Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 553
Filter
1.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39000550

ABSTRACT

The effect of the modulators of the mitochondrial ATP-dependent potassium channel (mitoKATP) on the structural and biochemical alterations in the substantia nigra and brain tissues was studied in a rat model of Parkinson's disease induced by rotenone. It was found that, in experimental parkinsonism accompanied by characteristic motor deficits, both neurons and the myelin sheath of nerve fibers in the substantia nigra were affected. Changes in energy and ion exchange in brain mitochondria were also revealed. The nucleoside uridine, which is a source for the synthesis of the mitoKATP channel opener uridine diphosphate, was able to dose-dependently decrease behavioral disorders and prevent the death of animals, which occurred for about 50% of animals in the model. Uridine prevented disturbances in redox, energy, and ion exchanges in brain mitochondria, and eliminated alterations in their structure and the myelin sheath in the substantia nigra. Cytochemical examination showed that uridine restored the indicators of oxidative phosphorylation and glycolysis in peripheral blood lymphocytes. The specific blocker of the mitoKATP channel, 5-hydroxydecanoate, eliminated the positive effects of uridine, suggesting that this channel is involved in neuroprotection. Taken together, these findings indicate the promise of using the natural metabolite uridine as a new drug to prevent and, possibly, stop the progression of Parkinson's disease.


Subject(s)
Mitochondria , Potassium Channels , Rotenone , Uridine , Animals , Uridine/pharmacology , Uridine/metabolism , Rats , Potassium Channels/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Male , Disease Models, Animal , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Parkinson Disease/pathology , Substantia Nigra/metabolism , Substantia Nigra/drug effects , Substantia Nigra/pathology , Neuroprotective Agents/pharmacology , Oxidative Phosphorylation/drug effects , Rats, Wistar , Decanoic Acids/pharmacology , Hydroxy Acids/pharmacology
2.
J Microbiol Biotechnol ; 34(6): 1314-1321, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38938006

ABSTRACT

Branched-chain hydroxy acids (BCHAs), produced by lactic acid bacteria, have recently been suggested as bioactive compounds contributing to the systemic metabolism and modulation of the gut microbiome. However, the relationship between BCHAs and gut microbiome remains unclear. In this study, we investigated the effects of BCHAs on the growth of seven different families in the gut microbiota. Based on in vitro screening, both 2-hydroxyisovaleric acid (HIVA) and 2-hydroxyisocaproic acid (HICA) stimulated the growth of Lactobacillaceae and Bifidobacteriaceae, with HIVA showing a significant growth promotion. Additionally, we observed not only the growth promotion of probiotic Lactobacillaceae strains but also growth inhibition of pathogenic B. fragilis in a dosedependent manner. The production of HIVA and HICA varied depending on the family of the gut microbiota and was relatively high in case of Lactobacillaceae and Lachnosporaceae. Furthermore, HIVA and HICA production by each strain positively correlated with their growth variation. These results demonstrated gut microbiota-derived BCHAs as active metabolites that have bacterial growth modulatory effects. We suggest that BCHAs can be utilized as active metabolites, potentially contributing to the treatment of diseases associated with gut dysbiosis.


Subject(s)
Gastrointestinal Microbiome , Hydroxy Acids , Gastrointestinal Microbiome/drug effects , Hydroxy Acids/metabolism , Hydroxy Acids/pharmacology , Probiotics , Caproates/metabolism , Caproates/pharmacology , Bacteria/drug effects , Bacteria/metabolism , Bacteria/growth & development , Bacteria/genetics , Bacteria/classification , Lactobacillaceae/metabolism , Humans , Pentanoic Acids/metabolism
3.
Nat Commun ; 13(1): 1343, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35292630

ABSTRACT

Meta-analyses suggest that yogurt consumption reduces type 2 diabetes incidence in humans, but the molecular basis of these observations remains unknown. Here we show that dietary yogurt intake preserves whole-body glucose homeostasis and prevents hepatic insulin resistance and liver steatosis in a dietary mouse model of obesity-linked type 2 diabetes. Fecal microbiota transplantation studies reveal that these effects are partly linked to the gut microbiota. We further show that yogurt intake impacts the hepatic metabolome, notably maintaining the levels of branched chain hydroxy acids (BCHA) which correlate with improved metabolic parameters. These metabolites are generated upon milk fermentation and concentrated in yogurt. Remarkably, diet-induced obesity reduces plasma and tissue BCHA levels, and this is partly prevented by dietary yogurt intake. We further show that BCHA improve insulin action on glucose metabolism in liver and muscle cells, identifying BCHA as cell-autonomous metabolic regulators and potential mediators of yogurt's health effects.


