ABSTRACT
The development of new bioactive molecules based on the molecular hybridization has been widely explored. In line with this, reliable tests should be employed to give information about the toxicology of these new molecules. In this sense, the use of in vitro tests is a valuable tool, especially the in vitro maturation of oocytes (IVM), which is an efficient resource to discover the potential toxicity of synthetic molecules. Thus, the aim of the present study was to evaluate the toxicological effects of the selenium-containing indolyl compound 3-(4-Chlorophenylselanyl)-1-methyl-1H-indole (CMI), on different quality parameters of bovine oocytes through the IVM. Different concentrations of the CMI compound (0, 25, 50, 100, 200⯵M) were supplemented during the in vitro maturation process. After, the oocyte maturation rate, glutathione (GSH) levels, reactive oxygen species (ROS) levels, membrane, and mitochondrial integrity were evaluated. The results showed that the lowest concentration of CMI induced the highest GSH production (Pâ¯<â¯0.05), an important marker of cytoplasmic quality and maturation. All treatments increased ROS production in relation to non-supplementation (Pâ¯<â¯0.05). In addition, oocyte maturation was reduced only with the highest concentration of CMI (Pâ¯<â¯0.05). Supplementation with CMI did not impact mitochondrial activity, integrity and cell membrane. To our knowledge, this is the first study that evaluates CMI on the oocyte in vitro maturation process. Importantly, our results did not find any toxic effect of CMI on bovine oocytes. CMI was efficient for cytoplasmic maturation by promoting an increase in the intracellular levels of glutathione.
Subject(s)
In Vitro Oocyte Maturation Techniques , Indoles/toxicity , Oocytes/drug effects , Selenium Compounds/toxicity , Animals , Cattle , Cell Survival/drug effects , Cells, Cultured , Female , Glutathione/metabolism , Oocytes/metabolism , Reactive Oxygen Species/metabolismABSTRACT
The discovery of aggregation inhibitors and the elucidation of their mechanism of action are key in the quest to mitigate the toxic consequences of amyloid formation. We have previously characterized the antiamyloidogenic mechanism of action of sodium phtalocyanine tetrasulfonate ([Na4(H2PcTS)]) on α-Synuclein (αS), demonstrating that specific aromatic interactions are fundamental for the inhibition of amyloid assembly. Here we studied the influence that metal preferential affinity and peripheral substituents may have on the activity of tetrapyrrolic compounds on αS aggregation. For the first time, our laboratory has extended the studies in the field of the bioinorganic chemistry and biophysics to cellular biology, using a well-established cell-based model to study αS aggregation. The interaction scenario described in our work revealed that both N- and C-terminal regions of αS represent binding interfaces for the studied compounds, a behavior that is mainly driven by the presence of negatively or positively charged substituents located at the periphery of the macrocycle. Binding modes of the tetrapyrrole ligands to αS are determined by the planarity and hydrophobicity of the aromatic ring system in the tetrapyrrolic molecule and/or the preferential affinity of the metal ion conjugated at the center of the macrocyclic ring. The different capability of phthalocyanines and meso-tetra (N-methyl-4-pyridyl) porphine tetrachloride ([H2PrTPCl4]) to modulate αS aggregation in vitro was reproduced in cell-based models of αS aggregation, demonstrating unequivocally that the modulation exerted by these compounds on amyloid assembly is a direct consequence of their interaction with the target protein.
Subject(s)
Amyloidogenic Proteins/metabolism , Indoles/metabolism , Porphyrins/metabolism , Protein Multimerization/drug effects , Zinc/metabolism , alpha-Synuclein/metabolism , Amino Acid Sequence , Amyloidogenic Proteins/chemistry , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Indoles/chemistry , Indoles/toxicity , Porphyrins/chemistry , Porphyrins/toxicity , Protein Binding , Zinc/chemistry , alpha-Synuclein/chemistryABSTRACT
The diamondback moth Plutella xylostella (Linnaeus, 1758) (Lepidoptera: Plutellidae) is a destructive pest of brassica crops of economic importance that have resistance to a range of insecticides. Indole derivates can exert diverse biological activities, and different effects may be obtained from small differences in their molecular structures. Indole is the parent substance of a large number of synthetic and natural compounds, such as plant and animal hormones. In the present study, we evaluate the insecticidal activity of 20 new synthesized indole derivatives against P. xylostella, and the selectivity of these derivatives against non-target hymenopteran beneficial arthropods: the pollinator Apis mellifera (Linnaeus, 1758) (Hymenoptera: Apidae), and the predators Polybia scutellaris (White, 1841), Polybia sericea (Olivier, 1791) and Polybia rejecta (Fabricius, 1798) (Hymenoptera: Vespidae). Bioassays were performed in the laboratory to determine the lethal and sublethal effects of the compounds on P. xylostella and to examine their selectivity to non-target organisms by topical application and foliar contact. The treatments consisted of two synthesized derivatives (most and least toxic), the positive control (deltamethrin) and the negative control (solvent). The synthesized compound 4e [1-(1H-indol-3-yl)hexan-1-one] showed high toxicity (via topical application and ingestion) and decreased the leaf consumption by P. xylostella, displaying a higher efficiency than the pyrethroid deltamethrin, widely used to control this pest. In addition, the synthesized indole derivatives were selective to the pollinator A. mellifera and the predators P. scutellaris, P. sericea and P. rejecta, none of which were affected by deltamethrin. Our results highlight the promising potential of the synthesized indole derivatives for the generation of new chemical compounds for P. xylostella management.
Subject(s)
Bees/drug effects , Indoles/toxicity , Insecticides/toxicity , Moths/drug effects , Wasps/drug effects , Animals , Indoles/pharmacology , Insecticides/pharmacology , Larva/drug effectsABSTRACT
Glioblastoma multiforme is considered to be one of the most aggressive types of tumors of the central nervous system, with a poor prognosis and short survival periods of ~ one year. The current protocol for glioblastoma treatment includes the surgical excision of the primary tumor followed by radio and chemotherapy. Photodynamic therapy (PDT) is considered a promising strategy for the treatment of several types of tumors. Phthalocyanines (Pcs) are good photosensitizers (PSs) for PDT because they induce cell death in several cellular models. ZnPc (Zn(II)phthalocyanine) is a well-known Pc, extensively tested in different cells and tumor models, but its evaluation on a glioblastoma model has been poorly studied. Herein, we compare the capacity of ZnPc and one of its derivatives, Zn(II)tetraminephthalocyanine (TAZnPc), to photoinactivate glioblastoma cells (T98G, MO59, LN229 and U87-MG) in culture. We measured the cellular uptake, the toxicity in the dark and the subcellular localization of the different Pcs, as well as the clonogenic capacity of surviving cells after PDT. The mechanism of cell death induced after PDT was determined by measuring caspase 3 activation, DNA fragmentation, phosphatidylserine externalization, mitochondrial morphological changes and loss of mitochondrial membrane potential as well as lysosomal membrane integrity. Overall, ZnPc and TAZnPc present good properties to be used as PSs with photoinactivation capacity on glioblastoma cells.
Subject(s)
Glioblastoma/drug therapy , Indoles/toxicity , Organometallic Compounds/toxicity , Photochemotherapy/methods , Photosensitizing Agents/toxicity , Caspase 3/metabolism , Cell Line, Tumor , DNA Fragmentation , Humans , Indoles/chemistry , Indoles/pharmacology , Isoindoles , Lysosomes/drug effects , Mitochondria/drug effects , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Zinc CompoundsABSTRACT
Two Streptomyces spp. strains responsible for potato common scab infections in Uruguay which do not produce diketopiperazines were identified through whole-genome sequencing, and the virulence factor produced by one of them was isolated and characterized. Phylogenetic analysis showed that both pathogenic strains can be identified as S. niveiscabiei, and the structure of the phytotoxin was elucidated as that of the polyketide desmethylmensacarcin using MS and NMR methods. The metabolite is produced in yields of â¼200 mg/L of culture media, induces deep necrotic lesions on potato tubers, stuns root and shoot growth in radish seedlings, and is comparatively more aggressive than thaxtomin A. This is the first time that desmethylmensacarcin, a member of a class of compounds known for their antitumor and antibiotic activity, is associated with phytotoxicity. More importantly, it represents the discovery of a new virulence factor related to potato common scab, an economically-important disease affecting potato production worldwide.
Subject(s)
Plant Diseases/microbiology , Solanum tuberosum/microbiology , Streptomyces/chemistry , Diketopiperazines , Indoles/toxicity , Molecular Structure , Phylogeny , Piperazines/toxicity , Plant Diseases/etiology , Raphanus/microbiology , Streptomyces/pathogenicity , Virulence Factors/chemistry , Virulence Factors/isolation & purificationABSTRACT
Thioredoxin plays an essential role in bacterial antioxidant machinery and virulence; however, its regulatory actions in the host are less well understood. Reduced human Trx activates transient receptor potential canonical 5 (TRPC5) in inflammation, but there is no evidence of whether these receptors mediate bacterial thioredoxin effects in the host. Importantly, TRPC5 can form functional complexes with other subunits such as TRPC4. Herein, E. coli-derived thioredoxin induced mortality in lipopolysaccharide- (LPS-) injected mice, accompanied by reduction of leukocyte accumulation, regulation of cytokine release into the peritoneum, and impairment of peritoneal macrophage-mediated phagocytosis. Dual TRPC4/TRPC5 blockade by ML204 increased mortality and hypothermia in thioredoxin-treated LPS mice but preserved macrophage's ability to phagocytose. TRPC5 deletion did not alter body temperature but promoted additional accumulation of peritoneal leukocytes and inflammatory mediator release in thioredoxin-administered LPS mice. Thioredoxin diminished macrophage-mediated phagocytosis in wild-type but not TRPC5 knockout animals. TRPC5 ablation did not affect LPS-induced responses. However, ML204 caused mortality associated with exacerbated hypothermia and decreased peritoneal leukocyte numbers and cytokines in LPS-injected mice. These results suggest that bacterial thioredoxin effects under LPS stimuli are mediated by TRPC4 and TRPC5, shedding light on the additional mechanisms of bacterial virulence and on the pathophysiological roles of these receptors.
Subject(s)
Escherichia coli/chemistry , Lipopolysaccharides/toxicity , Systemic Inflammatory Response Syndrome/metabolism , TRPC Cation Channels/metabolism , Thioredoxins/therapeutic use , Animals , Hydrogen Peroxide/metabolism , Indoles/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Phagocytosis/drug effects , Piperidines/toxicity , Systemic Inflammatory Response Syndrome/chemically induced , TRPC Cation Channels/antagonists & inhibitors , Virulence/drug effectsABSTRACT
Treatment of leishmaniasis involves the use of antimonials, miltefosine, amphotericin B or pentamidine. However, the side effects of these drugs and the reports of drug-resistant parasites demonstrate the need for new treatments that are safer and more efficacious. Histone deacetylase inhibitors are a new class of compounds with potential to treat leishmaniasis. Herein, we evaluated the effects of KH-TFMDI, a novel histone deacetylase inhibitor, on Leishmania amazonensis promastigotes and intracellular amastigotes. The IC50 values of this compound for promastigotes and intracellular amastigotes were 1.976 and 1.148 µM, respectively, after 72 h of treatment. Microscopic analyses revealed that promastigotes became elongated and thinner in response to KH-TFMDI, indicating changes in cytoskeleton organization. Immunofluorescence microscopy, western blotting and flow cytometry using an anti-acetylated tubulin antibody revealed an increase in the expression of acetylated tubulin. Furthermore, transmission electron microscopy revealed several ultrastructural changes, such as (a) mitochondrial swelling, followed by the formation of many vesicles inside the matrix; (b) presence of lipid bodies randomly distributed through the cytoplasm; (c) abnormal chromatin condensation; and (d) formation of blebs on the plasma membrane. Physiological studies for mitochondrial function, flow cytometry with propidium iodide and TUNEL assay confirmed the alterations in the mitochondrial metabolism, cell cycle, and DNA fragmentation, respectively, which could result to cell death by mechanisms related to apoptosis-like. All these together indicate that histone deacetylases are promising targets for the development of new drugs to treat Leishmania, and KH-TFMDI is a promising drug candidate that should be tested in vivo.
Subject(s)
Benzylidene Compounds/pharmacology , Cell Death/drug effects , Cytoskeleton/drug effects , Histone Deacetylase Inhibitors/pharmacology , Indoles/pharmacology , Leishmania/drug effects , Mitochondria/drug effects , Sirtuins/antagonists & inhibitors , Animals , Antiparasitic Agents/pharmacology , Antiparasitic Agents/toxicity , Apoptosis/drug effects , Benzylidene Compounds/toxicity , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Cytoskeleton/metabolism , Histone Deacetylase Inhibitors/toxicity , Indoles/toxicity , Inhibitory Concentration 50 , Leishmania/cytology , Leishmania/growth & development , Leishmania/ultrastructure , Leishmaniasis/drug therapy , Leishmaniasis/parasitology , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Microtubules/drug effects , Microtubules/metabolism , Oxidative Stress/drug effectsABSTRACT
Photodynamic therapy is emerging as a hopeful method for the treatment of oncological diseases. In the search of novel therapeutic strategies for colorectal cancer, in this work we reported the photocytotoxic activity of a lipophilic zinc(II) phthalocyanine on a murine colon adenocarcinoma cell line (CT26 cells). The 2,9(10),16(17),23(24) tetrakis[(2-dimethylamino)ethylsulfanyl]phthalocyaninatozinc(II), named Pc9, was encapsulated into Tetronic® 1107 polymeric poloxamine micelles (T1107) and assayed in 2D and 3D cell cultures. We showed that the formulation Pc9-T1107 was efficient to reduce cell viability after photodynamic treatment both in 2D cultures (IC50 10±2nM) as well as in CT26 spheroids (IC50 370±11nM). Cellular uptake of Pc9-T1107 was a time- and concentration-dependent process, being the phthalocyanine formulation mainly incorporated into lysosomal vesicles and endoplasmic reticulum cisterns, but not in mitochondria. Pc9-T1107 also induced the formation of reactive oxygen species immediately after cell irradiation. We also found that the phototoxic action of Pc9-T1107 was partially reversed in the presence of antioxidants, such as TROLOX and N-acetyl-cysteine. In addition, we showed that Pc9-T1107 treatment triggered an apoptotic cell death, as suggested by the detection of pyknotic nuclei, the reduction in the expression levels of procaspase-3 and the increase in caspase-3 enzymatic activity.
Subject(s)
Apoptosis/drug effects , Drug Carriers/chemistry , Indoles/chemistry , Indoles/toxicity , Micelles , Organometallic Compounds/toxicity , Photosensitizing Agents/toxicity , Antioxidants/chemistry , Antioxidants/metabolism , Apoptosis/radiation effects , Caspase 3/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Endoplasmic Reticulum/metabolism , Humans , Isoindoles , Light , Lysosomes/metabolism , Organometallic Compounds/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Zinc CompoundsSubject(s)
Benzhydryl Compounds/toxicity , Gene Expression Regulation, Developmental/drug effects , Genistein/toxicity , Indoles/toxicity , Mammary Glands, Animal/drug effects , Phenols/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Animals , Anticarcinogenic Agents/toxicity , Benzhydryl Compounds/administration & dosage , Estrogens, Non-Steroidal/toxicity , Female , Genistein/administration & dosage , Indoles/administration & dosage , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/pathology , Phenols/administration & dosage , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/pathology , Random Allocation , Rats, Sprague-DawleyABSTRACT
A novel series of twenty 3-thiocyanato-1H-indoles, carrying diversification at positions N-1, C-2 and C-5 of the heterocyclic core, were synthesized; their antiproliferative activity against four human cancer cell lines (HL60, HEP-2, NCI-H292 and MCF-7) was evaluated, employing doxorubicin as positive control. Indole, N-methylindole and 2-(4-chlorophenyl)-N-methylindole demonstrated to be essentially inactive, whereas several of their congener 3-thiocyanato-1H-indoles displayed good to excellent levels of potency (IC50 ≤ 6 µM), while being non-hemolytic. N-Phenyl-3-thiocyanato-1H-indole and 1-methyl-2-(4-chlorophenyl)-3-thiocyanato-1H-indole showed good to high potency against all the cell lines. On the other side, the N-(4-chlorophenyl)-, 2-(4-chlorophenyl)- and 2-phenyl- 3-thiocyanato-1H-indole derivatives were slightly less active against the test cell lines. Overall, these results suggest that the indole-3-thiocyanate motif can be suitably decorated to afford highly cytotoxic compounds and that the substituted indole can be employed as a useful scaffold toward more potent compounds.
Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Indoles/chemical synthesis , Indoles/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Drug Screening Assays, Antitumor , Hemolysis/drug effects , Humans , Indoles/chemistry , Indoles/toxicityABSTRACT
BACKGROUND: Violacein is a purple pigment from Chromobacterium violaceum that possesses diverse biological and pharmacological properties. Among these, pro-oxidant and antioxidant activities have been suggested. However, the cytotoxic mechanisms induced by violacein are poorly understood and the improvement in knowledge regarding these cell death mechanisms will be useful to develop new therapeutic approaches. Considering this, in our work, we investigated the pro-oxidant effects of violacein in non-tumor (CHO-K1 and MRC-5) and tumor (HeLa) cell lines, searching for a better understanding of reactive oxygen species (ROS) production and cell death induction. RESULTS: Cytotoxicity induced by violacein was observed in the three cell lines; however, MRC-5 and HeLa cells were shown to be more sensitive to violacein treatment. Although punctual alterations in the antioxidant apparatus and increase in oxidative stress biomarkers was observed in some violacein concentrations, no association was found between increased oxidative stress and induction of cell death. However, the increase of mitochondrial membrane potential was observed. CONCLUSIONS: In fact, the increase of mitochondrial membrane potential in MRC-5 and HeLa cells suggests that mitochondrial membrane hyperpolarization might be the main cause of cell death triggered by violacein.
Subject(s)
Indoles/toxicity , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , CHO Cells , Cell Death , Cell Line , Cell Survival/drug effects , Cricetulus , HeLa Cells , Humans , Indoles/pharmacologyABSTRACT
BACKGROUND & AIMS: Sustained activation of the cytosolic calcium concentration induces injury to pancreatic acinar cells and necrosis. The calcium release-activated calcium modulator ORAI1 is the most abundant Ca(2+) entry channel in pancreatic acinar cells; it sustains calcium overload in mice exposed to toxins that induce pancreatitis. We investigated the roles of ORAI1 in pancreatic acinar cell injury and the development of acute pancreatitis in mice. METHODS: Mouse and human acinar cells, as well as HEK 293 cells transfected to express human ORAI1 with human stromal interaction molecule 1, were hyperstimulated or incubated with human bile acid, thapsigargin, or cyclopiazonic acid to induce calcium entry. GSK-7975A or CM_128 were added to some cells, which were analyzed by confocal and video microscopy and patch clamp recordings. Acute pancreatitis was induced in C57BL/6J mice by ductal injection of taurolithocholic acid 3-sulfate or intravenous' administration of cerulein or ethanol and palmitoleic acid. Some mice then were given GSK-7975A or CM_128, which inhibit ORAI1, at different time points to assess local and systemic effects. RESULTS: GSK-7975A and CM_128 each separately inhibited toxin-induced activation of ORAI1 and/or activation of Ca(2+) currents after Ca(2+) release, in a concentration-dependent manner, in mouse and human pancreatic acinar cells (inhibition >90% of the levels observed in control cells). The ORAI1 inhibitors also prevented activation of the necrotic cell death pathway in mouse and human pancreatic acinar cells. GSK-7975A and CM_128 each inhibited all local and systemic features of acute pancreatitis in all 3 models, in dose- and time-dependent manners. The agents were significantly more effective, in a range of parameters, when given at 1 vs 6 hours after induction of pancreatitis. CONCLUSIONS: Cytosolic calcium overload, mediated via ORAI1, contributes to the pathogenesis of acute pancreatitis. ORAI1 inhibitors might be developed for the treatment of patients with pancreatitis.
Subject(s)
Acinar Cells/drug effects , Benzamides/pharmacology , Calcium Channels/drug effects , Calcium Channels/metabolism , Calcium/metabolism , Pancreatitis/drug therapy , Pyrazoles/pharmacology , Acinar Cells/cytology , Acute Disease , Animals , Bile Acids and Salts/toxicity , Calcium/toxicity , Cells, Cultured , Disease Models, Animal , HEK293 Cells , Humans , Indoles/toxicity , Mice , Mice, Inbred C57BL , ORAI1 Protein , Pancreatitis/chemically induced , Pancreatitis/metabolism , Thapsigargin/toxicity , Time Factors , Treatment OutcomeABSTRACT
In this work, the in vitro anti-Leishmania activity of photodynamic liposomes made of soybean phosphatidylcholine, sodium cholate, total polar archaeolipids (TPAs) extracted from the hyperhalophile archaea Halorubrum tebenquichense and the photosensitizer zinc phthalocyanine (ZnPcAL) was compared to that of ultradeformable photodynamic liposomes lacking TPAs (ZnPcUDLs). We found that while ZnPcUDLs and ZnPcALs (130 nm mean diameter and -35 mV zeta potential) were innocuous against promastigotes, a low concentration (0.01 µM ZnPc and 7.6 µM phospholipids) of ZnPcALs irradiated at a very low-energy density (0.2 J/cm(2)) eliminated L. braziliensis amastigotes from J774 macrophages, without reducing the viability of the host cells. In such conditions, ZnPcALs were harmless for J774 macrophages, HaCaT keratinocytes, and bone marrow-derived dendritic cells. Therefore, topical photodynamic treatment would not likely affect skin-associated lymphoid tissue. ZnPcALs were extensively captured by macrophages, but ZnPcUDLs were not, leading to 2.5-fold increased intracellular delivery of ZnPc than with ZnPcUDLs. Despite mediating low levels of reactive oxygen species, the higher delivery of ZnPc and the multiple (caveolin- and clathrin-dependent plus phagocytic) intracellular pathway followed by ZnPc would have been the reason for the higher antiamastigote activity of ZnPcALs. The leishmanicidal activity of photodynamic liposomal ZnPc was improved by TPA-containing liposomes.
Subject(s)
Antiprotozoal Agents/pharmacology , Glyceryl Ethers/pharmacology , Indoles/pharmacology , Leishmania/drug effects , Leishmania/radiation effects , Liposomes/pharmacology , Organometallic Compounds/pharmacology , Animals , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacokinetics , Antiprotozoal Agents/toxicity , Cell Line , Cell Survival/drug effects , Glyceryl Ethers/chemistry , Glyceryl Ethers/pharmacokinetics , Glyceryl Ethers/toxicity , Humans , Indoles/chemistry , Indoles/pharmacokinetics , Indoles/toxicity , Isoindoles , Liposomes/chemistry , Liposomes/pharmacokinetics , Liposomes/toxicity , Macrophages/metabolism , Mice , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/toxicity , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Zinc CompoundsABSTRACT
In order to find a novel photosensitizer to be used in photodynamic therapy for cancer treatment, we have previously showed that the cationic zinc(II) phthalocyanine named Pc13, the sulfur-linked dye 2,9(10),16(17),23(24)-tetrakis[(2-trimethylammonium) ethylsulfanyl]phthalocyaninatozinc(II) tetraiodide, exerts a selective phototoxic effect on human nasopharynx KB carcinoma cells and induces an apoptotic response characterized by an increase in the activity of caspase-3. Since the activation of an apoptotic pathway by chemotherapeutic agents contributes to the elimination of malignant cells, in this study we investigated the molecular mechanisms underlying the antitumor action of Pc13. We found that after light exposure, Pc13 induced the production of reactive oxygen species (ROS), which are mediating the resultant cytotoxic action on KB cells. ROS led to an early permeabilization of lysosomal membranes as demonstrated by the reduction of lysosome fluorescence with acridine orange and the release of lysosomal proteases to cytosol. Treatment with antioxidants inhibited ROS generation, preserved the integrity of lysosomal membrane and increased cell proliferation in a concentration-dependent manner. Lysosome disruption was followed by mitochondrial depolarization, cytosolic release of cytochrome C and caspases activation. Although no change in the total amount of Bax was observed, the translocation of Bax from cytosol to mitochondria, the cleavage of the pro-apoptotic protein Bid, together with the decrease of the anti-apoptotic proteins Bcl-XL and Bcl-2 indicated the involvement of Bcl-2 family proteins in the induction of the mitochondrial pathway. It was also demonstrated that cathepsin D, but not caspase-8, contributed to Bid cleavage. In conclusion, Pc13-induced cell photodamage is triggered by ROS generation and activation of the mitochondrial apoptotic pathway through the release of lysosomal proteases. In addition, our results also indicated that Pc13 induced a caspase-dependent apoptotic response, being activation of caspase-8, -9 and -3 the result of a post-mitochondrial event.
Subject(s)
Dermatitis, Phototoxic/metabolism , Dermatitis, Phototoxic/pathology , Indoles/toxicity , Lysosomes/metabolism , Mitochondria/metabolism , Organometallic Compounds/toxicity , Caspases/metabolism , Cathepsins/antagonists & inhibitors , Cathepsins/metabolism , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Tumor , Cytochromes c/metabolism , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Humans , Indoles/chemistry , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Intracellular Membranes/radiation effects , Isoindoles , Lysosomes/drug effects , Lysosomes/radiation effects , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/radiation effects , Mitochondria/drug effects , Mitochondria/radiation effects , Models, Biological , Organometallic Compounds/chemistry , Permeability/drug effects , Permeability/radiation effects , Photochemotherapy , Protein Transport/drug effects , Protein Transport/radiation effects , Radiation, Ionizing , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Zinc Compounds , bcl-2-Associated X Protein/metabolismABSTRACT
A highly regio- and stereoselective method has been developed and expanded for the synthesis of a 20-membered library of spirooxindole 1'-nitro pyrrolizidines via 1,3-dipolar cycloaddition of azomethine ylides, generated in situ by a decarboxylative route from a common set of diverse isatins and L-proline derivatives, with substituted ß-nitrostyrenes under aqueous medium. Among various reaction conditions, water proved to be necessary for the interaction of the reagents as well as heating the reaction at 90 °C for one hour, during which time the desired products were obtained in good yields and with excellent regio- and stereoselectivities. We subsequently applied in silico drug discovery computational methods to (i) identify the ADME properties, based on Lipinski's rule, (ii) screen the toxicological profile, and (iii) predict the penetration through the blood brain barrier (BBB) of the synthesized compounds. Next, the LC50 values of all these spirocyclic oxindoles were determined in zebrafish embryos cultured individually in buffer solutions of each compound and, finally, the phenotypes induced by these molecules in the zebrafish embryos at concentrations below their LC50 were analyzed at 48, 72 and 96 hours post fertilization.
Subject(s)
Embryo, Nonmammalian/drug effects , Indoles/chemistry , Indoles/chemical synthesis , Pyrroles/chemistry , Water/chemistry , Zebrafish/embryology , Animals , Blood-Brain Barrier/metabolism , Chemistry Techniques, Synthetic , Computer Simulation , Indoles/metabolism , Indoles/toxicity , Oxindoles , Phenotype , Stereoisomerism , Substrate SpecificityABSTRACT
BACKGROUND: Renal cell carcinoma (RCC) is the most common type of kidney cancer, and represents the third most common urological malignancy. Despite the advent of targeted therapies for RCC and the improvement of the lifespan of patients, its cost-effectiveness restricted the therapeutic efficacy. In a recent report, we showed that synthetic phosphoethanolamine (Pho-s) has a broad antitumor activity on a variety of tumor cells and showed potent inhibitor effects on tumor progress in vivo. METHODOLOGY/PRINCIPAL FINDINGS: We show that murine renal carcinoma (Renca) is more sensitive to Pho-s when compared to normal immortalized rat proximal tubule cells (IRPTC) and human umbilical vein endothelial cells (HUVEC). In vitro anti-angiogenic activity assays show that Pho-s inhibits endothelial cell proliferation, migration and tube formation. In addition, Pho-s has anti-proliferative effects on HUVEC by inducing a cell cycle arrest at the G2/M phase. It causes a decrease in cyclin D1 mRNA, VEGFR1 gene transcription and VEGFR1 receptor expression. Pho-s also induces nuclear fragmentation and affects the organization of the cytoskeleton through the disruption of actin filaments. Additionally, Pho-s induces apoptosis through the mitochondrial pathway. The putative therapeutic potential of Pho-s was validated in a renal carcinoma model, on which our remarkable in vivo results show that Pho-s potentially inhibits lung metastasis in nude mice, with a superior efficacy when compared to Sunitinib. CONCLUSIONS/SIGNIFICANCE: Taken together, our findings provide evidence that Pho-s is a compound that potently inhibits lung metastasis, suggesting that it is a promising novel candidate drug for future developments.
Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Ethanolamines/pharmacology , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/toxicity , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Caspase 3/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclin D1/genetics , Cytoskeleton/drug effects , Ethanolamines/chemical synthesis , Ethanolamines/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Indoles/pharmacology , Indoles/toxicity , Kidney Tubules, Proximal/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasm Metastasis/drug therapy , Neovascularization, Pathologic/drug therapy , Oxidation-Reduction/drug effects , Pyrroles/pharmacology , Pyrroles/toxicity , Rats , Sunitinib , Transcription, Genetic , Vascular Endothelial Growth Factor Receptor-1/genetics , Wound Healing/drug effectsABSTRACT
New assays with HepG2 cells indicate that Indigo Carmine (IC), a dye that is widely used as additive in many food and pharmaceutical industries exhibited cytotoxic effects. This work describes the development of a bicomponent nanostructured Ti/TiO2/WO3 electrode prepared by "template" method and investigates its efficiency in a photoelectrocatalytic method by using visible light irradiation and applied potential of 1V. After 2h of treatment there are reduction of 97% discoloration, 62% of mineralization and formation of three byproducts assigned as: 2-amine-5-sulfo-benzoic acid, 2,3-dioxo-14-indole-5-sulfonic acid, and 2-amino-α-oxo-5-sulfo-benzeneacetic acid were identified by HPLC-MS/MS. But, cytotoxicity was completely removed after 120 min of treatment.
Subject(s)
Electrodes , Indoles/analysis , Nanostructures/chemistry , Water Pollutants, Chemical/analysis , Water Purification/methods , Indigo Carmine , Indoles/chemistry , Indoles/toxicity , Models, Chemical , Oxides/chemistry , Photochemical Processes , Titanium/chemistry , Tungsten/chemistry , Water Pollutants, Chemical/chemistry , Water Pollutants, Chemical/toxicityABSTRACT
PURPOSE: To evaluate the in vivo and in vitro toxicity of sunitinib malate, a multikinase inhibitor molecule. DESIGN: Experimental, Prospective, Controlled. METHODS: Human retinal pigment epithelial (ARPE-19) and human umbilical vein endothelialcells (HUVECS) were used in a culture toxicity test and exposed to different concentrations of sunitinib malate for 18 hours. The HUVECs also were cultured to evaluate the angiogenesis inhibitory effect of sunitinib malate. Fundus photography and angiographic, electrophysiologic, and histopathologic evaluations with light and electron microscopy were performed in two groups of five rabbits each that received different intravitreal concentrations of the drug. Each rabbit received 0.1 ml of sunitinib malate in the right eye (one group with 12.5 mg/ml, the other group with 25 mg/ml); all animals received 0.1 ml of physiologic saline solution in the left eye. After sacrifice, the eyes were enucleated and fixed with modified Karnovsky solution. RESULTS: No toxicity related to sunitinib malate was observed using an in vitro model with the 12.5 and 25 mg/ml solutions in HUVEC and ARPE cell cultures. No toxicity was observed in the in vivo model with 12.5 mg/ml, but light microscopy showed that the 25 mg/ml solution damaged the photoreceptors layer. No functional changes in the electroretinogram were observed in any group. CONCLUSIONS: Sunitinib malate 12.5 mg/ml caused no toxicity in in vivo and in vitro models, but the 25 mg/ml concentration caused retinal changes suggesting toxicity in the in vivo model. Further research with the drug is needed in models of ocular neovascularization.
Subject(s)
Angiogenesis Inhibitors/toxicity , Antineoplastic Agents/toxicity , Endothelium, Vascular/drug effects , Indoles/toxicity , Photoreceptor Cells, Vertebrate/drug effects , Pyrroles/toxicity , Retinal Pigment Epithelium/drug effects , Animals , Cell Count , Cell Line , Dose-Response Relationship, Drug , Electroretinography/drug effects , Fluorescein Angiography , Humans , Intravitreal Injections , Photoreceptor Cells, Vertebrate/ultrastructure , Protein-Tyrosine Kinases/antagonists & inhibitors , Rabbits , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/ultrastructure , Sunitinib , Umbilical Veins/cytologyABSTRACT
Gastrotoxicity is a major problem for long-term therapy with non-steroidal anti-inflammatory drugs (NSAIDs). DICCIC (1-(2,6-dichlorophenyl)indolin-2-one) is a new diclofenac prodrug, which has proven anti-inflammatory activity without gastroulcerogenic effect. The aim of this work was to compare the pharmacokinetic profiles of diclofenac from DICCIC (7.6 mg/kg equivalent to 8.1 mg/kg diclofenac) and diclofenac (8.1 mg/kg) administration in Wistar rats weighing 250-300 g (n=20). The doses were calculated by interspecific allometric scaling based on the 2 mg/kg from diary human dose of diclofenac. Blood samples were collected in heparinized tubes via the femoral artery through the implanted catheter. The plasma was separated and quantitation was made in a HPLC system with a UV-Vis detector. The confidence limits of the bioanalytical method were appropriate for its application in a preclinical pharmacokinetic study. The AUC of diclofenac from DICCIC (53.7± 5.8 ug/mL.min) was significantly less (Mann Whitney test, p<0.05) than that of diclofenac from diclofenac administration (885.9 ± 124,8 ug/mL.min). Terminal half-life of diclofenac from DICCIC (50.1 ± 17.2 min) was significantly less (Mann Whitney test, p<0.05) than that of diclofenac from diclofenac administration (247.4 ± 100.9 min). Still the parameters clearance and distribution volume were calculated for diclofenac from diclofenac, whose results were 9.2 ±1.2 mL/min.kg and 3.3 ±1.2 L/kg, respectively. The results of DICCIC from DICCIC administration were 108.9 ± 19.6 mL/min.kg and 7.8 ± 2.4 L/kg for clearance and distribution volume, respectively. The pharmacokinetic profile demonstrated that there was an increase in diclofenac elimination and a lower exposure to diclofenac with administration of DICCIC compared to diclofenac.
Subject(s)
Indoles/pharmacokinetics , Prodrugs/pharmacokinetics , Animals , Diclofenac/pharmacokinetics , Diclofenac/toxicity , Indoles/toxicity , Male , Prodrugs/toxicity , Rats , Rats, Wistar , Stomach Ulcer/chemically inducedABSTRACT
The compounds 1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)methyl nitrate (C1), (1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)ethyl nitrate (C2), 3-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)benzyl nitrate (C3), 4-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)-N-hydroxy-benzenesulfonamide (C4), 4-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)benzyl nitrate (C5), and 2-[4-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)phenyl]ethyl nitrate (C6) were evaluated with a micronucleus test using mouse peripheral blood to identify new candidate drugs for the treatment of sickle cell disease (SCD) that are safer than hydroxyurea. The compounds induced an average frequency of micronucleated reticulocytes (MNRET) of less than six per 1,000 cells at 12.5, 25, 50, and 100 mg/kg, whereas hydroxyurea induced an average MNRET frequency of 7.8, 9.8, 15, and 33.7 per 1000 cells respectively, at the same concentrations. Compounds C1-C6 are new non-genotoxic in vivo candidate drugs for the treatment of SCD symptoms.