ABSTRACT
Generation 4 of polyamidoamine dendrimer (G4-PAMAM) has several biological effects due to its tridimensional globular structure, repetitive branched amides, tertiary amines, and amino-terminal subunit groups liked to a common core. G4-PAMAM is cytotoxic due to its positive charges. However, its cytotoxicity could increase in cancer cells due to the excessive intracellular negative charges in these cells. Furthermore, this work reports G4-PAMAM chemical structural characterization using UHPLC-QTOF-MS/MS (LC-MS) by electrospray ionization to measure its population according to its positive charges. Additionally, the antiproliferative effects and intracellular localization were explored in the HMC-1 and K-562 cell lines by confocal microscopy. The LC-MS results show that G4-PAMAM generated multivalent mass spectrum values, and its protonated terminal amino groups produced numerous positive charges, which allowed us to determine its exact mass despite having a high molecular weight. Additionally, G4-PAMAM showed antiproliferative activity in the HMC-1 tumor cell line after 24 h (IC50 = 16.97 µM), 48 h (IC50 = 7.02 µM) and 72 h (IC50 = 5.98 µM) and in the K-562 cell line after 24 h (IC50 = 15.14 µM), 48 h (IC50 = 14.18 µM) and 72 h (IC50 = 9.91 µM). Finally, our results showed that the G4-PAMAM dendrimers were located in the cytoplasm and nucleus in both tumor cell lines studied.
Subject(s)
Dendrimers/pharmacology , Leukemia/drug therapy , Leukemia/metabolism , Nylons/pharmacology , Antineoplastic Agents/analysis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chromatography, Liquid , Dendrimers/analysis , Dendrimers/pharmacokinetics , Drug Screening Assays, Antitumor/methods , Humans , Inhibitory Concentration 50 , Intracellular Space/drug effects , Intracellular Space/metabolism , K562 Cells , Leukemia/pathology , Nylons/analysis , Nylons/pharmacokinetics , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Tissue DistributionABSTRACT
Several reports have suggested that photobiomodulation, owing to its analgesic, anti-inflammatory, and healing effects, may be an effective therapeutic option for local effects of snakebites when the availability and accessibility of conventional serum therapy are inefficient and far from medical care centers. Although there have been studies that demonstrate the application of photobiomodulation in the treatment of local adverse events due to snakebites from snakes of the genus Bothrops, its role in the activation of leukocytes, particularly macrophages, has not been evaluated. Here, we assessed the effect of light-emitting diode (LED) treatment on macrophage activation induced by B. jararacussu venom (BjV). LED treatment caused an increase in the viability of macrophages incubated with BjV. This treatment reduced reactive oxygen species (ROS) and nitric oxide (NO) production by macrophages after incubation with BjV. However, LED treatment did not interfere with IL-1ß and IL-10 production by macrophages after incubation with BjV. In conclusion, this study showed that LED treatment has the potential to be used in combination with conventional serum therapy to prevent or minimize the progression of local to severe symptoms after Bothrops envenomation.
Subject(s)
Bothrops , Crotalid Venoms/toxicity , Low-Level Light Therapy/instrumentation , Macrophages/radiation effects , Semiconductors , Snake Bites/immunology , Snake Bites/radiotherapy , Animals , Cell Survival/drug effects , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Intracellular Space/radiation effects , Macrophages/immunology , Male , Mice , Nitric Oxide/biosynthesis , Reactive Oxygen Species/metabolism , Snake Bites/metabolism , Snake Bites/pathology , Superoxides/metabolismABSTRACT
The effect of N-geranyl-ethane-1,2-diamine dihydochloride (GIB24), a synthetic diamine, was assayed against different developmental forms of the parasitic protozoan Trypanosoma cruzi (strain Dm28c). The compound was effective against culture epimastigote forms (IC50/24h = 5.64 µM; SI = 16.4) and intracellular amastigotes (IC50/24h = 12.89 µM; SI = 7.18), as detected by the MTT methodology and by cell counting, respectively. Incubation of epimastigotes for 6h with 6 µM GIB24 (IC50/24h value) resulted in significant dissipation of the mitochondrial membrane potential, prior to permeabilization of the plasma membrane. Rounded epimastigotes with cell size reduction were observed by scanning electron microscopy. These morpho-physiological changes induced by GIB24 suggest an incidental death process. Treatment of infected Vero cells did not prevent the intracellular amastigotes from completing the intracellular cycle. However, there was a decrease in the number of released parasites, increasing the ratio amastigotes/trypomastigotes. Proteomic analysis of 15 µM GIB24 resistant epimastigotes indicated that the compound acts mainly on mitochondrial components involved in the Krebs cycle and in maintaining the oxidative homeostasis of the parasites. Our data suggest that GIB24 is active against the main morphological forms of T. cruzi.