Subject(s)
Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/prevention & control , Fermentation , Hydroxy Acids/pharmacology , Mice , Mice, Obese , Yogurt
4.
Orthop Surg ; 14(4): 704-713, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35195339

ABSTRACT

OBJECTIVES: To evaluate the efficacy of resistance training (RT) combined with beta-hydroxy-beta-methylbutyric acid (HMB) in the treatment of elderly patients with sarcopenia after hip replacement. METHODS: From January 1, 2018 to December 31, 2018, 200 elderly patients (68 men, mean age 76.3 years and 137 women, mean age 79.1 years) who experienced femoral neck fracture with sarcopenia after hip arthroplasty were assigned to four groups: RT + HMB group, RT group, HMB group, and negative control group. Baseline data, body composition, grip strength, Barthel index (BI), Harris hip score (HHS), and visual analog scale score (VAS) were compared among the four groups before and 3 months after surgery. RESULTS: A total of 177 participants completed the trial, including 43 in the HMB + RT group, 44 in the HMB group, 45 in the RT group, and 45 in the negative control group. At the 3-month follow-up, the body composition and grip strength of the HMB + RT group and RT group were significantly improved compared with those before operation. The HMB group had no significant change, while the measures in the negative control group significantly decreased. Postoperative BI and HSS did not reach pre-injury levels in any of the four groups, but postoperative VAS score was significantly improved. However, there was no significant difference in BI, HSS, or VAS among the four groups. CONCLUSION: RT, with or without HMB supplementation, can effectively improve body composition and grip strength in elderly patients with sarcopenia after hip replacement at short-term follow-up. Simultaneously, use of exclusive HMB supplementation alone may also help to prevent decreases in muscle mass and grip strength in these patients.


Subject(s)
Arthroplasty, Replacement, Hip , Resistance Training , Sarcopenia , Aged , Dietary Supplements , Female , Humans , Hydroxy Acids/pharmacology , Male , Muscle, Skeletal , Sarcopenia/pathology , Sarcopenia/prevention & control , Valerates/pharmacology , Valerates/therapeutic use
5.
Eur J Pharmacol ; 901: 174095, 2021 Jun 15.
Article in English | MEDLINE | ID: mdl-33862063

ABSTRACT

Previous clinical studies have shown that anisodamine could improve no-reflow phenomenon and prevent reperfusion arrhythmias, but whether this protective effect is related to the antagonism of the M-type cholinergic receptor or other potential mechanisms is uncertain. The aim of the present study was to investigate the role of the mitochondrial ATP-sensitive potassium channel (mitoK ATP ) in cardioprotective effect of anisodamine against ischemia/reperfusion injury. Anisodamine and 5- hydroxydecanoic acid were used to explore the relationship between anisodamine and mitoK ATP . Using a Langendorff isolated heart ischemia/reperfusion injury model, hemodynamic parameters and reperfusion ventricular arrhythmia were evaluated; in addition, changes in myocardial infarct size, cTnI from coronary effluent and myocardial ultrastructure, as well as ATP, MDA and SOD in myocardial tissues, were detected. In the hypoxia/reoxygenation injury model of neonatal rat cardiomyocyte, cTnI release in the culture medium and levels of ATP, MDA and SOD in cardiomyocytes and mitochondrial membrane potential, were analyzed. Overall, anisodamine could significantly improve the hemodynamic indexes of isolated rat heart injured by ischemia/reperfusion, reduce the occurrence of ventricular reperfusion arrhythmia and myocardial infarction area, and improve the ultrastructural damage of myocardium and mitochondria. The in vitro results demonstrated that anisodamine could improve mitochondrial energy metabolism, reduce oxidative stress and stabilize mitochondrial membrane potential. The cardioprotective effects were significantly inhibited by 5-hydroxydecanoic acid. In conclusion, this study suggests that the opening of mitoK ATP could play an important role in the protective effect of anisodamine against myocardial ischemia/reperfusion injury.


Subject(s)
Cardiotonic Agents/therapeutic use , Mitochondria, Heart/drug effects , Myocardial Reperfusion Injury/prevention & control , Potassium Channels/drug effects , Reperfusion Injury/prevention & control , Solanaceous Alkaloids/therapeutic use , Adenosine Triphosphate/metabolism , Animals , Arrhythmias, Cardiac/prevention & control , Decanoic Acids/pharmacology , Energy Metabolism/drug effects , Hemodynamics/drug effects , Hydroxy Acids/pharmacology , In Vitro Techniques , Male , Malondialdehyde/metabolism , Rats , Rats, Sprague-Dawley , Solanaceous Alkaloids/antagonists & inhibitors , Superoxide Dismutase/metabolism
6.
Biochem Biophys Res Commun ; 551: 161-167, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33740623

ABSTRACT

Physiological oxygen concentration (physioxia) ranges from 1 to 8% in human tissues while many researchers cultivate mammalian cells under an atmospheric concentration of 21% (hyperoxia). Oxygen is one of the significant gases which functions in human cells including energy production in mitochondria, metabolism in peroxidase, and transcription of various genes in company with HIF (Hypoxia-inducible factors) in the nucleus. Thus, mammalian cell culture should be deliberated on the oxygen concentration to mimic in vivo physiology. Here, we studied if the cultivation of human skin cells under physiological conditions could affect skin significant genes in barrier functions and dermal matrix formation. We further examined that some representative active ingredients in dermatology such as glycolic acid, gluconolactone, and salicylic acid work in different ways depending on the oxygen concentration. Taken together, we present the importance of oxygen concentration in skin cell culture for proper screening of novel ingredients as well as the mechanistic study of skin cell regulation.


Subject(s)
Hydroxy Acids/pharmacology , Oxygen/pharmacology , Skin , Cell Line , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Fibroblasts/drug effects , Fibroblasts/metabolism , Filaggrin Proteins , Gene Expression Regulation , Gluconates/metabolism , Glycolates/metabolism , Humans , Keratin-1/genetics , Keratinocytes/drug effects , Keratinocytes/metabolism , Lactones/metabolism , Matrix Metalloproteinase 1/genetics , Oxygen/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , S100 Proteins/genetics , Salicylic Acid/metabolism , Skin/cytology , Skin/drug effects , Skin/metabolism
7.
Clin Exp Hypertens ; 43(5): 462-473, 2021 Jul 04.
Article in English | MEDLINE | ID: mdl-33775188

ABSTRACT

BACKGROUND: Signal transduction of Angiotensin II (Ang II) induced autophagy and its role in Ang II-induced dysfunction of HUVECs are still unclear. METHODS: HUVECs are stimulated with different doses of Ang II (10-9-10-5 mol/L) for different time (6-48 hours). Autophagy-related protein markers: LC3, Beclin-1 and SQSTM1/p62 are measured by western blot. RESULTS: Incubation with Ang II increases autophagic flux (Beclin-1, autophagosomes formation, and degradation of SQSTM1/p62, LC3-I). Increased autophagic levels are inhibited by pretreatment with Ang II type 1 receptor (AT1) blocker (Candesartan), NADPH Oxidase inhibitor (apocycin), mitochondrial KATP channels inhibitor (5-hydroxydecanoate, 5HD). 3-Methyladenine (inhibitors of autophagy) and rapamycin (activator of autophagy) respectively inhibits or activates Ang II-induced autophagy levels. Ang II decreases phosphorylation of endothelial nitric oxide synthase (eNOS) and NO production in HUVECs. L-NAME (NOS inhibitor) totally mimics the actions of Ang II on eNOS, NO production and autophagy levels. Rapamycin further decreases NO production combined with Ang II. Silence Atg5 completely reverses Ang II-activated autophagy levels. CONCLUSIONS: Our results demonstrate that Ang II stimulation increases autophagy levels via AT1 receptor, NADPH oxidase, mitochondrial KATP channel, eNOS, Atg5 signal pathway in HUVECs, and activation of autophagy contributes to Ang II induced dysfunction of HUVECs.


Subject(s)
Angiotensin II/toxicity , Autophagy , Human Umbilical Vein Endothelial Cells/pathology , Acetophenones/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Autophagy-Related Proteins/metabolism , Benzimidazoles/pharmacology , Biphenyl Compounds/pharmacology , Decanoic Acids/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Hydroxy Acids/pharmacology , Models, Biological , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Sirolimus/pharmacology , Tetrazoles/pharmacology , Time Factors
8.
Int J Mol Sci ; 22(2)2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33440780

ABSTRACT

Since the European Union (EU) announced their animal testing ban in 2013, all animal experiments related to cosmetics have been prohibited, creating a demand for alternatives to animal experiments for skin studies. Here, we investigated whether an ex vivo live porcine skin model can be employed to study the safety and skin barrier-improving effects of hydroxyacids widely used in cosmetics for keratolytic peels. Glycolic acid (1-10%), salicylic acid (0.2-2%), and lactobionic acid (1.2-12%) were used as representative substances for α-hydroxyacid (AHA), ß-hydroxyacid (BHA), and polyhydroxyacid (PHA), respectively. When hydroxyacids were applied at high concentrations on the porcine skin every other day for 6 days, tissue viability was reduced to 50-80%, suggesting that the toxicity of cosmetic ingredients can be evaluated with this model. Based on tissue viability, the treatment scheme was changed to a single exposure for 20 min. The protective effects of a single exposure of hydroxyacids on skin barrier function were evaluated by examining rhodamine permeability and epidermal structural components of barrier function using immunohistochemistry (IHC) and immunofluorescence (IF) staining. Lactobionic acid (PHAs) improved skin barrier function most compared to other AHAs and BHAs. Most importantly, trans-epidermal water loss (TEWL), an important functional marker of skin barrier function, could be measured with this model, which confirmed the significant skin barrier-protective effects of PHAs. Collectively, we demonstrated that the ex vivo live full-thickness porcine skin model can be an excellent alternative to animal experiments for skin studies on the safety and efficacy of cosmetic ingredients.


Subject(s)
Skin Physiological Phenomena , Skin/metabolism , Animals , Biomarkers , Epidermis/drug effects , Epidermis/metabolism , Fluorescent Antibody Technique , Histocytochemistry , Humans , Hydroxy Acids/chemistry , Hydroxy Acids/pharmacology , In Vitro Techniques , Permeability , Rhodamines/pharmacology , Salicylic Acid/chemistry , Salicylic Acid/pharmacology , Skin/cytology , Skin/drug effects , Skin Physiological Phenomena/drug effects , Swine , Tissue Culture Techniques
9.
Biomed Res Int ; 2020: 3039184, 2020.
Article in English | MEDLINE | ID: mdl-33134375

ABSTRACT

Inflammatory response during myocardial ischemia reperfusion injury (MIRI) is essential for cardiac healing, while excessive inflammation extends the infarction and promotes adverse cardiac remodeling. Understanding the mechanism of these uncontrolled inflammatory processes has a significant impact during the MIRI therapy. Here, we found a critical role of ATP-sensitive potassium channels (KATP) in the inflammatory response of MIRI and its potential mechanism and explored the effects of Panax Notoginseng Saponins (PNS) during this possess. Rats underwent 40 min ischemia by occlusion of the left anterior descending (LAD) coronary artery and 60 min of reperfusion. PNS was treated at the corresponding time point before operation; 5-hydroxydecanoate (5-HD) and glybenclamide (Gly) (or Nicorandil (Nic)) were used as pharmacological blocker (or nonselective opener) of KATP. Cardiac function and pathomorphology were evaluated and a set of molecular signaling experiments was tested. KATP current density was measured by patch-clamp. Results revealed that in MIRI, PNS pretreatment restored cardiac function, reduced infarct size, and ameliorated inflammation through KATP. However, inhibiting KATP by 5-HD and Gly significantly reversed the effects, including NLRP3 inflammasome and inflammatory mediators IL-6, MPO, TNF-α, and MCP-1. Moreover, PNS inhibited the phosphorylation and nuclear translocation of NF-κB in I/R myocardium when the KATP was activated. Importantly, PNS promoted the expression of subunits and activation of KATP. The study uncovered KATP served as a new potential mechanism during PNS modulating MIRI-induced inflammation and promoting injured heart recovery. The manipulation of KATP could be a potential therapeutic approach for MIRI and other inflammatory diseases.


Subject(s)
Cardiotonic Agents/pharmacology , Drugs, Chinese Herbal/chemistry , KATP Channels/genetics , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/drug therapy , Saponins/pharmacology , Animals , Cardiotonic Agents/isolation & purification , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Decanoic Acids/pharmacology , Gene Expression Regulation , Glyburide/pharmacology , Hydroxy Acids/pharmacology , Inflammation , Interleukin-6/genetics , Interleukin-6/metabolism , KATP Channels/agonists , KATP Channels/antagonists & inhibitors , KATP Channels/metabolism , Male , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Nicorandil/pharmacology , Patch-Clamp Techniques , Peroxidase/genetics , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Saponins/isolation & purification , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
Int J Biol Macromol ; 163: 298-308, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32603731

ABSTRACT

Molecular properties and biological functions of Pyrenaican SF-1 as a novel biological macromolecule extracted from a fungal isolate were studied. The isolate was identified as Daldinia pyrenaica on the basis of 5.8S rDNA sequencing. Pyrenaican SF-1 was obtained from the culture filtrate of the fungal isolate. The partial characterization of biochemical structure of Pyrenaican SF-1 was conducted. The fungal extract was also tested for the treatment of AGS, MDA and HeLa cell lines to assess cells proliferation, cells cycle and apoptosis. Furthermore, Pyrenaican SF-1 extract was tested for its antibacterial and antioxidant activity. Initial chemical analysis revealed that Pyrenaican SF-1 extract was composed of various monosaccharides such as d-glucose, D- mannitol, D-arabinose and ß-D-ribopyranose. In vitro study indicated that Pyrenaican SF-1 could effectively elevate percentage of apoptosis and necrosis of cancer cells and block cell cycle phase of the control group. The fungal extract could inhibit proliferation of Hela and MDA cell up to 67% and 56%, respectively. Moreover, Pyrenaican SF-1 represented a strong antioxidant activity compared to that one obtained from vitamin C. On the other hand, Pyrenaican SF-1 exhibited growth inhibitory effects against different Gram-negative and Gram-positive bacterial strains. Pyrenaican SF-1 can be considered as a bioactive macromolecule with promising application in pharmaceutical and medical sectors.


Subject(s)
Ascomycota/chemistry , Biological Products/chemistry , Biological Products/pharmacology , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gas Chromatography-Mass Spectrometry , Humans , Hydroxy Acids/chemistry , Hydroxy Acids/pharmacology , Spectroscopy, Fourier Transform Infrared , Viscosity , beta Carotene
11.
BMC Mol Cell Biol ; 21(1): 31, 2020 Apr 19.
Article in English | MEDLINE | ID: mdl-32306897

ABSTRACT

BACKGROUND: Cytoprotection afforded by mitochondrial ATP-sensitive K+-channel (mKATP-channel) opener diazoxide (DZ) largely depends on the activation of potassium cycle with eventual modulation of mitochondrial functions and ROS production. However, generally these effects were studied in the presence of Mg∙ATP known to block K+ transport. Thus, the purpose of our work was the estimation of DZ effects on K+ transport, K+ cycle and ROS production in rat liver mitochondria in the absence of Mg∙ATP. RESULTS: Without Mg·ATP, full activation of native mKATP-channel, accompanied by the increase in ATP-insensitive K+ uptake, activation of K+-cycle and respiratory uncoupling, was reached at ≤0.5 µM of DZ,. Higher diazoxide concentrations augmented ATP-insensitive K+ uptake, but not mKATP-channel activity. mKATP-channel was blocked by Mg·ATP, reactivated by DZ, and repeatedly blocked by mKATP-channel blockers glibenclamide and 5-hydroxydecanoate, whereas ATP-insensitive potassium transport was blocked by Mg2+ and was not restored by DZ. High sensitivity of potassium transport to DZ in native mitochondria resulted in suppression of mitochondrial ROS production caused by the activation of K+-cycle on sub-micromolar scale. Based on the oxygen consumption study, the share of mKATP-channel in respiratory uncoupling by DZ was found. CONCLUSIONS: The study of mKATP-channel activation by diazoxide in the absence of MgATP discloses novel, not described earlier, aspects of mKATP-channel interaction with this drug. High sensitivity of mKATP-channel to DZ results in the modulation of mitochondrial functions and ROS production by DZ on sub-micromolar concentration scale. Our experiments led us to the hypothesis that under the conditions marked by ATP deficiency affinity of mKATP-channel to DZ can increase, which might contribute to the high effectiveness of this drug in cardio- and neuroprotection.


Subject(s)
Adenosine Triphosphate/metabolism , Diazoxide/pharmacology , Mitochondria, Liver/drug effects , Potassium Channels/metabolism , Potassium/metabolism , Animals , Decanoic Acids/pharmacology , Energy Metabolism/drug effects , Female , Glyburide/pharmacology , Hydroxy Acids/pharmacology , Ion Transport/drug effects , Ion Transport/genetics , KATP Channels/metabolism , Magnesium/metabolism , Mitochondria, Liver/metabolism , Oxygen Consumption/drug effects , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels/genetics , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
12.
Molecules ; 24(22)2019 Nov 08.
Article in English | MEDLINE | ID: mdl-31717454

ABSTRACT

Endophytes have been recognized as a source for structurally novel and biologically active secondary metabolites. Among the host plants for endophytes, some medicinal plants that produce pharmaceuticals have been reported to carry endophytes, which could also produce bioactive secondary metabolites. In this study, the medicinal plant Aconitum carmichaeli was selected as a potential source for endophytes. An endophytic microorganism, Aureobasidium pullulans AJF1, harbored in the flower of Aconitum carmichaeli, was cultured on a large scale and extracted with an organic solvent. Extensive chemical investigation of the extracts resulted in isolation of three lipid type compounds (1-3), which were identified to be (3R,5R)-3,5-dihydroxydecanoic acid (1), (3R,5R)-3-(((3R,5R)-3,5-dihydroxydecanoyl)oxy)-5-hydroxydecanoic acid (2), and (3R,5R)-3-(((3R,5R)-5-(((3R,5R)-3,5-dihydroxydecanoyl)oxy)-3-hydroxydecanoyl)oxy)-5-hydroxydecanoic acid (3) by chemical methods in combination with spectral analysis. Compounds 2 and 3 had new structures. Absolute configurations of the isolated compounds (1-3) were established using modified Mosher's method together with analysis of NMR data for their acetonide derivatives. All the isolates (1-3) were evaluated for antibiotic activities against Escherichia coli, Staphylococcus aureus, Bacillus subtilis, Pseudomonas aeruginosa, and their cytotoxicities against MCF-7 cancer cells. Unfortunately, they showed low antibiotic activities and cytotoxic activities.


Subject(s)
Ascomycota/metabolism , Decanoic Acids/chemistry , Decanoic Acids/metabolism , Hydroxy Acids/chemistry , Hydroxy Acids/metabolism , Aconitum/genetics , Aconitum/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Ascomycota/genetics , Bacteria/drug effects , Decanoic Acids/chemical synthesis , Decanoic Acids/pharmacology , Humans , Hydroxy Acids/chemical synthesis , Hydroxy Acids/pharmacology , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Molecular Structure
13.
Eur J Pharmacol ; 862: 172636, 2019 Nov 05.
Article in English | MEDLINE | ID: mdl-31491405

ABSTRACT

Several studies have reported that CORM-3, a water-soluble carbon monoxide releasing molecule, elicits cardioprotection against myocardial infarction but the mechanism remains to be investigated. Numerous reports indicate that inhibition of pH regulators, the Na+/H+ exchanger (NHE) and Na+/HCO3- symporter (NBC), protect cardiomyocytes from hypoxia/reoxygenation injury by delaying the intracellular pH (pHi) recovery at reperfusion. Our goal was to explore whether CORM-3-mediated cytoprotection involves the modulation of pH regulation. When added at reoxygenation, CORM-3 (50 µM) reduced the mortality of cardiomyocytes exposed to 3 h of hypoxia and 2 h of reoxygenation in HCO3--buffered solution. This effect was lost when using inactive iCORM-3, which is depleted of CO and used as control, thus implicating CO as the mediator of this cardioprotection. Interestingly, the cardioprotective effect of CORM-3 was abolished by switching to a bicarbonate-free medium. This effect of CORM-3 was also inhibited by 5-hydroxydecanoate, a mitochondrial ATP-dependent K+ (mKATP) channel inhibitor (500 µM) or PD098059, a MEK1/2 inhibitor (10 µM). In additional experiments and in the absence of hypoxia-reoxygenation, intracellular pH was monitored in cardiomyocytes exposed to cariporide to block NHE activity. CORM-3 inhibited alkalinisation and this effect was blocked by PD098059 and 5-HD. In conclusion, CORM-3 protects the cardiomyocyte against hypoxia-reoxygenation injury by inhibiting a bicarbonate transporter at reoxygenation, probably the Na+/HCO3- symporter. This cardioprotective effect of CORM-3 requires the activation of mKATP channels and the activation of MEK1/2.


Subject(s)
Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Organometallic Compounds/pharmacology , Protective Agents/pharmacology , Animals , Carbon Monoxide/metabolism , Cell Hypoxia/drug effects , Cell Survival/drug effects , Cells, Cultured , Culture Media/chemistry , Decanoic Acids/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Flavonoids/pharmacology , Humans , Hydrogen-Ion Concentration , Hydroxy Acids/pharmacology , KATP Channels/antagonists & inhibitors , KATP Channels/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Male , Mice , Mitochondria/chemistry , Mitochondria/drug effects , Mitochondria/pathology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/pathology , Organometallic Compounds/therapeutic use , Primary Cell Culture , Protective Agents/therapeutic use
14.
Sci Rep ; 9(1): 9418, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31263219

ABSTRACT

The effect of the activation of the mitochondrial ATP-dependent potassium channel (mitoKATP) on the ultrastructure of rat lung in acute hypoxic hypoxia (7% of oxygen in nitrogen, exposure 30 min) was studied. It was shown that uridine, a precursor of the mitoKATP activator UDP, exerted a protective effect against hypoxic damage to the lung. The administration of uridine to animals prior to hypoxia decreased the number of mitochondria with altered ultrastructure and prevented the hypoxia-induced mitochondrial swelling. Uridine also protected the epithelial, interstitial and endothelial layers of the air-blood barrier from the hypoxia-induced hyperhydration. The protective action of uridine against hypoxia-induced lung injury was eliminated by the selective blocker of mitoKATP 5-hydroxydecanoate. These data suggest that one of the mechanisms of the positive effect of uridine is related to the activation of the mitoKATP channel, which, according to the literature and our data, is involved in the protection of tissues from hypoxia and leads to adaptation to it. A possible role of uridine in the maintenance of the mitochondrial structure upon hypoxia-induced lung injury and the optimization of oxygen supply of the organism is discussed.


Subject(s)
Lung Injury/drug therapy , Protective Agents/therapeutic use , Uridine/therapeutic use , Animals , Decanoic Acids/pharmacology , Hydroxy Acids/pharmacology , Hypoxia/pathology , Lung Injury/etiology , Male , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/ultrastructure , Oxygen/toxicity , Potassium Channels/chemistry , Potassium Channels/metabolism , Protective Agents/pharmacology , Rats , Rats, Wistar , Uridine/pharmacology
15.
PLoS One ; 14(4): e0215104, 2019.
Article in English | MEDLINE | ID: mdl-30978206

ABSTRACT

A mild ischemic load applied after a lethal ischemic insult reduces the subsequent ischemia-reperfusion injury, and is called ischemic postconditioning (PostC). We studied the effect of ischemic PostC on synaptic glutamate release using a whole-cell patch-clamp technique. We recorded spontaneous excitatory post-synaptic currents (sEPSCs) from CA1 pyramidal cells in mouse hippocampal slices. The ischemic load was perfusion of artificial cerebrospinal fluid (ACSF) equilibrated with mixed gas (95% N2 and 5% CO2). The ischemic load was applied for 7.5 min, followed by ischemic PostC 30 s later, consisting of three cycles of 15 s of reperfusion and 15 s of re-ischemia. We found that a surging increase in sEPSCs frequency occurred during the immediate-early reperfusion period after the ischemic insult. We found a significant positive correlation between cumulative sEPSCs and the number of dead CA1 neurons (r = 0.70; p = 0.02). Ischemic PostC significantly suppressed this surge of sEPSCs. The mitochondrial KATP (mito-KATP) channel opener, diazoxide, also suppressed the surge of sEPSCs when applied for 15 min immediately after the ischemic load. The mito-KATP channel blocker, 5-hydroxydecanoate (5-HD), significantly attenuated the suppressive effect of both ischemic PostC and diazoxide application on the surge of sEPSCs. These results suggest that the opening of mito-KATP channels is involved in the suppressive effect of ischemic PostC on synaptic glutamate release and protection against neuronal death. We hypothesize that activation of mito-KATP channels prevents mitochondrial malfunction and breaks mutual facilitatory coupling between glutamate release and Ca2+ entry at presynaptic sites.


Subject(s)
Glutamic Acid/metabolism , Hippocampus/metabolism , Ischemic Postconditioning/methods , Mitochondria/metabolism , Myocardial Reperfusion Injury/prevention & control , Neurons/metabolism , Potassium Channels/metabolism , Adenosine Triphosphate/metabolism , Animals , Decanoic Acids/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , Hippocampus/pathology , Hydroxy Acids/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Neurons/drug effects , Neurons/pathology , Potassium Channel Blockers/pharmacology , Potassium Channels/chemistry
16.
Bull Exp Biol Med ; 166(6): 806-810, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31020578

ABSTRACT

We studied the effects of in vivo modulation of activity of mitochondrial ATP-dependent potassium channel (mitoKATP) by uridine on the morphofunctional state of mitochondria in rat cardiomyocytes under conditions of acute hypoxia. Preinjection of uridine to animals reduced the number of structurally modified mitochondria, but had practically no effect on their morphogenesis after hypoxia. Uridine in vivo stimulated the formation of micromitochondria and their release into the cytoplasm. The number of "maternal" mitochondria containing three and more new micromitochondria, increased as well. The use of mitoKATP blocker 5-hydroxydecanoate in parallel with uridine abolished its protective effect, as it significantly inhibited the formation of micromitochondria in rat cardiomyocytes after acute hypoxic exposure.


Subject(s)
Hypoxia/metabolism , Myocardium/metabolism , Potassium Channels/metabolism , Uridine/pharmacology , Animals , Cell Hypoxia , Decanoic Acids/antagonists & inhibitors , Decanoic Acids/pharmacology , Hydroxy Acids/antagonists & inhibitors , Hydroxy Acids/pharmacology , Hypoxia/drug therapy , Hypoxia/pathology , Male , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar
17.
Cell Physiol Biochem ; 49(2): 717-727, 2018.
Article in English | MEDLINE | ID: mdl-30165359

ABSTRACT

BACKGROUND/AIMS: The phosphatidylinositol-3-kinase -AKT (PI3K-AKT) is an important intracellular signal pathway in regulating cell proliferation, differentiation and apoptosis. In previous studies, we've demonstrated that PI3K-AKT pathway protects cardiomyocytes from ischemic and hypoxic apoptosis through mitochondrial function. However, the molecular mechanisms underlying hypoxia-induced cardiomyocyte apoptosis via PI3K-AKT pathway remain ill-defined. Here, we addressed this question. METHODS: Cardiomyocytes were exposed to hypoxia, with/without different inhibitors and then protein levels were assessed by Western blotting. RESULTS: We found that the PI3K-AKT pathway was activated in cardiomyocytes that were exposed to hypoxia. Moreover, the phospho-AKT (pAKT) translocated from cytosol to mitochondria via mitochondrial adenosine triphosphate-dependent potassium (mitoKATP), leading to an increase in cytochrome c oxidase (CcO) activity to suppress apoptosis. On the other hand, the mitoKATP specific blocker, 5-hydroxydecanote (5-HD), or suppression of CcO using siRNA, inhibited the pAKT mitochondrial translocation to maintain the CcO activity, resulting in mitochondrial dysfunction and cellular apoptosis induced by hypoxia. CONCLUSION: These findings suggest that the anti-apoptotic effect of the PI3K-AKT pathway through pAKT translocation to mitochondrial via mitoKATP may be conducted through modification of CcO activity.


Subject(s)
Apoptosis , Cell Hypoxia , Phosphatidylinositol 3-Kinases/metabolism , Potassium Channels/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cells, Cultured , Chromones/pharmacology , Decanoic Acids/pharmacology , Electron Transport Complex IV/antagonists & inhibitors , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Hydroxy Acids/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Morpholines/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phosphoinositide-3 Kinase Inhibitors , Potassium Channels/chemistry , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
18.
Pharmacol Rep ; 70(4): 720-729, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29935398

ABSTRACT

BACKGROUND: The current study is focusing on the role of brain natriuretic peptide (BNP), a substrate of dipeptidyl peptidase-4 (DPP-4) enzyme, and its signaling survival pathway in the cardioprotective mechanism of sitagliptin, a DPP-4 inhibitor. METHODS: Male Wistar rats were randomized into 7 groups, sham, I/R, KT-5823 (selective protein kinase (PK) G inhibitor), 5-HD (selective mito-KATP channel blocker), sitagliptin (300mg/kg, po), sitagliptin+KT-5823, and sitagliptin+5-HD. Sitagliptin was administered for 3 days prior to induction of coronary I/R, while either KT-5823 or 5-HD was administered intravenously 5min before coronary ligation. RESULTS: Pretreatment with sitagliptin provided significant protection against I/R injury as manifested by decreasing, percentage of infarct size, suppressing the elevated ST segment, reducing the increased cardiac enzymes, as well as DPP-4 activity and elevating both heart rate (HR) and left ventricular developed pressure (LVDP). However, the addition of either blocker to sitagliptin regimen reversed partly its cardioprotective effects. Although I/R increased BNP content, it unexpectedly decreased that of cGMP; nevertheless, sitagliptin elevated both parameters, an effect that was not affected by the use of the two blockers. On the molecular level, sitagliptin decreased caspase-3 activity and downregulated the mRNA levels of BNP, Bax, and Cyp D, while upregulated that of Bcl2. The use of either KT-5823 or 5-HD with sitagliptin hindered its effect on the molecular markers tested. CONCLUSIONS: The results of the present study suggest that the cardioprotective effect of sitagliptin is mediated partly, but not solely, through the BNP/cGMP/PKG survival signaling pathway.


Subject(s)
Natriuretic Peptide, Brain/metabolism , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Sitagliptin Phosphate/pharmacology , Animals , Carbazoles/pharmacology , Cardiotonic Agents/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Caspase 3/biosynthesis , Cyclic GMP/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/biosynthesis , Decanoic Acids/pharmacology , Dipeptidyl Peptidase 4/metabolism , Hemodynamics/drug effects , Hydroxy Acids/pharmacology , Male , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Sitagliptin Phosphate/antagonists & inhibitors , bcl-2-Associated X Protein/biosynthesis
19.
Molecules ; 23(4)2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29642579

ABSTRACT

AHAs are organic acids with one hydroxyl group attached to the alpha position of the acid. AHAs including glycolic acid, lactic acid, malic acid, tartaric acid, and citric acid are often used extensively in cosmetic formulations. AHAs have been used as superficial peeling agents as well as to ameliorate the appearance of keratoses and acne in dermatology. However, caution should be exercised in relation to certain adverse reactions among patients using products with AHAs, including swelling, burning, and pruritus. Whether AHAs enhance or decrease photo damage of the skin remains unclear, compelling us to ask the question, is AHA a friend or a foe of the skin? The aim of this manuscript is to review the various biological effects and mechanisms of AHAs on human keratinocytes and in an animal model. We conclude that whether AHA is a friend or foe of human skin depends on its concentration. These mechanisms of AHAs are currently well understood, aiding the development of novel approaches for the prevention of UV-induced skin damage.


Subject(s)
Acne Vulgaris/drug therapy , Hydroxy Acids/pharmacology , Keratinocytes/drug effects , Animals , Cosmetics , Humans , Hydroxy Acids/adverse effects , Hydroxy Acids/chemistry , Hydroxy Acids/therapeutic use , Keratinocytes/cytology , Keratinocytes/metabolism , Molecular Structure , Signal Transduction/drug effects
20.
Int Urol Nephrol ; 49(11): 2079-2086, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28748494

ABSTRACT

PURPOSE: The present study investigated the putative mechanisms underlying effects of KATP channel on high glucose (HG)-induced mesangial cell proliferation and tissue inhibitors of metalloproteinases (TIMP)-2 and Collagen IV production. METHODS: Rat mesangial cells were subjected to whole cell patch clamp to record the KATP channel currents under high glucose (HG, 30 mM) condition. Cell proliferation was measured using a CCK-8 assay. The production of TIMP-2 and Collagen IV and AMP-activated protein kinase (AMPK)-signaling pathway activity was assessed by ELISA and Western blotting, respectively. AMPK agonist (AICAR) was used to analyze the role of this kinase. The expression of KATP subunit (Kir6.1, Kir6.2, SUR1, SUR2A and SUR2B) was examined using quantitative real-time PCR (RT-PCR). RESULTS: We found that HG was significant decreases in the expression of Kir6.1, SUB2A and SUB2B, three subunits of KATP, TIMP-2 production, KATP channel activity and AMPK activity, while it promoted the cell proliferation and Collagen IV production in rat mesangial cells. Pretreatment with KATP selective opener (diazoxide, DZX) significantly inhibited HG-induced mesangial cell proliferation, Collagen IV production and decrease in KATP channel activity in rat mesangial cells, which were reversed by pretreatment of 5-hydroxydecanoate, a selective inhibitor of KATP. Moreover, AICAR pretreatment inhibited HG-induced decrease in KATP channel activity. CONCLUSIONS: Taken together, activating AMPK-KATP signaling may protect against HG-induced mesangial cell proliferation and Collagen IV production, and, thereby, provides new insights into the molecular mechanisms underlying early diabetic nephropathy (DN).


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Proliferation/drug effects , Collagen Type IV/biosynthesis , Glucose/pharmacology , KATP Channels/metabolism , Mesangial Cells/metabolism , Sulfonylurea Receptors/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cells, Cultured , Decanoic Acids/pharmacology , Diabetic Nephropathies/metabolism , Diazoxide/pharmacology , Glucose/administration & dosage , Hydroxy Acids/pharmacology , Hypoglycemic Agents/pharmacology , KATP Channels/antagonists & inhibitors , Male , Mesangial Cells/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology , Signal Transduction/drug effects , Sulfonylurea Receptors/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinase-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL