Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 218: 115927, 2023 12.
Article in English | MEDLINE | ID: mdl-37992998

ABSTRACT

The search for effective antimalarial agents remains a critical priority because malaria is widely spread and drug-resistant strains are becoming more prevalent. In this review, a variety of small molecules capable of modulating redox processes were showcased for their potential as antimalarial agents. The compounds were designed to target the redox balance of Plasmodium parasites, which has a pivotal function in their ability to survive and multiply within the host organism. A thorough screening method was utilized to assess the effectiveness of these compounds against both drug-sensitive and drug-resistant strains of Plasmodium falciparum, the malaria-causing parasite. The results revealed that several of the tested compounds exhibited significant effectiveness against malaria, displaying IC50 values at a low micromolar range. Furthermore, these compounds displayed promising selectivity for the parasite, as they exhibited low cytotoxicity towards mammalian cells. Thorough mechanistic studies were undertaken to clarify how the active compounds exert their mode of action. The findings revealed that these compounds disrupted the parasites' redox balance, causing oxidative stress and interfering with essential cellular functions. Additionally, the compounds showed synergistic effects when combined with existing antimalarial drugs, suggesting their potential for combination therapies to combat drug resistance. Overall, this study highlights the potential of redox-modulating small molecules as effective antimalarial agents. The identified compounds demonstrate promising antimalarial activity, and their mechanism of action offers insights into targeting the redox balance of Plasmodium parasites. Further optimization and preclinical studies are warranted to determine their efficacy, safety, and potential for clinical development as novel antimalarial therapeutics.


Subject(s)
Antimalarials , Malaria , Animals , Humans , Antimalarials/pharmacology , Antimalarials/therapeutic use , Malaria/drug therapy , Malaria/parasitology , Plasmodium falciparum , Oxidation-Reduction , Oxidative Stress , Mammals
2.
Free Radic Biol Med ; 209(Pt 2): 211-227, 2023 11 20.
Article in English | MEDLINE | ID: mdl-37898387

ABSTRACT

The pursuit of effective anticancer therapies has led to a burgeoning interest in the realm of redox modulation. This review provides a comprehensive exploration of the intricate mechanisms by which diverse anticancer molecules leverage redox pathways for therapeutic intervention. Redox modulation, encompassing the fine balance of oxidation-reduction processes within cells, has emerged as a pivotal player in cancer treatment. This review delves into the multifaceted mechanisms of action employed by various anticancer compounds, including small molecules and natural products, to disrupt cancer cell proliferation and survival. Beginning with an examination of the role of redox signaling in cancer development and resistance, the review highlights how aberrant redox dynamics can fuel tumorigenesis. It then meticulously dissects the strategies employed by anticancer agents to induce oxidative stress, perturb redox equilibrium, and trigger apoptosis within cancer cells. Furthermore, the review explores the challenges and potential side effects associated with redox-based treatments, along with the development of novel redox-targeted agents. In summary, this review offers a profound understanding of the dynamic interplay between redox modulation and anticancer molecules, presenting promising avenues to revolutionize cancer therapy and enhance patient outcomes.


Subject(s)
Apoptosis , Biological Products , Humans , Oxidation-Reduction , Carcinogenesis , Cell Proliferation
8.
Free Radic Biol Med ; 65: 456-467, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23892052

ABSTRACT

Nonsteroidal anti-inflammatory drug (NSAID)-induced mitochondrial oxidative stress (MOS) is an important prostaglandin (PG)-independent pathway of the induction of gastric mucosal injury. However, the molecular mechanism behind MOS-mediated gastric pathology is still obscure. In various pathological conditions of tissue injury oxidative stress is often linked with inflammation. Here we report that MOS induced by indomethacin (an NSAID) induces gastric mucosal inflammation leading to proinflammatory damage. Indomethacin, time dependently stimulated the expression of proinflammatory molecules such as intercellular adhesion molecule 1(ICAM-1), vascular cell adhesion molecule 1(VCAM-1), interleukin1ß (IL-1ß), and monocyte chemotactic protein-1 (MCP-1) in gastric mucosa in parallel with the increase of neutrophil infiltration and injury of gastric mucosa in rat. Western immunoblotting and confocal microscopic studies revealed that indomethacin induced nuclear translocation of nuclear factor kappa-B (NF-κB) in gastric mucosal cells, which resulted in proinflammatory signaling. The prevention of MOS by antioxidant tryptamine-gallic acid hybrid (SEGA) inhibited indomethacin-induced expression of ICAM-1, VCAM-1, IL-1ß, and MCP-1. SEGA also prevented indomethacin-induced NF-κB activation and neutrophil infiltration as documented by chromatin immunoprecipitation studies and neutrophil migration assay, respectively. Heme oxygenase-1 (HO-1), a cytoprotective enzyme associated with tissue repair mechanisms is stimulated in response to oxidative stress. We have investigated the role of HO-1 against MOS and MOS-mediated inflammation in recovering from gastropathy. Indomethacin stimulated the expression of HO-1 and indomethacin-stimulated HO-1 expression was reduced by SEGA, an antioxidant, which could prevent MOS. Thus, the data suggested that the induction of HO-1 was a protective response against MOS developed by indomethacin. Moreover, the induction of HO-1 by cobalt protoporphyrin inhibited inflammation and chemical silencing of HO-1 by zinc protoporphyrin aggravated the inflammation by indomethacin. Thus, NSAID by promoting MOS-induced proinflammatory response damaged gastric mucosa and HO-1 protected NSAID-induced gastric mucosal damage by preventing NF-κB activation and proinflammatory activity.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Heme Oxygenase-1/metabolism , NF-kappa B/metabolism , Animals , Blotting, Western , Chromatin Immunoprecipitation , Gastric Mucosa/injuries , Indomethacin/adverse effects , Inflammation/chemically induced , Microscopy, Confocal , Mitochondria/drug effects , Mitochondria/metabolism , Neutrophil Infiltration/drug effects , Oxidative Stress , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley
9.
J Biol Chem ; 287(32): 26630-46, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22696214

ABSTRACT

We have investigated the impact of persistent intravascular hemolysis on liver dysfunction using the mouse malaria model. Intravascular hemolysis showed a positive correlation with liver damage along with the increased accumulation of free heme and reactive oxidants in liver. Hepatocytes overinduced heme oxygenase-1 (HO-1) to catabolize free heme in building up defense against this pro-oxidant milieu. However, in a condition of persistent free heme overload in malaria, the overactivity of HO-1 resulted in continuous transient generation of free iron to favor production of reactive oxidants as evident from 2',7'-dichlorofluorescein fluorescence studies. Electrophoretic mobility shift assay documented the activation of NF-κB, which in turn up-regulated intercellular adhesion molecule 1 as evident from chromatin immunoprecipitation studies. NF-κB activation also induced vascular cell adhesion molecule 1, keratinocyte chemoattractant, and macrophage inflammatory protein 2, which favored neutrophil extravasation and adhesion in liver. The infiltration of neutrophils correlated positively with the severity of hemolysis, and neutrophil depletion significantly prevented liver damage. The data further documented the elevation of serum TNFα in infected mice, and the treatment of anti-TNFα antibodies also significantly prevented neutrophil infiltration and liver injury. Deferoxamine, which chelates iron, interacts with free heme and bears antioxidant properties that prevented oxidative stress, NF-κB activation, neutrophil infiltration, hepatocyte apoptosis, and liver damage. Furthermore, the administration of N-acetylcysteine also prevented NF-κB activation, neutrophil infiltration, hepatocyte apoptosis, and liver damage. Thus, hepatic free heme accumulation, TNFα release, oxidative stress, and NF-κB activation established a link to favor neutrophil infiltration in inducing liver damage during hemolytic conditions in malaria.


Subject(s)
Heme/metabolism , Hemolysis , Liver/physiopathology , Malaria/physiopathology , NF-kappa B/metabolism , Neutrophil Infiltration , Animals , Base Sequence , Blotting, Western , DNA Primers , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Heme Oxygenase (Decyclizing)/metabolism , In Situ Nick-End Labeling , Liver/metabolism , Malaria/parasitology , Male , Mice , Mice, Inbred BALB C , Oxidative Stress , Plasmodium yoelii/isolation & purification , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
10.
J Biol Chem ; 287(29): 24844-61, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22645149

ABSTRACT

Macrophage migration inhibitory factor (MIF) is responsible for proinflammatory reactions in various infectious and non-infectious diseases. We have investigated the mechanism of anti-inflammatory activity of epoxyazadiradione, a limonoid purified from neem (Azadirachta indica) fruits, against MIF. Epoxyazadiradione inhibited the tautomerase activity of MIF of both human (huMIF) and malaria parasites (Plasmodium falciparum (PfMIF) and Plasmodium yoelii (PyMIF)) non-competitively in a reversible fashion (K(i), 2.11-5.23 µm). Epoxyazadiradione also significantly inhibited MIF (huMIF, PyMIF, and PfMIF)-mediated proinflammatory activities in RAW 264.7 cells. It prevented MIF-induced macrophage chemotactic migration, NF-κB translocation to the nucleus, up-regulation of inducible nitric-oxide synthase, and nitric oxide production in RAW 264.7 cells. Epoxyazadiradione not only exhibited anti-inflammatory activity in vitro but also in vivo. We tested the anti-inflammatory activity of epoxyazadiradione in vivo after co-administering LPS and MIF in mice to mimic the disease state of sepsis or bacterial infection. Epoxyazadiradione prevented the release of proinflammatory cytokines such as IL-1α, IL-1ß, IL-6, and TNF-α when LPS and PyMIF were co-administered to BALB/c mice. The molecular basis of interaction of epoxyazadiradione with MIFs was explored with the help of computational chemistry tools and a biological knowledgebase. Docking simulation indicated that the binding was highly specific and allosteric in nature. The well known MIF inhibitor (S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid methyl ester (ISO-1) inhibited huMIF but not MIF of parasitic origin. In contrast, epoxyazadiradione inhibited both huMIF and plasmodial MIF, thus bearing an immense therapeutic potential against proinflammatory reactions induced by MIF of both malaria parasites and human.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Limonins/pharmacology , Macrophage Migration-Inhibitory Factors/pharmacology , Macrophages/drug effects , Animals , Azadirachta/chemistry , Cell Line , Electrophoretic Mobility Shift Assay , Humans , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Nitric Oxide/metabolism , Plasmodium falciparum/metabolism , Plasmodium yoelii/metabolism
11.
Free Radic Biol Med ; 53(1): 129-42, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22588006

ABSTRACT

We have synthesized a new series of aryl aryl methyl thio arenes (AAMTAs) and evaluated antimalarial activity in vitro and in vivo against drug-resistant malaria. These compounds interact with free heme, inhibit hemozoin formation, and prevent Plasmodium falciparum growth in vitro in a concentration-dependent manner. These compounds concentration dependently promote oxidative stress in Plasmodium falciparum as evident from the generation of intraparasitic oxidants, protein carbonyls, and lipid peroxidation products. Furthermore, AAMTAs deplete intraparasite GSH levels, which is essential for antioxidant defense and survival during intraerythrocytic stages. These compounds displayed potent antimalarial activity not only in vitro but also in vivo against multidrug-resistant Plasmodium yoelii dose dependently in a mouse model. The mixtures of enantiomers of AAMTAs containing 3-pyridyl rings were found to be more efficient in providing antimalarial activity. Efforts have been made to synthesize achiral AAMTAs 17-23 and among them, compound 18 showed significant antimalarial activity in vivo.


Subject(s)
Antimalarials/therapeutic use , Drug Resistance, Multiple/drug effects , Hydrocarbons, Aromatic/chemistry , Malaria/prevention & control , Oxidative Stress/drug effects , Plasmodium falciparum/drug effects , Animals , Glutathione/metabolism , Heme/metabolism , Malaria/parasitology , Male , Mice , Mice, Inbred BALB C
12.
J Biol Chem ; 287(5): 3495-509, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22157011

ABSTRACT

We have investigated the gastroprotective effect of SEGA (3a), a newly synthesized tryptamine-gallic acid hybrid molecule against non-steroidal anti-inflammatory drug (NSAID)-induced gastropathy with mechanistic details. SEGA (3a) prevents indomethacin (NSAID)-induced mitochondrial oxidative stress (MOS) and dysfunctions in gastric mucosal cells, which play a pathogenic role in inducing gastropathy. SEGA (3a) offers this mitoprotective effect by scavenging of mitochondrial superoxide anion (O(2)(·-)) and intramitochondrial free iron released as a result of MOS. SEGA (3a) in vivo blocks indomethacin-mediated MOS, as is evident from the inhibition of indomethacin-induced mitochondrial protein carbonyl formation, lipid peroxidation, and thiol depletion. SEGA (3a) corrects indomethacin-mediated mitochondrial dysfunction in vivo by restoring defective electron transport chain function, collapse of transmembrane potential, and loss of dehydrogenase activity. SEGA (3a) not only corrects mitochondrial dysfunction but also inhibits the activation of the mitochondrial pathway of apoptosis by indomethacin. SEGA (3a) inhibits indomethacin-induced down-regulation of bcl-2 and up-regulation of bax genes in gastric mucosa. SEGA (3a) also inhibits indometacin-induced activation of caspase-9 and caspase-3 in gastric mucosa. Besides the gastroprotective effect against NSAID, SEGA (3a) also expedites the healing of already damaged gastric mucosa. Radiolabeled ((99m)Tc-labeled SEGA (3a)) tracer studies confirm that SEGA (3a) enters into mitochondria of gastric mucosal cell in vivo, and it is quite stable in serum. Thus, SEGA (3a) bears an immense potential to be a novel gastroprotective agent against NSAID-induced gastropathy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Apoptosis/drug effects , Gallic Acid/pharmacology , Indomethacin/adverse effects , Mitochondria/metabolism , Stomach Diseases/chemically induced , Stomach Diseases/drug therapy , Tryptamines/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Electron Transport Chain Complex Proteins/metabolism , Gastric Mucosa/metabolism , Gene Expression Regulation/drug effects , Humans , Indomethacin/pharmacology , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Superoxides/metabolism , bcl-2-Associated X Protein/biosynthesis
13.
Antioxid Redox Signal ; 17(4): 555-82, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22122517

ABSTRACT

SIGNIFICANCE: Parasitic diseases affect hundreds of millions of people worldwide and represent major health problems. Treatment is becoming extremely difficult due to the emergence of drug resistance, the absence of effective vaccines, and the spread of insecticide-resistant vectors. Thus, identification of affordable and readily available drugs against resistant parasites is of global demand. RECENT ADVANCES: Susceptibility of many parasites to oxidative stress is a well-known phenomenon. Therefore, generation of reactive oxygen species (ROS) or inhibition of endogenous antioxidant enzymes would be a novel therapeutic approach to develop antiparasitic drugs. This article highlights the unique metabolic pathways along with redox enzymes of unicellular (Plasmodium falciparum, Trypanosoma cruzi, Trypanosoma brucei, Leishmania donovani, Entamoeba histolytica, and Trichomonas vaginalis) and multicellular parasites (Schistosoma mansoni), which could be utilized to promote ROS-mediated toxicity. CRITICAL ISSUES: Enzymes involved in various vital redox reactions could be potential targets for drug development. FUTURE DIRECTIONS: The identification of redox-active antiparasitic drugs along with their mode of action will help researchers around the world in designing novel drugs in the future.


Subject(s)
Antiparasitic Agents/pharmacology , Animals , Entamoeba histolytica/drug effects , Entamoeba histolytica/enzymology , Entamoeba histolytica/metabolism , Humans , Oxidation-Reduction , Oxidative Stress , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Plasmodium falciparum/metabolism , Reactive Oxygen Species/metabolism , Trichomonas vaginalis/drug effects , Trichomonas vaginalis/enzymology , Trichomonas vaginalis/metabolism , Trypanosomatina/drug effects , Trypanosomatina/enzymology , Trypanosomatina/metabolism
14.
Nucleic Acids Res ; 40(3): 1174-90, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22006844

ABSTRACT

We have investigated the DNA-binding nature as well as the function of a putative Alba (Acetylation lowers binding affinity) family protein (PfAlba3) from Plasmodium falciparum. PfAlba3 possesses DNA-binding property like Alba family proteins. PfAlba3 binds to DNA sequence non-specifically at the minor groove and acetylation lowers its DNA-binding affinity. The protein is ubiquitously expressed in all the erythrocytic stages of P. falciparum and it exists predominantly in the acetylated form. PfAlba3 inhibits transcription in vitro by binding to DNA. Plasmodium falciparum Sir2 (PfSir2A), a nuclear localized deacetylase interacts with PfAlba3 and deacetylates the lysine residue of N-terminal peptide of PfAlba3 specific for DNA binding. PfAlba3 is localized with PfSir2A in the periphery of the nucleus. Fluorescence in situ hybridization studies revealed the presence of PfAlba3 in the telomeric and subtelomeric regions. ChIP and ChIP ReChIP analyses further confirmed that PfAlba3 binds to the telomeric and subtelomeric regions as well as to var gene promoter.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Plasmodium falciparum , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Amino Acid Sequence , Chromatin Immunoprecipitation , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/analysis , Lysine/metabolism , Models, Molecular , Molecular Sequence Data , Peptides/metabolism , Protein Binding , Protozoan Proteins/analysis , RNA-Binding Proteins/chemistry , Sirtuin 2/metabolism , Transcription, Genetic
15.
Bioorg Med Chem ; 19(24): 7365-73, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22088307

ABSTRACT

Macrophage migration inhibitory factor (MIF), a pro-inflammatory cytokine, is involved in the development of an array of inflammatory disorders including rheumatoid arthritis, inflammatory bowel disease, psoriasis, multiple sclerosis and sepsis. The synthesis of MIF-inhibitor is a rationale approach to develop novel anti-inflammatory agent to treat multitude of inflammatory diseases. In this work, we have synthesized and evaluated MIF-inhibitory activity of a series of small molecules containing isoxazoline skeleton. Mode of binding of this inhibitor to human MIF (huMIF) was determined by docking studies. The synthesized molecules inhibit tautomerase activity of huMIF. The anti-inflammatory activity of the most active inhibitor, 4-((3-(4-hydroxy-3-methoxyphenyl)-4, 5-dihydroisoxazol-5-yl) methoxy) benzaldehyde (4b) was evaluated against huMIF-induced inflammation in a cellular model (RAW 264.7 cell). Compound 4b significantly inhibits huMIF-mediated NF-κB translocation to the nucleus, up-regulation of inducible nitric oxide synthase and nitric oxide production in RAW 264.7 cell which are the markers for inflammation. The compound 4b is not cytotoxic as evident from cell viability assay. Hence, the compound 4b has potential to be a novel anti-inflammatory agent.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Isoxazoles/chemistry , Isoxazoles/pharmacology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Animals , Anti-Inflammatory Agents/chemical synthesis , Binding Sites , Cell Line , Cell Survival/drug effects , Humans , Isoxazoles/chemical synthesis , Macrophage Migration-Inhibitory Factors/chemistry , Macrophage Migration-Inhibitory Factors/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Models, Molecular , NF-kappa B/metabolism , Nitric Oxide/immunology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Up-Regulation
16.
J Biol Chem ; 286(45): 39387-402, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21908612

ABSTRACT

The mechanism of action of heme oxygenase-1 (HO-1) in mitochondrial oxidative stress (MOS)-mediated apoptotic tissue injury was investigated. MOS-mediated gastric mucosal apoptosis and injury were introduced in rat by indomethacin, a non-steroidal anti-inflammatory drug. Here, we report that HO-1 was not only induced but also translocated to mitochondria during gastric mucosal injury to favor repair mechanisms. Furthermore, mitochondrial translocation of HO-1 resulted in the prevention of MOS and mitochondrial pathology as evident from the restoration of the complex I-driven mitochondrial respiratory control ratio and transmembrane potential. Mitochondrial translocation of HO-1 also resulted in time-dependent inhibition of apoptosis. We searched for the plausible mechanisms responsible for HO-1 induction and mitochondrial localization. Free heme, the substrate for HO-1, was increased inside mitochondria during gastric injury, and mitochondrial entry of HO-1 decreased intramitochondrial free heme content, suggesting that a purpose of mitochondrial translocation of HO-1 is to detoxify accumulated heme. Heme may activate nuclear translocation of NF-E2-related factor 2 to induce HO-1 through reactive oxygen species generation. Electrophoretic mobility shift assay and chromatin immunoprecipitation studies indicated nuclear translocation of NF-E2-related factor 2 and its binding to HO-1 promoter to induce HO-1 expression during gastric injury. Inhibition of HO-1 by zinc protoporphyrin aggravated the mucosal injury and delayed healing. Zinc protoporphyrin further reduced the respiratory control ratio and transmembrane potential and enhanced MOS and apoptosis. In contrast, induction of HO-1 by cobalt protoporphyrin reduced MOS, corrected mitochondrial dysfunctions, and prevented apoptosis and gastric injury. Thus, induction and mitochondrial localization of HO-1 are a novel cytoprotective mechanism against MOS-mediated apoptotic tissue injury.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Apoptosis/drug effects , Gastric Mucosa/enzymology , Gastric Mucosa/injuries , Heme Oxygenase (Decyclizing)/metabolism , Indomethacin/adverse effects , Mitochondria/enzymology , Oxidative Stress/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Indomethacin/pharmacology , Membrane Potential, Mitochondrial/drug effects , NF-E2 Transcription Factor/metabolism , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley
17.
Bioorg Med Chem Lett ; 21(12): 3563-7, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21600767

ABSTRACT

Heme is an important prosthetic molecule for various hemoproteins and serves important function in living aerobic organisms. But degradation of hemoprotein, for example, hemoglobin during different pathological conditions leads to the release of heme, which is very toxic as it induces oxidative stress and inflammation due to its pro-oxidant nature. Thus, synthesis of compound that will detoxify free heme by interacting with it would be fruitful for the management of heme-induced pathogenesis. Here, we report the synthesis of a novel natural product arborinine and some other acridone derivatives, which interact with free heme. These acridones in vitro block heme-mediated protein oxidation and degradation, markers for heme-induced oxidative stress.


Subject(s)
Acridines/chemical synthesis , Hemeproteins/metabolism , Acridines/chemistry , Acridines/pharmacology , Acridones , Hemeproteins/antagonists & inhibitors , Hemeproteins/chemistry , Inhibitory Concentration 50 , Models, Biological , Molecular Structure , Oxidation-Reduction/drug effects , Oxidative Stress , Protein Binding
18.
Free Radic Biol Med ; 50(11): 1659-68, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21406224

ABSTRACT

Plasmodium falciparum macrophage migration inhibitory factor (PfMIF) exhibits thioredoxin (Trx)-like oxidoreductase activity but the active site for this activity and its function have not been evaluated. A bioinformatics search revealed that the conserved CXXC motif, which is responsible for Trx-like oxidoreductase activity, is absent from PfMIF. In contrast, the adjacent N-terminal Cys-3 and Cys-4 are conserved in MIF across species of malarial parasites. Mutation of either vicinal Cys-3 or Cys-4 of PfMIF abolished the Trx-like activity, whereas the mutation of the remaining Cys-59 or Cys-103 did not affect it. PfMIF has an antioxidant function. It prevents reactive oxygen species-mediated lipid peroxidation and oxidative damage of DNA as evident from DNA nicking assay. Interestingly, chemical modification of the vicinal cysteines by phenylarsine oxide (PAO), a specific vicinal thiol modifier, significantly prevented this antioxidant activity. Modification of Cys-3 and Cys-4 was confirmed by MALDI-TOF mass spectroscopy of peptide fragments obtained after cyanogen bromide digestion of PAO-modified PfMIF. Furthermore, mutation of either Cys-3 or Cys-4 of PfMIF resulted in the loss of both Trx-like oxidoreductase and antioxidant activities of PfMIF. Altogether, our results suggest that the vicinal Cys-3 and Cys-4 play a critical role in the Trx-like oxidoreductase activity and antioxidant property of PfMIF.


Subject(s)
Cysteine/metabolism , Erythrocytes/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Malaria/metabolism , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , Animals , Arsenicals/pharmacology , Computational Biology , Cysteine/chemistry , Cysteine/genetics , DNA Damage/drug effects , Enzyme Activation/drug effects , Erythrocytes/parasitology , Erythrocytes/pathology , Macrophage Migration-Inhibitory Factors/chemistry , Malaria/genetics , Malaria/parasitology , Mutagenesis, Site-Directed , Mutation/genetics , Oxidative Stress/drug effects , Oxidoreductases/metabolism , Plasmodium falciparum/pathogenicity , Protozoan Proteins/chemistry , Rabbits , Stereoisomerism , Thioredoxins/chemistry , Thioredoxins/metabolism
19.
Free Radic Biol Med ; 49(2): 258-67, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20406680

ABSTRACT

Nonsteroidal anti-inflammatory drug (NSAID)-induced oxidative stress plays a critical role in gastric mucosal cell apoptosis and gastropathy. NSAIDs induce the generation of hydroxyl radical ((*)OH) through the release of free iron, which plays an important role in developing gastropathy. Thus, molecules having both iron-chelating and antiapoptotic properties will be beneficial in preventing NSAID-induced gastropathy. Gallic acid (GA), a polyphenolic natural product, has the capacity to chelate free iron. Here, we report that GA significantly prevents, as well as heals, NSAID-induced gastropathy. In vivo, GA blocks NSAID-mediated mitochondrial oxidative stress by preventing mitochondrial protein carbonyl formation, lipid peroxidation, and thiol depletion. In vitro, GA scavenges free radicals and blocks (*)OH-mediated oxidative damage. GA also attenuates gastric mucosal cell apoptosis in vivo as well as in vitro in cultured gastric mucosal cells as evident from the TUNEL assay. GA prevents NSAID-induced activation of caspase-9, a marker for the mitochondrial pathway of apoptosis, and restores NSAID-mediated collapse of the mitochondrial transmembrane potential and dehydrogenase activity. Thus, the inhibition of mitochondrial oxidative stress by GA is associated with the inhibition of NSAID-induced mitochondrial dysfunction and activation of apoptosis in gastric mucosal cells, which are responsible for gastric injury or gastropathy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Apoptosis , Gallic Acid/pharmacology , Iron Chelating Agents/pharmacology , Mitochondria/drug effects , Stomach Diseases/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Apoptosis/drug effects , Caspase 9/genetics , Caspase 9/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Gallic Acid/administration & dosage , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Iron Chelating Agents/administration & dosage , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Protein Carbonylation/drug effects , Rats , Rats, Sprague-Dawley , Stomach Diseases/chemically induced , Stomach Diseases/pathology , Stomach Diseases/physiopathology
20.
J Pineal Res ; 46(3): 314-23, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19220725

ABSTRACT

Augmentation of gastric mucosal cell apoptosis due to development of oxidative stress is one of the main pathogenic events in the development of nonsteroidal anti-inflammatory drug (NSAID)-induced gastropathy. Identification of a nontoxic, anti-apoptotic molecule is warranted for therapy against NSAID-induced gastropathy. The objective of the present study was to define the mechanism of the anti-apoptotic effect of melatonin, a nontoxic molecule which scavenges reactive oxygen species. Using an array of experimental approaches, we have shown that melatonin prevents the development of mitochondrial oxidative stress and activation of mitochondrial pathway of apoptosis induced by indomethacin (a NSAID) in the gastric mucosa. Melatonin inhibits the important steps of indomethacin-induced activation of mitochondrial pathway of apoptosis such as upregulation of the expression of Bax and Bak, and the downregulation of Bcl-2 and BclxL. Melatonin also prevents indomethacin-induced mitochondrial translocation of Bax and prevents the collapse of mitochondrial membrane potential. Moreover, melatonin reduces indomethacin-mediated activation of caspase-9 and caspase-3 by blocking the release of cytochrome c and finally rescues gastric mucosal cells from indomethacin-induced apoptosis as measured by the TUNEL assay. Histologic studies of gastric mucosa further document that melatonin almost completely protects against gastric damage induced by indomethacin. Thus, melatonin has significant anti-apoptotic effects to protect gastric mucosa from NSAID-induced apoptosis and gastropathy, which makes its use as potential therapy against gastric damage during NSAID treatment.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Gastric Mucosa/drug effects , Indomethacin/pharmacology , Melatonin/pharmacology , Mitochondria/metabolism , Oxidative Stress/drug effects , Analysis of Variance , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cyclooxygenase Inhibitors/pharmacology , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , In Situ Nick-End Labeling , Membrane Potential, Mitochondrial/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...