Subject(s)
Diamines/pharmacology , Drug Resistance , Intracellular Space/drug effects , Proteomics , Terpenes/chemistry , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/growth & development , Animals , Chlorocebus aethiops , Diamines/chemistry , Intracellular Space/parasitology , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/metabolism , Vero CellsABSTRACT
A new source of pectin with a cytotoxic effect on glioblastoma cells is presented. A homogeneous GWP-FP-S fraction (Mw of 29,170â¯gâ¯mol-1) was obtained by fractionating the crude pectin extract (GW) from Campomanesia xanthocarpa pulp. According to the monosaccharide composition, the GWP-FP-S was composed of galacturonic acid (58.8%), arabinose (28.5%), galactose (11.3%) and rhamnose (1.1%), comprising 57.7% of homogalacturonans (HG) and 42.0% of type I rhamnogalacturonans (RG-I). These structures were characterized by chromatographic and spectroscopic methods; GW and GWP-FP-S fractions were evaluated by MTT and crystal violet assays for their cytotoxic effects. Both fractions induced cytotoxicity (15.55-37.65%) with concomitant increase in the cellular ROS levels in human glioblastoma cells at 25-400⯵g mL-1, after 48â¯h of treatment, whereas no cytotoxicity was observed for normal NIH 3T3 cells. This is the first report of in vitro bioactivity and the first investigation of the antitumor potential of gabiroba pectins.
Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Glioblastoma/pathology , Pectins/chemistry , Pectins/pharmacology , Pimenta/chemistry , Antineoplastic Agents/isolation & purification , Cell Line, Tumor , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Monosaccharides/analysis , Pectins/isolation & purification , Reactive Oxygen Species/metabolismABSTRACT
Cardiac arrhythmias are among the most important pathologies that lead to sudden death. The discovery of new therapeutic options against arrhythmias with low adverse effects is of paramount importance. Farnesol is found in essential oils with antioxidant, anti-inflammatory and cardioprotective properties. The aim of this work was to investigate the effects of farnesol on the contractile and electrophysiological properties in rat heart and evaluate its antiarrhythmic action. It was evaluated farnesol effects on the left ventricular developed pressure, ECG, potassium (Ik) and L-type Ca2+ currents (ICa,L), action potential, intracellular Ca2+ transient, Ca2+ sparks and waves and reactive oxygen species production. Antiarrhythmic activity of farnesol was determined in vivo and ex vivo. The results showed that 50⯵M farnesol did not alter left ventricular developed pressure, heart rate, ECG parameters and intracellular Ca2+ transient but reduced ICa,L. Farnesol reduced action potential duration at 90% repolarization. Notably, farnesol improved arrhythmia score and the incidence of the most severe arrhythmias. Farnesol attenuated the generation of reactive oxygen species, Ca2+ sparks and waves in isolated cardiomyocytes submitted to Ca2+ overload. In conclusion, farnesol has antiarrhythmic effect mediated by reducing of ICa,L and IK along with a decrease of reactive oxygen species production and normalized Ca2+ sparks and waves.
Subject(s)
Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Calcium Channel Blockers/pharmacology , Calcium/metabolism , Farnesol/pharmacology , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Calcium Channel Blockers/therapeutic use , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Electrocardiography/drug effects , Farnesol/therapeutic use , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Myocardial Contraction/drug effects , Oxygen/metabolism , Potassium/metabolism , Rats , Rats, Wistar , Ventricular Dysfunction, Left/drug therapyABSTRACT
Stroke is the second cause of death and first cause of physical disability around the world; it affects the brain parenchyma through oxygen deficiency and spreads excitotoxicity. The complexity of the disease has made it difficult to find effective therapies. It is necessary to identify new treatments that effectively act within the narrow therapeutic window but also offer long-term protection poststroke. Our previous work found that oral linalool reversed the hippocampal and peripheral pro-inflammatory phospholipidomic biomarkers in ischemic rats; based on these observations, the "proof of concept" was to demonstrate that intranasal administration of linalool has a faster delivery to the central nervous system to protect it after focal ischemia in Wistar rats. The ischemic animals treated with linalool (25â¯mg/kg) showed a decrease in infarct volume at 24â¯h and seven days, and the treated animals had better neurological and motor skills at both poststroke times. Additionally, one month after daily intranasal administration of linalool, the ischemic rats showed improved relearning performance in the Morris water maze test. They also exhibited a reduction in microgliosis and decreased COX2, IL-1Beta and Nrf2 markers in the cerebral cortex and hippocampus. In astrocyte and microglial cultures, linalool reduced pro-inflammation and had a potent effect on microglial cells, generating Nrf2 subcellular redistribution under glutamate excitotoxicity conditions. Together, our findings indicate an acute and chronic recovery after ischemia induced by a daily intranasal puff of linalool, which mainly acts on microglial populations with anti-inflammatory actions.
Subject(s)
Acyclic Monoterpenes/administration & dosage , Acyclic Monoterpenes/pharmacology , Brain Ischemia/pathology , Microglia/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Acyclic Monoterpenes/pharmacokinetics , Administration, Intranasal , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain Ischemia/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Microglia/metabolism , Microglia/pathology , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacokinetics , Protein Transport/drug effects , Rats , Rats, Wistar , Tissue DistributionABSTRACT
Mechanisms underlying the vasorelaxant effects of the synthetic nitro compound, trans-4-methoxy-ß-nitrostyrene (T4MN) were studied in isolated small resistance arteries from spontaneously hypertensive rats (SHRs). T4MN caused vasorelaxation in endothelium-intact third-order branches of the mesenteric artery pre-contracted with noradrenaline (NA). This effect was unchanged by indomethacin and atropine but was significantly reduced by endothelium removal, L-NAME, LY294002, glybenclamide, TEA, apamin, TRAM 34, or by the association of apamin and TRAM 34. Pretreatment with the sGC inhibitor, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) reduced the T4MN-induced relaxation in endothelium-intact, but not in denuded preparations. Incubation of small resistance arteries with T4MN increased nitric oxide (NO) production, an effect that was blocked by L-NAME. In Ca2+-free medium, T4MN inhibits the contractions induced by (i) NA, (ii) exogenous calcium through receptor- or voltage-operated Ca2+ channels and (iii) those evoked by Ca2+ influx through stores-operated Ca2+ channels activated by thapsigargin-induced Ca2+ store depletion. In contrast, T4MN was inert against the transient contraction induced by caffeine in Ca2+-free medium. In conclusion, T4MN induced effective vasorelaxant effects in isolated small resistance arteries from SHRs. This vasorelaxation seems to be mediated partly by an endothelium-dependent mechanism involving activation of Akt/eNOS/NO pathway and partly by an endothelium-independent mechanism through activation of sGC/cGMP/PKG pathway in vascular smooth muscle, leading to inhibition of Ca2+ influx from the extracellular milieu and IP3-sensitive intracellular Ca2+ release as well as activation of potassium channels.
Subject(s)
Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Styrenes/pharmacology , Vasodilation/drug effects , Animals , Blood Pressure/drug effects , Calcium Channels/metabolism , Calcium Signaling/drug effects , Enzyme Activation/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptors, Muscarinic/metabolism , Receptors, Prostaglandin/metabolism , Vasoconstriction/drug effectsABSTRACT
BACKGROUND: Low molecular weight 1,2,3-triazoles and naphthoquinones are endowed with various types of biological activity, such as against cancer, HIV and bacteria. However, in some cases, the conjugation of these two nuclei considerably increases their biological activities. OBJECTIVE: In this work, we decided to study the synthesis and screening of bis-naphthoquinones and xanthenes tethered to 1,2,3-triazoles against cancer cell lines, specifically the human breast cancer cell line MCF-7. RESULTS: Starting from lawsone and aryl-1H-1,2,3-triazole-4-carbaldehydes (10a-h) several new 7- (1-aryl-1H-1,2,3-triazol-4-yl)-6H-dibenzo[b,h]xanthene-5,6,8,13(7H)-tetraones (12a-h) and 3,3'- ((1-aryl-1H-1,2,3-triazol-4-yl)methylene)bis(2-hydroxynaphthalene-1,4-diones) 11a-h were synthesized and evaluated for their cytotoxic activities using the human breast cancer cell line MCF-7 and the non-tumor cell line MCF10A as control. We performed test of cell viability, cell proliferation, intracellular ATP content and cell cytometry to determine reactive oxygen species (ROS) formation. CONCLUSIONS: Based on these results, we found that compound 12a promotes ROS production, interfering with energy metabolism, cell viability and proliferation, and thus promoting whole cell damage.
Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Drug Design , Reactive Oxygen Species/metabolism , Triazoles/chemistry , Xanthenes/chemical synthesis , Xanthenes/pharmacology , Adenosine Triphosphate/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemistry Techniques, Synthetic , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , MCF-7 Cells , Xanthenes/chemistryABSTRACT
Phagocytosis, a process involved in host defense, requires coordination of a variety of signaling reactions. MT-II, a catalytically-inactive Lys49-PLA2¸ and MT-III, an active Asp49-PLA2 isolated from Bothrops asper snake venom, activate phagocytosis in macrophages. In this study the signal pathways mediating zymosan phagocytosis, focusing in lipidic second messengers, were investigated. Macrophages collected from male Swiss mouse peritoneum were obtained 96h after i.p. injection of thioglycollate. Phagocytosis was evaluated with non-opsonized zymosan in the presence or absence of specific inhibitors. Data showed that both venom PLA2s increased phagocytosis. Zileuton, Etoricoxib, PACOCF3 (5-LO, COX-2 and iPLA2 inhibitors, respectively), as well as WEB2170 (PAF receptor antagonist) significantly reduced phagocytosis induced by both venom PLA2s. However, Indomethacin (COX-1/COX-2 inhibitor) and Montelukast (CysL receptor antagonist) did not affect the toxins-induced phagocytosis. Moreover, while PACOCF3 (iPLA2 inhibitor), reduced the phagocytosis induced by MT-II and MT-III, AACOCF3 (cPLA2 inhibitor) significantly reduced the MT-II, but not MT-III-induced phagocytosis. These data suggest the effect of both sPLA2s depends on iPLA2 and that the effect of MT-II depends on activation of cPLA2. COX-2 and 5-LO-derived metabolites as well as PAF are involved in the signaling events required for phagocytosis induced by both venom sPLA2s.
Subject(s)
Bothrops , Crotalid Venoms/enzymology , Macrophages/cytology , Macrophages/drug effects , Phospholipases A2, Secretory/pharmacology , Signal Transduction/drug effects , Zymosan/metabolism , Animals , Dinoprostone/biosynthesis , Intracellular Space/drug effects , Intracellular Space/metabolism , Macrophages/metabolism , Male , Mice , Receptors, Immunologic/metabolism , p38 Mitogen-Activated Protein Kinases/metabolismABSTRACT
Sticholysin II (StII) is a pore-forming toxin of biomedical interest that belongs to the actinoporin protein family. Sticholysins are currently under examination as an active immunomodulating component of a vaccinal platform against tumoral cells and as a key element of a nucleic acids delivery system to cell cytosol. These proteins form pores in the plasma membrane leading to ion imbalance and cell lysis. However, the intracellular mechanisms triggered by actinoporins upon binding to membranes and its consequences for cell death are barely understood. Here, we have examined the cytotoxicity and intracellular responses induced by StII upon binding to human B-cell lymphoma Raji in vitro. StII cytotoxicity involves a functional actin cytoskeleton, induces cellular swelling, lysis and the concomitant release of cytosol content. In addition, StII induces calcium release mainly from the Endoplasmic Reticulum, activates Mitogen-Activated Protein Kinase ERK and impairs mitochondrial membrane potential. Furthermore, StII stimulates the expression of receptor interacting protein kinase 1 (RIP1), normally related to different forms of regulated cell death such as apoptosis and necroptosis. In correspondence, necrostatin-1, an inhibitor of this kinase, reduces StII cytotoxicity. However, the mechanism of cell death activated by StII does not involve caspases activation, typical molecular features of apoptosis and pyroptosis. Our results suggest that, beyond pore-formation and cell lysis, StII-induced cytotoxicity could involve other regulated intracellular mechanisms connected to RIP1-MEK1/2 -ERK1/2- pathways. This opens new perspectives and challenges the general point of view that these toxins induce a completely unregulated mechanism of necrotic cell death. This study contributes to a better understanding of the molecular mechanisms involved in toxin-cell interaction and the implications for cell functioning, with connotation for the exploitations of these toxins in clinical settings.
Subject(s)
Cell Death/drug effects , Cnidarian Venoms/toxicity , Cytotoxins/toxicity , Intracellular Space/drug effects , Intracellular Space/metabolism , Cell Line , Cell Membrane/drug effects , Cell Survival/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Signal Transduction/drug effectsABSTRACT
1-Nitro-2-phenylethene (NPe) induces a more potent vasorelaxant effect in rat aorta than its structural analog 1-nitro-2-phenylethane, but mediated through a different mechanism, independent of soluble guanylate cyclase (sGC) stimulation. We hypothesized that introducing an electron donor into the aromatic moiety might stabilize NPe, enhancing its potency and/or interaction with sGC. Therefore, trans-4-methoxy-ß-nitrostyrene (T4MN) was synthesized, and mechanisms underlying its vasorelaxant effects were studied in rat aortic ring preparations. In endothelium-intact preparations, T4MN fully relaxed contractions induced by phenylephrine (PHE) with a potency similar to that of its parent drug, NPe. This vasorelaxant effect that was unchanged by endothelium removal, pretreatment with L-NAME, indomethacin, or MDL-12,330A, but was significantly reduced by tetraethylammonium, 4-aminopyridine, methyl blue, or ODQ. Under Ca2+-free conditions, T4MN did not alter contractions evoked by caffeine, but significantly reduced, in an ODQ-preventable manner, those induced by either PHE or extracellular Ca2+ restoration following depletion of intracellular Ca2+ stores in thapsigargin-treated aortic preparations. Under the same conditions, T4MN also reduced contractions induced by protein kinase C activator phorbol-12,13-dibutyrate with a potency similar to that evoked by this nitroderivative against PHE-induced contractions. In conclusion, T4MN induces potent vasorelaxation in rat aorta by stimulating the sGC-cGMP pathway through a NO-independent mechanism. Introduction of a methoxy group into the aromatic moiety apparently stabilizes NPe, thereby enhancing its interaction with sGC.
Subject(s)
Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Guanylate Cyclase/chemistry , Guanylate Cyclase/metabolism , Styrenes/pharmacology , Vasodilation/drug effects , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/metabolism , Calcium Channels/metabolism , Calcium Signaling/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Phenylephrine/pharmacology , Potassium Channels/metabolism , Potassium Chloride/pharmacology , Rats , Rats, Wistar , Solubility , Styrenes/chemical synthesis , Vasoconstriction/drug effectsABSTRACT
Mechanisms underlying the vasorelaxant effects of trans-4-methyl-ß-nitrostyrene (T4MeN) were studied in rat aortic rings. In endothelium-intact preparations, T4MeN fully and similarly relaxed contractions induced by phenylephrine (PHE) (IC50 = 61.41 [35.40-87.42] µmol/L) and KCl (IC50 = 83.50 [56.63-110.50] µmol/L). The vasorelaxant effect of T4MeN was unchanged by endothelium removal, pretreatment with L-NAME, indomethacin, tetraethylammonium, ODQ or MDL-12,330A. Under Ca2+ -free conditions, T4MeN significantly reduced with a similar potency: (i) phasic contractions induced by PHE, but not by caffeine; (ii) contractions due to CaCl2 in aortic preparations stimulated with PHE (in the presence of verapamil) or high KCl; (iii) contractions evoked by the restoration of external Ca2+ levels after depletion of intracellular Ca2+ stores in the presence of thapsigargin. In contrast, T4MeN was more potent at inhibiting contractions evoked by the tyrosine phosphatase inhibitor, sodium orthovanadate, than those induced by the activator of PKC, phorbol-12,13-dibutyrate. These results suggest that T4MeN induces an endothelium- independent vasorelaxation that appears to occur intracellularly through the inhibition of contractions that are independent of Ca2+ influx from the extracellular milieu but involve phosphorylation of tyrosine residues.
Subject(s)
Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Styrenes/pharmacology , Vasodilation/drug effects , Vasodilator Agents/chemical synthesis , Vasodilator Agents/pharmacology , Animals , Calcium Signaling/drug effects , Chemistry Techniques, Synthetic , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Potassium Chloride/pharmacology , Rats , Styrenes/chemistry , Vanadates/pharmacology , Vasodilator Agents/chemistryABSTRACT
BACKGROUND: Bergenin, a compound derived from gallic acid, is a secondary metabolite of the plant Peltophorum dubium (Spreng.) Taub. OBJECTIVE: In this study, we aimed to characterize the ability of bergenin to eliminate the radicals in non-biological systems. METHODS: We evaluated bergenin's ability to protect erythrocytes from oxidative damage in a biological system. We have elucidated bergenin structure using nuclear magnetic resonance, X-ray diffraction, Fourier transform infrared spectroscopy, and differential scanning calorimetry. We then evaluated its antioxidant capacity in vitro against DPPHâ¢, ABTSâ¢+, hydroxyl radicals, and nitric oxide, and determined its ability to transfer electrons owing to its reduction potential and ability to chelate iron. We also evaluated its protective capacity against oxidative damage produced by AAPH in erythrocytes, its hemolytic properties, its ability to inhibit hemolysis, and its ability to maintain intracellular reduced glutathione homeostasis. RESULTS: Bergenin concentrations between 0.1 and 3mM significantly (p < 0.05) and dose dependently decreased formation of ABTSâ¢+, DPPHâ¢, nitrite ions, OHâ¢, reduced formation ferricyanide, ferrozine-Fe2+complex, inhibited AAPH-induced oxidative hemolysis of erythrocytes, raised GSH levels in the presence of AAPH, inhibited AAPH-induced lipid peroxidation in erythrocytes. CONCLUSION: Bergenin may represent a novel alternative antioxidant, with potential applications in various industries, including drugs, cosmetics, and foods.
Subject(s)
Antioxidants/isolation & purification , Antioxidants/pharmacology , Benzopyrans/isolation & purification , Benzopyrans/pharmacology , Erythrocytes/drug effects , Fabaceae/chemistry , Animals , Antioxidants/chemistry , Benzopyrans/chemistry , Benzothiazoles/chemistry , Biphenyl Compounds/chemistry , Electron Transport/drug effects , Erythrocytes/metabolism , Female , Glutathione/metabolism , Hemolysis/drug effects , Homeostasis/drug effects , Hydroxyl Radical/chemistry , Intracellular Space/drug effects , Intracellular Space/metabolism , Iron/chemistry , Lipid Peroxidation/drug effects , Models, Molecular , Molecular Conformation , Nitrites/chemistry , Picrates/chemistry , Rats , Rats, Wistar , Sulfonic Acids/chemistryABSTRACT
Mammalian sperm require to spend a limited period of time in the female reproductive tract to become competent to fertilize in a process called capacitation. It is well established that HCO3- is essential for capacitation because it activates the atypical soluble adenylate cyclase ADCY10 leading to cAMP production, and promotes alkalinization of cytoplasm, and membrane hyperpolarization. However, how HCO3- is transported into the sperm is not well understood. There is evidence that CFTR activity is involved in the human sperm capacitation but how this channel is integrated in the complex signaling cascades associated with this process remains largely unknown. In the present work, we have analyzed the extent to which CFTR regulates different events in human sperm capacitation. We observed that inhibition of CFTR affects HCO3- -entrance dependent events resulting in lower PKA activity. CFTR inhibition also affected cAMP/PKA-downstream events such as the increase in tyrosine phosphorylation, hyperactivated motility, and acrosome reaction. In addition, we demonstrated for the first time, that CFTR and PKA activity are essential for the regulation of intracellular pH, and membrane potential in human sperm. Addition of permeable cAMP partially recovered all the PKA-dependent events altered in the presence of inh-172 which is consistent with a role of CFTR upstream of PKA activation. J. Cell. Physiol. 232: 1404-1414, 2017. © 2016 Wiley Periodicals, Inc.
Subject(s)
Alkalies/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Membrane Potentials , Sperm Capacitation , Acrosome Reaction/drug effects , Benzoates/metabolism , Cell Movement/drug effects , Chlorides/metabolism , Cyclic AMP/agonists , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Humans , Hydrogen-Ion Concentration , Intracellular Space/drug effects , Intracellular Space/metabolism , Isoquinolines/pharmacology , Membrane Potentials/drug effects , Models, Biological , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Sperm Capacitation/drug effects , Sulfonamides/pharmacology , Thiazolidines/metabolismABSTRACT
BACKGROUND: Heavy metals can cause great harm to Siberian tigers in the natural environment. Cadmium (Cd2+) is an environmental contaminant that affects multiple cellular processes, including cell proliferation, differentiation, and survival. It has been shown to induce apoptosis in a variety of cell types and tissues. RESULTS: We investigated the apoptotic effects of Cd2+ on Siberian tiger fibroblasts in vitro. Our research revealed the typical signs of apoptosis after Cd2+ exposure. Apoptosis was dose- (0-4.8 µM) and duration-dependent (12-48 h), and proliferation was strongly inhibited. Cd2+ increased the activity of caspase-3, -8, and -9 and disrupted calcium homeostasis by causing oxidative stress and mitochondrial dysfunction. It also increased K+ efflux and altered the mRNA levels of Bax, Bcl-2, caspase-3, caspase-8, Fas, and p53. CONCLUSIONS: Our results suggest that Cd2+ triggers the apoptosis of Siberian tiger fibroblasts by disturbing intracellular homeostasis. These results will aid in our understanding of the effects of Cd2+ on Siberian tigers and in developing interventions to treat and prevent cadmium poisoning.
Subject(s)
Apoptosis/drug effects , Cadmium/toxicity , Fibroblasts/drug effects , Homeostasis/drug effects , Intracellular Space/drug effects , Tigers , Animals , Apoptosis/genetics , Caspases/analysis , Caspases/drug effects , Cell Cycle/drug effects , Cells, Cultured , Comet Assay/veterinary , DNA Damage , Fibroblasts/physiology , Homeostasis/physiology , Membrane Potential, Mitochondrial/drug effects , Microscopy, Electron, Transmission , Polymerase Chain Reaction , Reactive Oxygen Species/analysis , Reverse Transcription , SiberiaABSTRACT
Comparable intracellular concentrations (≈30pmol/10(6) cells) of bovine serum albumin-ZnPc (BSA) adduct outperformed dipalmitoyl-phosphatidyl-choline (DPPC) liposomes containing ZnPc at photodynamic-killing of human cervical cancer cells (HeLa) after only 15min of irradiation using red light (λ>620nm, 30W/cm(2)). This result could not be simply explained in terms of dye aggregation within the carrier, since in the liposomes the dye was considerably less aggregated than in bovine serum albumin, formulation that was capable to induce cell apoptosis upon red light exposure. Thus, using specific organelle staining, our cumulative data points towards intrinsic differences in intra-cellular localization depending on the cargo vehicle used, being ZnPc:BSA preferentially located in the near vicinity of the nucleus and in the Golgi structures, while the liposomal formulation ZnPc:DPPC was preferentially located in cellular membrane and cytoplasm. In addition to those differences, using real-time advanced fluorescence lifetime imaging of HeLa cells loaded with the photosensitizer contained in the different vehicles, we have found that only for the ZnPc:BSA formulation, there was no significant changes in the fluorescence lifetime of the photosensitizer inside the cells. This contrasts with the in situ≈two-fold reduction of the fluorescence lifetime measured for the liposomal ZnPc formulation. Those observations point towards the superiority of the protein to preserve dye aggregation, and its photochemical activity, post-cell uptake, demonstrating the pivotal role of the delivery vehicle at determining the ultimate fate of a photosensitizer.
Subject(s)
1,2-Dipalmitoylphosphatidylcholine/chemistry , Indoles/administration & dosage , Indoles/pharmacology , Organometallic Compounds/administration & dosage , Organometallic Compounds/pharmacology , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacology , Serum Albumin, Bovine/chemistry , Animals , Biological Transport , Cattle , HeLa Cells , Humans , Indoles/chemistry , Indoles/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Intracellular Space/radiation effects , Isoindoles , Liposomes , Organometallic Compounds/chemistry , Organometallic Compounds/metabolism , Photosensitizing Agents/chemistry , Photosensitizing Agents/metabolism , Zinc CompoundsABSTRACT
We investigated the mechanisms underlying the vascular effects of succinate. Vascular reactivity experiments were performed in aortic rings isolated from male Wistar rats and C57BL/6 wild type (WT) or GPR91(-/-) mice. Nitrate/nitrite (NOx) was measured colorimetrically whereas 6-keto-prostaglandin F1α (stable product of prostacyclin) was measured by enzyme immunoassay (EIA). Phosphorylation of endothelial nitric oxide synthase (eNOS) was assessed by western immunoblotting. Functional assays revealed that the direct effect of succinate in the vasculature is biphasic. At lower concentrations succinate induced relaxation while at higher concentrations succinate induced vascular contraction. Succinate concentration dependently relaxed rat aortic rings with intact endothelium. Endothelial removal reduced, but not abolished succinate-induced relaxation. Similarly, succinate relaxed endothelium-intact and endothelium-denuded aortas isolated from both C57BL/6 and GPR91(-/-) mice. Pre-incubation of endothelium-intact, but not endothelium-denuded rat aortic rings with l-NAME, indomethacin and tetraethylammonium (TEA) reduced succinate-induced relaxation. In endothelium-intact rings, succinate-induced relaxation was attenuated by ODQ, haemoglobin, Rp-8-Br-Pet-cGMPS, thapsigargin, wortmannin and SC-560. Blockade of K(+) channels with 4-aminopyridine, apamin and charybdotoxin reduced succinate-induced relaxation. Succinate increased the concentration of NOx and 6-keto-prostaglandin F1α as well as eNOS phosphorylation at ser(1177) residue. CaCl2-induced contraction of endothelium-intact or endothelium-denuded aortas was not affected by succinate. The major finding of our study is that it first demonstrates a direct effect of succinate in the vasculature. Succinate displays a biphasic and concentration-dependent effect. The vascular relaxation induced by succinate is partially mediated by endothelial GPR91 receptors via the NO-cGMP pathway, a vasodilator cyclooxygenase (COX) product(s) and the opening of K(+) channels.
Subject(s)
Aorta, Thoracic/drug effects , Succinic Acid/pharmacology , Vasodilator Agents/pharmacology , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiology , Calcium/metabolism , Calcium Chloride/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Mice , Nitrogen Oxides/metabolism , Phenylephrine/pharmacology , Rats , Rats, Wistar , Vasoconstriction/drug effectsABSTRACT
Leishmaniasis is a neglected tropical disease (NTDs), endemic in 88 countries that affect more than 12 million people. Current drugs are limited due to their toxicity, development of biological resistance, length of treatment and high cost. Thus, the search for new effective and less toxic treatments is an urgent need. In this study, we report the synthesis of 3 new amide derivatives of hederagenin (22-24) with yields between 70% and 90%, along with 57 other derivatives of hederagenin (1-21, 25-60) carrying different groups at C-28 previously reported by our group, and the results of their in vitro ability to inhibit the growth of Leishmania infantum. Some derivatives (3, 4, 44, 49 and 52), showed activity at micromolar level and low toxicity against BGM and HepG2 cells. Moreover, the ability of hederagenin derivatives 3 (IC50 = 9.7 µM), 4 (12 µM), 44 (11 µM) and 49 (2 µM), to prevent proliferation of intracellular amastigote forms of L. infantum and their higher selectivity index and low toxicity compared to commercial positive drug control of choice (potassium antimonyl tartrate trihydrate) (IC50 = 80 µM, SI = 0.1), make these compounds promising candidates for the treatment of leishmaniasis.
Subject(s)
Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacology , Leishmania infantum/drug effects , Oleanolic Acid/analogs & derivatives , Saponaria/chemistry , Antiprotozoal Agents/toxicity , Hep G2 Cells , Humans , Intracellular Space/drug effects , Intracellular Space/parasitology , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , Oleanolic Acid/toxicity , Structure-Activity RelationshipABSTRACT
Thiazolidinediones (TZDs) are peroxisome proliferator-activated receptor-γ (PPARγ) agonists used in treating type 2 diabetes that may exhibit beneficial pleiotropic effects on endothelial cells. In this study, we characterized the effects of three new TZDs [GQ-32 (3-biphenyl-4-ylmethyl-5-(4-nitro-benzylidene)-thiazolidine-2,4-dione), GQ-169 (5-(4-chloro-benzylidene)-3-(2,6-dichloro-benzyl)-thiazolidine-2,4-dione), and LYSO-7 (5-(5-bromo-1H-indol-3-ylmethylene)-3-(4-chlorobenzyl)-thiazolidine-2,4-dione)] on endothelial cells. The effects of the new TZDs were evaluated on the production of nitric oxide (NO) and reactive oxygen species (ROS), cell migration, tube formation and the gene expression of adhesion molecules and angiogenic mediators in human umbilical vein endothelial cells (HUVECs). PPARγ activation by new TZDs was addressed with a reporter gene assay. The three new TZDs activated PPARγ and suppressed the tumor necrosis factor α-induced expression of vascular cell adhesion molecule 1 and intercellular adhesion molecule 1. GQ-169 and LYSO-7 also inhibited the glucose-induced ROS production. Although NO production assessed with 4-amino-5-methylamino-2',7'-difluorofluorescein-FM probe indicated that all tested TZDs enhanced intracellular levels of NO, only LYSO-7 treatment significantly increased the release of NO from HUVEC measured by chemiluminescence analysis of culture media. Additionally, GQ-32 and GQ-169 induced endothelial cell migration and tube formation by the up-regulation of angiogenic molecules expression, such as vascular endothelial growth factor A and interleukin 8. GQ-169 also increased the mRNA levels of basic fibroblast growth factor, and GQ-32 enhanced transforming growth factor-ß expression. Together, the results of this study reveal that these new TZDs act as partial agonists of PPARγ and modulate endothelial cell activation and endothelial dysfunction besides to stimulate migration and tube formation.
Subject(s)
Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Thiazolidinediones/pharmacology , Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Nitric Oxide/metabolism , PPAR gamma/agonists , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacologyABSTRACT
Activated microglia that produce reactive nitrogen species (RNS), inflammatory factors, reactive oxygen species (ROS), and other neurovirulent factors may lead to the development of neurodegenerative diseases. Certain compounds can inhibit the activation of microglia. However, these mechanisms remain unclear. In the present study, we investigated the inhibitory effect of Gamma-decanolactone (GD) on the production of reactive oxygen species and inducible nitric oxide synthase (iNOS) in lipopolysaccharide (LPS) - stimulated N9 murine microglial cells through the p38 MAPK signaling pathway. The results showed that GD attenuated the activation of N9 cells and inhibited intracellular reactive oxygen species and the expression of iNOS and TNF-α induced by LPS in the cells. In addition, GD blocked the phosphorylation of p38 and inhibited cleaved caspase-9 and DNA damage. These data indicate that GD has therapeutic potential for the treatment of neurodegenerative diseases, and that it exerts its effects by inhibiting inflammation.