPR40 activation increased BDNF expression and p38 activation. The chemical inhibition of p38 abolished GPR40 effect in inducing neurogenesis markers in neurospheres, whereas BDNF immunoneutralization inhibited GPR40-induced cell proliferation in the hypothalamus of adult mice. Thus, GPR40 acts through p38 and BDNF to induce hypothalamic neurogenesis. This study provides mechanistic advance in the understating of how a fatty acid receptor regulates adult hypothalamic neurogenesis.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Neurogenesis/physiology , Receptors, G-Protein-Coupled/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Hypothalamus/drug effects , Imidazoles/pharmacology , Interleukin-6/physiology , Ligands , Male , Methylamines/pharmacology , Mice , Mice, Inbred C57BL , Models, Neurological , Neurons/drug effects , Neurons/physiology , Propionates/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Receptors, G-Protein-Coupled/agonists , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
5.
Elife ; 92020 06 08.
Article in English | MEDLINE | ID: mdl-32510330

ABSTRACT

Trimethylamine-oxide (TMAO) is present in seafood which is considered to be beneficial for health. Deep-water animals accumulate TMAO to protect proteins, such as lactate dehydrogenase (LDH), against hydrostatic pressure stress (HPS). We hypothesized that TMAO exerts beneficial effects on the circulatory system and protects cardiac LDH exposed to HPS produced by the contracting heart. Male, Sprague-Dawley and Spontaneously-Hypertensive-Heart-Failure (SHHF) rats were treated orally with either water (control) or TMAO. In vitro, LDH with or without TMAO was exposed to HPS and was evaluated using fluorescence correlation spectroscopy. TMAO-treated rats showed higher diuresis and natriuresis, lower arterial pressure and plasma NT-proBNP. Survival in SHHF-control was 66% vs 100% in SHHF-TMAO. In vitro, exposure of LDH to HPS with or without TMAO did not affect protein structure. In conclusion, TMAO reduced mortality in SHHF, which was associated with diuretic, natriuretic and hypotensive effects. HPS and TMAO did not affect LDH protein structure.


Heart failure is a common cause of death in industrialized countries with aging populations. Japan, however, has lower rates of heart failure and fewer deaths linked to this disease than the United States or Europe, despite having the highest proportion of elderly people in the world. Dietary differences between these regions may explain the lower rate of heart failure in Japan. The Japanese diet is rich in seafood, which contains nutrients that promote heart health, such as omega-3 fatty acids. Seafood also contains other compounds, including trimethylamine oxide (TMAO). Fish that live in deep waters undergo high pressures, which can damage their proteins, but TMAO seems to protect the proteins from harm. In humans, eating seafood increases TMAO levels in the blood and urine, but it is unclear what effects this has on heart health. Increased levels of TMAO in the blood are associated with cardiovascular diseases, but scientists are not sure whether TMAO itself harms the heart. A toxic byproduct of gut bacteria called TMA is converted in TMAO in the body, so it is possible that TMA rather than TMAO is to blame. To assess the effects of dietary TMAO on heart failure, Gawrys-Kopczynska et al. fed the compound to healthy rats and rats with heart failure for one year. TMAO had no effects on the healthy rats. Of the rats with heart failure that were fed TMAO, all of them survived the year, while one third of rats with heart failure that were not fed TMAO died. TMAO-treated rats with heart failure had lower blood pressure and urinated more than untreated rats with the condition. The experiments suggest that dietary TMAO may mimic the effects of heart failure treatments, which remove excess water and salt and lower pressure on the heart. More studies are needed to confirm whether TMAO has this same effect on humans.


Subject(s)
Diuresis/drug effects , Heart Failure/drug therapy , Methylamines/chemistry , Methylamines/pharmacology , Seafood/analysis , Angiotensins/genetics , Angiotensins/metabolism , Animals , Gene Expression Regulation/drug effects , Kidney/drug effects , Male , Methylamines/administration & dosage , Microfluidic Analytical Techniques , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/genetics , Receptor, Angiotensin, Type 2/metabolism , Temperature
6.
Int J Mol Sci ; 21(3)2020 Feb 05.
Article in English | MEDLINE | ID: mdl-32033285

ABSTRACT

l-carnitine supplementation has been used for cardiovascular health protection for a long time. Recently, trimethylamine-N-oxide (TMAO), which is an end product of l-carnitine metabolism via the activity of microbiota, has been identified as a cardiovascular disease (CVD) biomarker. The aim of this study was to assess the effect of 6 months of l-carnitine supplementation in a group of aged women engaged in a regular physical training. Platelet mitochondrial DNA methylation, an emerging and innovative biomarker, lipid profile and TMAO levels have been measured. TMAO increased after l-carnitine supplementation (before 344.3 ± 129.8 ng/mL vs. after 2216.8 ± 1869.0 ng/mL; n = 9; paired t-test, p = 0.02). No significant effects on TMAO were exerted by training alone (n = 9) or by l-leucine supplementation (n = 12). TMAO levels after 6 months of l-carnitine supplementation were associated with higher low-density lipoprotein-cholesterol (LDL-c) (Spearman Rho = 0.518, p = 0.003) and total cholesterol (TC) (Spearman Rho = 0.407, p = 0.026) levels. l-carnitine supplementation increased D-loop methylation in platelets (+6.63%; paired t-test, p = 0.005). D-loop methylation was not directly correlated to the TMAO augmentation observed in the supplemented group, but its increase inversely correlated with TC (Pearson coefficient = -0.529, p = 0.029) and LDL-c (Pearson coefficient = -0.439, p = 0.048). This evidence supports the hypothesis that the correlation between l-carnitine, TMAO and atherosclerosis might be more complex than already postulated, and the alteration of mitochondrial DNA (mtDNA) methylation in platelets could be involved in the pathogenesis of this multifactorial disease.


Subject(s)
Atherosclerosis/metabolism , Biomarkers/metabolism , Blood Platelets/drug effects , Carnitine/pharmacology , DNA Methylation/drug effects , DNA, Mitochondrial/drug effects , Methylamines/pharmacology , Oxides/pharmacology , Aged , Atherosclerosis/drug therapy , Blood Platelets/metabolism , Cardiovascular System/drug effects , Cardiovascular System/metabolism , Dietary Supplements , Female , Humans , Lipid Metabolism/drug effects , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Pilot Projects
7.
ChemMedChem ; 14(20): 1783-1794, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31359587

ABSTRACT

Gi -protein-biased agonists with minimal ß-arrestin recruitment represent opportunities to overcome the serious adverse effects of human mu opioid receptor (µ-OR) agonists and developing alternative and safe treatments for pain. In order to discover novel non-morphinan opioid receptor agonists, we applied hierarchical virtual screening of our in-house database against a pharmacophore based on modeling the active conformations of opioid receptors. We discovered an initial hit compound, a novel µ-OR agonist with a pyrazoloisoquinoline scaffold. We applied computational R-group screening to this compound and synthesized 14 derivatives predicted to be the best. Of these, a new Gi -protein-biased compound, 1-{5-(3-chlorophenyl)-7,8-dimethoxy-3-[4-(methylsulfonyl)benzyl]-3H-pyrazolo[3,4-c]isoquinolin-1-yl}-N,N-dimethylmethanamine, showed an EC50 value of 179 nm against the µ-OR. This resulted in significant pain relief for mice in the phase II period of formalin response tests. This study provides a new strategy to identify diverse sets of promising compounds that might prove useful for the development of drugs that target other G-protein-coupled receptors.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Discovery , Methylamines/pharmacology , Pain/drug therapy , Receptors, Opioid, mu/agonists , Analgesics, Opioid/chemistry , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Formaldehyde/administration & dosage , Humans , Ligands , Methylamines/chemistry , Molecular Docking Simulation , Molecular Structure , Pain/chemically induced , Rats , Structure-Activity Relationship
8.
Exp Cell Res ; 382(1): 111451, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31173767

ABSTRACT

Trimethylamine N-oxide (TMAO) promotes atherosclerosis in association with the functions of endothelial cells. Clock and Bmal1, as two main components of molecular circadian clock, play important regulatory roles during progression of atherogenesis. However, whether Clock and Bmal1 are involved in the regulation of endothelial proliferation disturbed by TMAO are unclear. We observed that cell proliferation of human umbilical vein endothelial cells (HUVECs) was inhibited after exposed to TMAO for 24 h. Besides, TMAO caused increased expression of lncRNA-NEAT1, Clock and Bmal1, and inhibited MAPK pathways. While MAPK pathways were blocked, the expression of Clock and Bmal1 was elevated. NEAT1 showed a circadian rhythmic expression in HUVECs, and its overexpression reduced cell proliferation. Knockdown or overexpression of NEAT1 might decrease or increase the expression of Clock and Bmal1 respectively, while raised or suppressed the expression of MAPK pathways correspondingly. Asparagus extract (AE) was found to improve the TMAO-reduced HUVECs proliferation. Moreover, it ameliorated the disorders of NEAT1, Clock, Bmal1, and MAPK signaling pathways induced by TMAO. Therefore, our findings indicated that NEAT1 regulating Clock-Bmal1 via MAPK pathways was involved in TMAO-repressed HUVECs proliferation, and AE improved endothelial proliferation by TMAO, proposing a novel mechanism for cardiovascular disease prevention.


Subject(s)
Asparagaceae/chemistry , Circadian Rhythm/drug effects , Gene Expression Regulation/physiology , Methylamines/toxicity , Plant Extracts/pharmacology , RNA, Long Noncoding/physiology , ARNTL Transcription Factors/antagonists & inhibitors , ARNTL Transcription Factors/biosynthesis , ARNTL Transcription Factors/genetics , Atherosclerosis/genetics , Atherosclerosis/physiopathology , CLOCK Proteins/biosynthesis , CLOCK Proteins/genetics , Cell Division/drug effects , Circadian Rhythm/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Methylamines/pharmacology , Plant Stems/chemistry , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology
9.
Curr Opin Lipidol ; 30(2): 125-133, 2019 04.
Article in English | MEDLINE | ID: mdl-30664014

ABSTRACT

PURPOSE OF REVIEW: Not all of the risk of cardiovascular disease can be explained by diet and genetics, and the human microbiome, which lies at the interface of these two factors, may help explain some of the unaccounted risk. This review examines some of the well established links between the microbiome and cardiovascular health, and proposes relatively unexplored associations. RECENT FINDINGS: Byproducts of microbial metabolism are associated with health and disease: Trimethylamine N oxide is associated with atherosclerosis; whereas short-chain fatty acids are associated with decreased inflammation and increased energy expenditure. More broadly, a large number of association studies have been conducted to explore the connections between bacterial taxa and metabolic syndrome. In contrast, the relationship between the microbiome and triglycerides levels remains poorly understood. SUMMARY: We suggest that deeper understanding of the molecular mechanisms that drive linkages between the microbiome and disease can be determined by replacing 16S rRNA gene sequencing with shotgun metagenomic sequencing or other functional approaches. Furthermore, to ensure translatability and reproducibility of research findings, a combination of multiple different complementary '-omic' approaches should be employed.


Subject(s)
Atherosclerosis/microbiology , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/immunology , Lipid Metabolism/immunology , Metabolic Syndrome/microbiology , Methylamines/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Bile Acids and Salts/immunology , Bile Acids and Salts/metabolism , Carnitine/immunology , Carnitine/metabolism , Choline/immunology , Choline/metabolism , Energy Metabolism/genetics , Energy Metabolism/immunology , Fatty Acids, Volatile/immunology , Gastrointestinal Microbiome/genetics , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Lipid Metabolism/genetics , Metabolic Syndrome/genetics , Metabolic Syndrome/immunology , Metabolic Syndrome/pathology , Methylamines/immunology , Methylamines/pharmacology , Phosphatidylcholines/immunology , Phosphatidylcholines/metabolism , RNA, Ribosomal, 16S/genetics , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Triglycerides/immunology , Triglycerides/metabolism
10.
Reprod Domest Anim ; 53(4): 904-913, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29733477

ABSTRACT

The effect of organic phosphorus on metabolic, haematological and hormonal status, restoration of ovarian functions and conception rate in anoestrous Farafra ewes in subtropics were evaluated. Anoestrous Farafra ewes (n = 24; 34.72 ± 0.52 kg body weight) were allocated into two equal groups: control and phosphorus groups. The ewes of phosphorus group were treated with sodium 4-dimethylamino-2-methyl-phenyl-phosphonate as an organic bound phosphorous twice a week for successive 3 weeks. Ovarian follicle development and corpora lutea were checked three times a week till occurrence of oestrus using ultrasonography while pregnancy was confirmed at 30 days post-service. Plasma metabolites, reproductive hormones, thyroid hormones and minerals were detected at weeks -2, -1, 0 (mating day) and + 4 weeks post-oestrus. Phosphorus group had significantly (p < .05) short interval to oestrous resumption if compared to control ewes (2.1 ± 0.8 weeks vs. 4.6 ± 1.1 weeks). In addition, phosphorous supplementation significantly (p < .05) increased the number of antral follicles (developed and their sizes in addition to sizes of corpora lutea (8.72 ± 0.3 mm vs. 7.46 ± 0.9 mm) as well. Number of services per conception (2.6 vs. 1.4; p < .01) was higher in control group than that of phosphorus group. Pregnancy rate (80 vs. 50%) was significantly (p < .01) higher in phosphorus group when compared to control. White blood cells in treated ewes (10.8 ± 0.44; p < .05) and monocytes (2.93 ± 0.13; p < .01) were higher than that of control group (white blood cells; 9.53 ± 0.50 and monocytes; 2.24 ± 0.14). Metabolic parameters did not differ between phosphorus and control groups during different times of treatment. It could be concluded that phosphorous administration to anoestrous Farafra ewes in subtropics could improve reproductive performance and restore ovarian activity at the end of spring and early summer.


Subject(s)
Climate , Energy Metabolism/drug effects , Methylamines/pharmacology , Organophosphonates/pharmacology , Reproduction/drug effects , Seasons , Sheep/physiology , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Diet/veterinary , Dietary Supplements , Hot Temperature , Methylamines/administration & dosage , Organophosphonates/administration & dosage , Stress, Physiological
11.
Inflammopharmacology ; 26(1): 261-271, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28451776

ABSTRACT

BACKGROUND: We have developed a novel compound from acetylsalicylic acid (ASA) and 2-amino-2-(hydroxymethyl)-1,3-propanediol (Tris) precursors with ASA-like anti-inflammatory efficacy and reduced the mucosa-damaging side-effects. Our aim was to examine local and remote consequences of ASA-Tris administration in 2-,4-,6-trinitrobenzene-sulfonic acid (TNBS)-induced colitis as compared to ASA or mesalamine (5-aminosalicylate) treatment. METHODS: Sprague-Dawley rats were randomized to five groups (n = 6, each), and TNBS enemas were performed. Group 1 was the negative control; group 2 was the untreated colitis group. 12 hour after colitis induction repeated doses of ASA, ASA-Tris (both 0.55 mmol/kg) and mesalamine (0.77 mmol/kg) were given 3 times daily for 3 days to groups 3-5. On day 3 of colitis, the in vivo histology of the colon and stomach was investigated. Tissue xanthine-oxidoreductase, myeloperoxidase, nitrite/nitrate changes, and circulating TNF-alpha levels were measured. In addition, liver mitochondria were examined with high-resolution respirometry to analyze alterations in the electron transport chain. RESULTS: TNBS enema significantly elevated inflammatory enzyme activities, NO production, TNF-alpha concentration, and induced morphological damage in the colon. ASA-treatment reduced the inflammatory marker levels and mucosal injury in the colon, but gastric tissue damage was present. ASA-Tris- and mesalamine-treatments significantly reduced the cytokine levels, inflammatory enzyme activities, and colonic mucosal damage without inducing gastric injury. Also, ASA significantly reduced the Complex IV-linked respiration of liver mitochondria, which was not observed after ASA-Tris-treatment. CONCLUSION: As compared to ASA, ASA-Tris conjugation provides significant protection against the colonic injury and cytokine-mediated progression of inflammatory events in experimental colitis without influencing the gastric epithelial structure.


Subject(s)
Aspirin/pharmacology , Colitis/drug therapy , Colon/drug effects , Methylamines/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colitis/metabolism , Colon/metabolism , Disease Models, Animal , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Mesalamine/pharmacology , Nitrates/metabolism , Nitrites/metabolism , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Trinitrobenzenesulfonic Acid/pharmacology , Tumor Necrosis Factor-alpha/metabolism
12.
Article in English | MEDLINE | ID: mdl-28735625

ABSTRACT

Many cellular actions of omega-3 fatty acids are mediated by two G protein-coupled receptors, FFA1 and FFA4, free fatty acid receptor (FFAR) family members that are activated by these dietary constituents. FFAR agonists inhibit proliferation of human prostate and breast cancer cells. Since omega-3 fatty acids can inhibit ovarian cancer cell growth, the current study tested the potential role of FFARs in the response. OVCAR3 and SKOV3 human ovarian cancer cell lines express mRNA for FFA1; FFA4 mRNA was detected at low levels in SKOV3 but not OVCAR3. Lysophosphatidic acid (LPA) and epidermal growth factor (EGF) stimulated proliferation of both cell lines; these responses were inhibited by eicosopentaneoic acid (EPA) and by GW9508, a synthetic FFAR agonist. The LPA antagonist Ki16425 also inhibited LPA- and EGF-induced proliferation; FFAR agonists had no further effect when added with Ki16425. The results suggest that FFARs are potential targets for ovarian cancer therapy.


Subject(s)
Cell Proliferation/drug effects , Isoxazoles/pharmacology , Methylamines/pharmacology , Neoplasm Proteins/agonists , Ovarian Neoplasms/drug therapy , Propionates/pharmacology , Receptors, G-Protein-Coupled/agonists , Cell Line, Tumor , Female , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
13.
Eur J Pharmacol ; 762: 459-63, 2015 Sep 05.
Article in English | MEDLINE | ID: mdl-26071852

ABSTRACT

Previous studies have shown that the administration of docosahexaenoic acid (DHA) or GW9508, a GPR40/FFA1 (free fatty acid receptor) agonist, facilitates ß-endorphin release in the arcuate nucleus of the hypothalamus in mice. However, the mechanisms mediating ß-endorphin release induced by GPR40/FFA1 agonists remain unknown. In this study, we focused on the changes in expression of hypothalamic prohormone convertase (PC) 2, which is a calcium-dependent subtilisin-related proteolytic enzyme. The intracerebroventricular injection of DHA or GW9508 significantly increased PC2 protein expression in the hypothalamus. This increase in PC2 expression was inhibited by pretreatment with GW1100, a GPR40/FFA1 antagonist. Furthermore, PC2 protein expression gradually increased over time after complete Freund's adjuvant. These increase in PC2 expression were inhibited by pretreatment with GW1100. However, GW1100 by itself had no effect on PC2 levels. Taken together, our findings suggest that activation of the hypothalamic GPR40/FFA1 signaling pathway may regulate ß-endorphin release via PC2, and regulate the endogenous pain control system.


Subject(s)
Gene Expression Regulation, Enzymologic , Hypothalamus/metabolism , Proprotein Convertase 2/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Gene Expression Regulation, Enzymologic/drug effects , Hypothalamus/drug effects , Male , Methylamines/pharmacology , Mice , Pain/metabolism , Pain/pathology , Propionates/pharmacology , Protein Transport/drug effects , Receptors, G-Protein-Coupled/agonists , Signal Transduction/drug effects
14.
J Neuroinflammation ; 11: 60, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24674717

ABSTRACT

BACKGROUND: Overnutrition and the ensuing hypothalamic inflammation is a major perpetuating factor in the development of metabolic diseases, such as obesity and diabetes. Inflamed neurons of the CNS fail to properly regulate energy homeostasis leading to pathogenic changes in glucose handling, feeding, and body weight. Hypothalamic neurons are particularly sensitive to pro-inflammatory signals derived locally and peripherally, and it is these neurons that become inflamed first upon high fat feeding. Given the prevalence of metabolic disease, efforts are underway to identify therapeutic targets for this inflammatory state. At least in the periphery, omega-3 fatty acids and their receptor, G-protein coupled receptor 120 (GPR120), have emerged as putative targets. The role for GPR120 in the hypothalamus or CNS in general is poorly understood. METHODS: Here we introduce a novel, immortalized cell model derived from the rat hypothalamus, rHypoE-7, to study GPR120 activation at the level of the individual neuron. Gene expression levels of pro-inflammatory cytokines were studied by quantitative reverse transcriptase-PCR (qRT-PCR) upon exposure to tumor necrosis factor α (TNFα) treatment in the presence or absence of the polyunsaturated omega-3 fatty acid docosahexaenoic acid (DHA). Signal transduction pathway involvement was also studied using phospho-specific antibodies to key proteins by western blot analysis. RESULTS: Importantly, rHypoE-7 cells exhibit a transcriptional and translational inflammatory response upon exposure to TNFα and express abundant levels of GPR120, which is functionally responsive to DHA. DHA pretreatment prevents the inflammatory state and this effect was inhibited by the reduction of endogenous GPR120 levels. GPR120 activates both AKT (protein kinase b) and ERK (extracellular signal-regulated kinase); however, the anti-inflammatory action of this omega-3 fatty acid (FA) receptor is AKT- and ERK-independent and likely involves the GPR120-transforming growth factor-ß-activated kinase 1 binding protein (TAB1) interaction as identified in the periphery. CONCLUSIONS: Taken together, GPR120 is functionally active in the hypothalamic neuronal line, rHypoE-7, wherein it mediates the anti-inflammatory actions of DHA to reduce the inflammatory response to TNFα.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Neurons/drug effects , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Cell Line, Transformed , Docosahexaenoic Acids/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Hypothalamus/cytology , I-kappa B Proteins/metabolism , Immunoprecipitation , Methylamines/pharmacology , Propionates/pharmacology , Pyridines/pharmacology , RNA, Small Interfering/pharmacology , Rats , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
15.
PLoS One ; 8(12): e81563, 2013.
Article in English | MEDLINE | ID: mdl-24349089

ABSTRACT

GPR40 has been reported to be activated by long-chain fatty acids, such as docosahexaenoic acid (DHA). However, reports studying functional role of GPR40 in the brain are lacking. The present study focused on the relationship between pain regulation and GPR40, investigating the functional roles of hypothalamic GPR40 during chronic pain caused using a complete Freund's adjuvant (CFA)-induced inflammatory chronic pain mouse model. GPR40 protein expression in the hypothalamus was transiently increased at day 7, but not at days 1, 3 and 14, after CFA injection. GPR40 was co-localized with NeuN, a neuron marker, but not with glial fibrillary acidic protein (GFAP), an astrocyte marker. At day 1 after CFA injection, GFAP protein expression was markedly increased in the hypothalamus. These increases were significantly inhibited by the intracerebroventricular injection of flavopiridol (15 nmol), a cyclin-dependent kinase inhibitor, depending on the decreases in both the increment of GPR40 protein expression and the induction of mechanical allodynia and thermal hyperalgesia at day 7 after CFA injection. Furthermore, the level of DHA in the hypothalamus tissue was significantly increased in a flavopiridol reversible manner at day 1, but not at day 7, after CFA injection. The intracerebroventricular injection of DHA (50 µg) and GW9508 (1.0 µg), a GPR40-selective agonist, significantly reduced mechanical allodynia and thermal hyperalgesia at day 7, but not at day 1, after CFA injection. These effects were inhibited by intracerebroventricular pretreatment with GW1100 (10 µg), a GPR40 antagonist. The protein expression of GPR40 was colocalized with that of ß-endorphin and proopiomelanocortin, and a single intracerebroventricular injection of GW9508 (1.0 µg) significantly increased the number of neurons double-stained for c-Fos and proopiomelanocortin in the arcuate nucleus of the hypothalamus. Our findings suggest that hypothalamic GPR40 activated by free long chain fatty acids might have an important role in this pain control system.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Chronic Pain/drug therapy , Docosahexaenoic Acids/pharmacology , Hyperalgesia/drug therapy , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/physiopathology , Astrocytes/drug effects , Astrocytes/metabolism , Benzoates/pharmacology , Chronic Pain/chemically induced , Chronic Pain/metabolism , Chronic Pain/physiopathology , DNA-Binding Proteins , Disease Models, Animal , Flavonoids/pharmacology , Freund's Adjuvant , Gene Expression , Glial Fibrillary Acidic Protein , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Injections, Intraventricular , Male , Methylamines/pharmacology , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pain Management , Piperidines/pharmacology , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Propionates/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Time Factors , beta-Endorphin/genetics , beta-Endorphin/metabolism
16.
Mol Cell Endocrinol ; 361(1-2): 40-50, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22484460

ABSTRACT

The number of adipocyte progenitors is determined early in foetal and neonatal development in a process which may be altered by gender and excess nutrient intake, and which in turn determines fat mass in adulthood and the risk of developing obesity. Here we investigate the hypothesis that excess nutrients, in this case the long chain fatty acid palmitate, can program differentiating stem cells towards white fat lineages. The experiments were performed on mouse embryonic stem cells in chemically defined media (CDM) supplemented with bone morphogenetic protein 4 (BMP4) and all trans-retinoic acid (RA). Subsequent treatment for 21 days with palmitate not only promoted the expression of adipocyte markers and monolocular lipid deposition as observed by RT/QPCR and immunocytochemistry, but also stimulated a considerable enrichment in adipocytes as measured by flow cytometry and a lipolytic response to catecholamines. Palmitate increased protein levels of adiponectin that is preferentially expressed in subcutaneous fat, while inhibiting IGFBP2 and IGFBP3 that are associated with visceral fat. In keeping with this finding, palmitate also increased expression of the subcutaneous markers Shox2 and Twist1 and oestrogenising enzymes. Collectively, these results suggest that palmitate induces differentiation towards subcutaneous fat and that this could occur through its oestrogenising effects on the preadipocyte, suggesting a role for palmitate in programming fat development towards a metabolically favourable profile.


Subject(s)
Adipocytes, White/cytology , Adipocytes, White/drug effects , Cell Differentiation/drug effects , Cell Lineage/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Palmitates/pharmacology , Adipocytes, White/metabolism , Animals , Biomarkers/metabolism , Bone Morphogenetic Protein 4/pharmacology , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/genetics , Cell Line , Dehydroepiandrosterone/pharmacology , Embryonic Stem Cells/metabolism , Gene Expression Regulation/drug effects , Gonadal Steroid Hormones/metabolism , Methylamines/pharmacology , Mice , PPAR gamma/metabolism , Propionates/pharmacology , Time Factors , Tretinoin/pharmacology
17.
Brain Res ; 1432: 74-83, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22137657

ABSTRACT

G-protein receptor (GPR) 40 is known to be activated by docosahexaenoic acid (DHA). However, reports studying the role and functions (including pain regulation) of GPR40 in the brain are lacking. We investigated the involvement of GPR40 in the brain on DHA-induced antinociceptive effects. Expression of GPR40 protein was observed in the olfactory bulb, striatum, hippocampus, midbrain, hypothalamus, medulla oblongata, cerebellum and cerebral cortex in the brain as well as the spinal cord, whereas GPR120 protein expression in these areas was not observed. Intracerebroventricular (i.c.v.), but not intrathecal (i.t.) injection of DHA (25 and 50µg/mouse) and GW9508 (a GPR40- and GPR120-selective agonist; 0.1 and 1.0µg/mouse) significantly reduced formalin-induced pain behavior. These effects were inhibited by pretreatment with the µ opioid receptor antagonist ß-funaltrexamine (ß-FNA), naltrindole (δ opioid receptor antagonist) and anti-ß-endorphin antiserum. The κ opioid receptor antagonist norbinaltorphimine (nor-BNI) did not affect the antinociception of DHA or GW9508. Furthermore, the immunoreactivity of ß-endorphin in the hypothalamus increased at 10 and 20min after i.c.v. injection of DHA and GW9508. These findings suggest that DHA-induced antinociception via ß-endorphin release may be mediated (at least in part) through GPR40 signaling in the supraspinal area, and may provide valuable information on a novel therapeutic approach for pain control.


Subject(s)
Analgesics/pharmacology , Brain/physiology , Docosahexaenoic Acids/physiology , Pain Management/methods , Receptors, G-Protein-Coupled/physiology , Animals , Brain/metabolism , Drug Synergism , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular/methods , Male , Methylamines/pharmacology , Mice , Narcotic Antagonists/pharmacology , Pain Measurement , Propionates/pharmacology , Receptors, Opioid/drug effects , Receptors, Opioid/physiology , beta-Endorphin/physiology
18.
Nature ; 472(7341): 57-63, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21475195

ABSTRACT

Metabolomics studies hold promise for the discovery of pathways linked to disease processes. Cardiovascular disease (CVD) represents the leading cause of death and morbidity worldwide. Here we used a metabolomics approach to generate unbiased small-molecule metabolic profiles in plasma that predict risk for CVD. Three metabolites of the dietary lipid phosphatidylcholine--choline, trimethylamine N-oxide (TMAO) and betaine--were identified and then shown to predict risk for CVD in an independent large clinical cohort. Dietary supplementation of mice with choline, TMAO or betaine promoted upregulation of multiple macrophage scavenger receptors linked to atherosclerosis, and supplementation with choline or TMAO promoted atherosclerosis. Studies using germ-free mice confirmed a critical role for dietary choline and gut flora in TMAO production, augmented macrophage cholesterol accumulation and foam cell formation. Suppression of intestinal microflora in atherosclerosis-prone mice inhibited dietary-choline-enhanced atherosclerosis. Genetic variations controlling expression of flavin monooxygenases, an enzymatic source of TMAO, segregated with atherosclerosis in hyperlipidaemic mice. Discovery of a relationship between gut-flora-dependent metabolism of dietary phosphatidylcholine and CVD pathogenesis provides opportunities for the development of new diagnostic tests and therapeutic approaches for atherosclerotic heart disease.


Subject(s)
Cardiovascular Diseases/metabolism , Cardiovascular Diseases/microbiology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Phosphatidylcholines/metabolism , Animals , Atherosclerosis/chemically induced , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/microbiology , Betaine/blood , Betaine/metabolism , Biomarkers/blood , Biomarkers/metabolism , Cardiovascular Diseases/blood , Cardiovascular Diseases/diagnosis , Cholesterol, HDL/blood , Choline/administration & dosage , Choline/blood , Choline/metabolism , Choline/pharmacology , Diet/adverse effects , Dietary Fats/blood , Dietary Fats/metabolism , Dietary Fats/pharmacology , Female , Gene Expression Regulation , Germ-Free Life , Humans , Liver/enzymology , Macrophages/metabolism , Metabolomics , Methylamines/blood , Methylamines/metabolism , Methylamines/pharmacology , Mice , Mice, Inbred C57BL , Oxygenases/genetics , Oxygenases/metabolism , Phenotype , Phosphatidylcholines/administration & dosage , Phosphatidylcholines/blood , Phosphatidylcholines/pharmacology , Receptors, Scavenger/metabolism , Risk Assessment
19.
Proc Natl Acad Sci U S A ; 107(12): 5569-74, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20212125

ABSTRACT

Zellweger spectrum disorder (ZSD) is a heterogeneous group of diseases with high morbidity and mortality caused by failure to assemble normal peroxisomes. There is no therapy for ZSD, but management is supportive. Nevertheless, one-half of the patients have a phenotype milder than classic Zellweger syndrome and exhibit a progressive disease course. Thus, patients would benefit if therapies became available and were instituted early. Recent reports indicate several interventions that result in partial peroxisome recovery in ZSD fibroblasts. To identify drugs that recover peroxisome functions, we expressed a GFP-peroxisome targeting signal 1 reporter in fibroblasts containing the common disease allele, PEX1-p.Gly843Asp. The GFP reporter remained cytosolic at baseline, and improvement in peroxisome functions was detected by the redistribution of the GFP reporter from the cytosol to the peroxisome. We established a high-content screening assay based on this phenotype assay and evaluated 2,080 small molecules. The cells were cultured in chemical for 2 days and then, were fixed and imaged by epifluorescent microscopy on a high-content imaging platform. We identified four compounds that partially recover matrix protein import, and we confirmed three using independent assays. Our results suggest that PEX1-p.G843D is a misfolded protein amenable to chaperone therapy.


Subject(s)
Membrane Proteins/genetics , Peroxisomes/physiology , Zellweger Syndrome/drug therapy , Zellweger Syndrome/genetics , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/physiology , Amino Acid Substitution , Betaine/pharmacology , Cells, Cultured , Drug Evaluation, Preclinical , Genes, Reporter , Glycerol/pharmacology , Green Fluorescent Proteins/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/physiology , Methylamines/pharmacology , Mutation, Missense , Peroxisome-Targeting Signal 1 Receptor , Peroxisomes/drug effects , Peroxisomes/genetics , Protein Folding/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Zellweger Syndrome/physiopathology
20.
Biol Pharm Bull ; 30(6): 1123-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17541165

ABSTRACT

The objectives of this study were to investigate the antihyperglycemic effect of Cephalotaxus sinensis leaves and to identify the active components. The antihyperglycemic effect of various fractions (FA, FB, FC, FD) of the 80% ethanol extract of the leaves was evaluated in streptozotocin (STZ)-induced diabetic rats. Among the tested fractions, FC was the most active. FC (0.48 g/kg) given orally for 10 d reduced significantly (p<0.001) the blood glucose of STZ-induced diabetic rats. The food and water intakes of FC (0.48 g/kg)-treated diabetic rats were reduced significantly (p<0.001) when compared to the 0.5% carboxymethyl cellulose (CMC)-treated diabetic rats. The activity-guided fractionation of the ethanol extract of C. sinensis leaves furnished three flavonoid compounds, apigenin-5-O-[alpha-L-rhamnopyranosyl-(1-->4)-6-O-beta-D-acetylglucopyranoside] (1), apigenin (2), and apigenin-5-O-[alpha-L-rhamnopyranosyl-(1-->4)-6-O-beta-D-glucopyranoside] (3). The elevation of GLUT-4 protein level in membrane preparations from mice adipocytes was detected by Western blot analysis after adipocytes were pre-incubated with FC (0.1, 1, 10 mg/ml), apigenin (0.1, 2 mg/ml) and apigenin-5-O-[alpha-L-rhamnopyranosyl-(1-->4)-6-O-beta-D-acetylglucopyranoside] (0.1, 2 mg/ml), respectively. Phytochemical investigation and HPLC-DAD analysis of FC indicated that the flavonoids were the major constituents in this fraction. These results suggest that the fraction from C. sinensis leaves is a promising drug for the treatment of diabetes, and that the flavonoids from this plant are the active constituents.


Subject(s)
Cephalotaxus/anatomy & histology , Flavonoids/chemistry , Glucose Transporter Type 4/metabolism , Hypoglycemic Agents/pharmacology , Plant Leaves/chemistry , Adipocytes/cytology , Animals , Blood Glucose/analysis , Blotting, Western , Carboxymethylcellulose Sodium/pharmacology , Cell Membrane/chemistry , Chromatography, High Pressure Liquid , Diabetes Mellitus, Experimental , Dose-Response Relationship, Drug , Drinking/drug effects , Drug Evaluation, Preclinical , Eating/drug effects , Flavonoids/pharmacology , Glipizide/pharmacology , Insulin/pharmacology , Male , Methylamines/pharmacology , Mice , Molecular Structure , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL