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










Publication year range
1.
Dalton Trans ; 52(16): 5176-5191, 2023 Apr 25.
Article in English | MEDLINE | ID: mdl-36970749

ABSTRACT

Nitrosyl ruthenium complexes are promising platforms for nitric oxide (NO) and nitroxyl (HNO) release, which exert their therapeutic application. In this context, we developed two polypyridinic compounds with the general formula cis-[Ru(NO)(bpy)2(L)]n+, where L is an imidazole derivative. These species were characterized by spectroscopic and electrochemical techniques, including XANES/EXAFS experiments, and further supported by DFT calculations. Interestingly, assays using selective probes evidenced that both complexes can release HNO on reaction with thiols. This finding was biologically validated by HIF-1α detection. The latter protein is related to angiogenesis and inflammation processes under hypoxic conditions, which is selectively destabilized by nitroxyl. These metal complexes also presented vasodilating properties using isolated rat aorta rings and demonstrated antioxidant properties in free radical scavenging experiments. Based on these results, the new nitrosyl ruthenium compounds showed promising characteristics as potential therapeutic agents for the treatment of cardiovascular conditions such as atherosclerosis, deserving further investigation.


Subject(s)
Coordination Complexes , Ruthenium , Animals , Rats , Nitric Oxide/chemistry , Nitrogen Oxides/chemistry , Ruthenium/chemistry , Sulfhydryl Compounds/chemistry , Cardiovascular Diseases
2.
Mol Pharm ; 20(1): 6-22, 2023 01 02.
Article in English | MEDLINE | ID: mdl-36350781

ABSTRACT

For more than 70 years, sodium nitroprusside (SNP) has been used to treat severe hypertension in hospital emergency settings. During this time, a few other clinical uses have also emerged such as in the treatment of acute heart failure as well as improving mitral incompetence and in the intra- and perioperative management during heart surgery. This drug functions by releasing nitric oxide (NO), which modulates several biological processes with many potential therapeutic applications. However, this small molecule has a short lifetime, and it has been administered through the use of NO donor molecules such as SNP. On the other hand, SNP also has some setbacks such as the release of cyanide ions, high water solubility, and very fast NO release kinetics. Currently, there are many drug delivery strategies that can be applied to overcome many of these limitations, providing novel opportunities for the use of old drugs, including SNP. This Perspective describes some nitroprusside properties and highlights new potential therapeutic uses arising from the use of drug delivery systems, mainly silica-based nanoparticles. There is a series of great opportunities to further explore SNP in many medical issues as reviewed, which deserves a closer look by the scientific community.


Subject(s)
Nanoparticles , Nitric Oxide Donors , Nitroprusside , Nitric Oxide Donors/therapeutic use , Nitric Oxide , Drug Delivery Systems
3.
Eur J Pharmacol ; 921: 174869, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35247379

ABSTRACT

Metal coordination complexes are chemotherapeutic and anti-inflammatory agents. The ruthenium complex FOR811A ([Ru(bpy)2(2-MIM)Cl](PF6)3) FOR811A was evaluated in mice models of acute inflammation and behavioral tests. Animals received FOR811A (3, 10 or 30 mg/kg; i.p.), indomethacin (20 mg/kg; i.p.), L-NAME (20 mg/kg; i.v.) aminoguanidine (50 mg/kg; i.p.) or dexamethasone (0.5 mg/kg; s.c.) 30 min before inflammatory stimulation. Paw edema was induced by carrageenan (400 µg/paw), TNF-α or L-arginine (15 nmol/paw) (5 ng/paw) and evaluated by hydropletismometry 4 h later. Peritonitis was induced by carrageenan (500 µg; i.p.) and evaluated 4 h later for hypernociception and quantification of total/differential leukocytes, total protein reduced glutathione (GSH) and myeloperoxidase (MPO). FOR811A inhibited the paw edema induced by carrageenan at 3 (64%; p < 0.0001), 10 (73%; p < 0.0001) and 30 mg/kg (66%; p < 0.0001), and at 10 mg/kg that induced with L-arginine by 75% or TNF-α by 55% (p = 0.0012). Paw tissues histological analysis showed reduction in mast cells (46%; p = 0.0027), leukocyte infiltrate (66%; p < 0.0001), edema and hemorrhagic areas. Immunohistochemical evaluation revealed inhibition of iNOS (62%; p < 0.0001) and TNF-α (35%; p < 0.0001). In the peritonitis model FOR811A increased (2.8X; p < 0.0001) hypernociceptive threshold, reduced total leukocytes (29%; p < 0.0001), neutrophils (47%; p = 0.0003) and total proteins (36%; p = 0.0082). FOR811A also inhibited MPO (47%; p = 0.0296) and increased GSH (1.8X; p < 0.0001). In the behavioral tests, FOR811A reduced (30.6%) the number of crossings in the open field, and increased (16%) the number of falls in the Rota rod. Concluding, FOR811A presents anti-inflammatory and antioxidant effects, via nitric oxide pathway.


Subject(s)
Nitric Oxide , Organometallic Compounds , 2,2'-Dipyridyl/analogs & derivatives , Animals , Anti-Inflammatory Agents/adverse effects , Carrageenan/adverse effects , Edema/chemically induced , Edema/drug therapy , Edema/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Mice , Nitric Oxide/metabolism , Organometallic Compounds/pharmacology , Organometallic Compounds/therapeutic use
4.
J Inorg Biochem ; 228: 111666, 2022 03.
Article in English | MEDLINE | ID: mdl-34923187

ABSTRACT

This study aimed to investigate the synthesis and potential vasodilator effect of a novel ruthenium complex, cis-[Ru(bpy)2(2-MIM)(NO2)]PF6 (bpy = 2,2'-bipyridine and 2-MIM = 2-methylimidazole) (FOR711A), containing an imidazole derivative via an in silico molecular docking model using ß1 H-NOX (Heme-nitric oxide/oxygen binding) domain proteins of reduced and oxidized soluble guanylate cyclase (sGC). In addition, pharmacokinetic properties in the human organism were predicted through computational simulations and the potential for acute irritation of FOR711A was also investigated in vitro using the hen's egg chorioallantoic membrane (HET-CAM). FOR711A interacted with sites of the ß1 H-NOX domain of reduced and oxidized sGC, demonstrating shorter bond distances to several residues and negative values of total energy. The predictive study revealed molar refractivity (RM): 127.65; Log Po/w = 1.29; topological polar surface area (TPSA): 86.26 Å2; molar mass (MM) = 541.55 g/mol; low solubility, high unsaturation index, high gastrointestinal absorption; toxicity class 4; failure to cross the blood-brain barrier and to react with cytochrome P450 (CYP) enzymes CYP1A2, CYP2C19, CYP2C9, CYP2D6 and CYP3A4. After the HET-CAM assay, the FOR711A complex was classified as non-irritant (N.I.) and its vasodilator effect was confirmed through greater evidence of blood vessels after the administration and ending of the observation period of 5 min. These results suggest that FOR711A presented a potential stimulator/activator effect of sGC via NO/sGC/cGMP. However, results indicate it needs a vehicle for oral administration.


Subject(s)
Coordination Complexes/chemistry , Nitric Oxide/chemistry , Ruthenium/chemistry , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacology , Animals , Chickens , Chorioallantoic Membrane/metabolism , Heme/chemistry , Humans , Imidazoles/chemistry , Molecular Docking Simulation/methods , Nitric Oxide/metabolism , Oxygen/chemistry , Protein Domains , Soluble Guanylyl Cyclase/chemistry , Soluble Guanylyl Cyclase/metabolism
5.
Int J Pharm ; 610: 121220, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34687814

ABSTRACT

Nitric oxide (NO) has emerged as a promising antibacterial agent, where NO donor compounds have been explored. Here, we investigated the role of a silica nanoparticle containing nitroprusside (MPSi-NP) as a NO donor agent against methicillin-sensitive (ATCC 25,923 and ATCC 12228) and methicillin-resistant (ATCC 700,698 and ATCC 35984) Staphylococcus strains. Biofilm inhibition was studied along with antibiotic activity in combination with standard antibiotics (ampicillin and tetracycline). MPSi-NP exhibited thermal release of 63% of NO within 24 h, while free nitroprusside released only 18% during a dialysis assay, indicating an assisted release of NO mediated by the nanoparticles. This nanomaterial showed only a moderate activity in blocking biofilm production, but exhibited a significant decrease in the number of viable bacterial cells (over 600-fold for Staphylococcus aureus ATCC 700,698 and Staphylococcus epidermidis ATCC 35984). Remarkably, even using MPSi-NP at concentrations below any antibacterial action, its combination with ampicillin promoted a significant decrease in MIC for resistant strains of S. aureus ATCC 700,698 (2-fold) and S. epidermidis ATCC 35,984 (4-fold). A carbopol-based gel formulation with MPSi-NP (0.5% w/w) was prepared and showed a zone of inhibition of 7.7 ± 0.6 mm for S. epidermidis ATCC 35984. Topical use of MPSi-NP in combination with antibiotics might be a manageable strategy to prevent and eventually treat complicated resistant bacterial infections.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Anti-Bacterial Agents/pharmacology , Biofilms , Humans , Microbial Sensitivity Tests , Nitric Oxide Donors/pharmacology , Renal Dialysis , Staphylococcus aureus
6.
PLoS One ; 16(3): e0248394, 2021.
Article in English | MEDLINE | ID: mdl-33711054

ABSTRACT

We aimed at evaluating the anti-asthmatic effect of cis-[Ru(bpy)2(2-MIM)(NO)](PF6)3 (FOR811A), a nitrosyl-ruthenium compound, in a murine model of allergic asthma. The anti-asthmatic effects were analyzed by measuring the mechanical lung and morphometrical parameters in female Swiss mice allocated in the following groups: untreated control (Ctl+Sal) and control treated with FOR811A (Ctl+FOR), along asthmatic groups untreated (Ast+Sal) and treated with FOR811A (Ast+FOR). The drug-protein interaction was evaluated by in-silico assay using molecular docking. The results showed that the use of FOR811A in experimental asthma (Ast+FOR) decreased the pressure-volume curve, hysteresis, tissue elastance, tissue resistance, and airway resistance, similar to the control groups (Ctl+Sal; Ctl+FOR). However, it differed from the untreated asthmatic group (Ast+Sal, p<0.05), indicating that FOR811A corrected the lung parenchyma and relaxed the smooth muscles of the bronchi. Similar to control groups (Ctl+Sal; Ctl+FOR), FOR811A increased the inspiratory capacity and static compliance in asthmatic animals (Ast+Sal, p<0.05), showing that this metallodrug improved the capacity of inspiration during asthma. The morphometric parameters showed that FOR811A decreased the alveolar collapse and kept the bronchoconstriction during asthma. Beyond that, the molecular docking using FOR811A showed a strong interaction in the distal portion of the heme group of the soluble guanylate cyclase, particularly with cysteine residue (Cys141). In summary, FOR811A relaxed bronchial smooth muscles and improved respiratory mechanics during asthma, providing a protective effect and promising use for the development of an anti-asthmatic drug.


Subject(s)
Anti-Asthmatic Agents , Asthma , Nitric Oxide Donors , Organometallic Compounds , Respiratory Mechanics/drug effects , Ruthenium , Animals , Anti-Asthmatic Agents/chemistry , Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Asthma/physiopathology , Female , Mice , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/pharmacology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Ruthenium/chemistry , Ruthenium/pharmacology
7.
J Inorg Biochem ; 210: 111133, 2020 09.
Article in English | MEDLINE | ID: mdl-32619898

ABSTRACT

Nitric oxide (NO) and nitroxyl (HNO) have gained broad attention due to their roles in several physiological and pathophysiological processes. Remarkably, these sibling species can exhibit opposing effects including the promotion of angiogenic activity by NO compared to HNO, which blocks neovascularization. While many NO donors have been developed over the years, interest in HNO has led to the recent emergence of new donors. However, in both cases there is an expressive lack of iron-based compounds. Herein, we explored the novel chemical reactivity and stability of the trans-[Fe(cyclam)(NO)Cl]Cl2 (cyclam = 1,4,8,11-tetraazacyclotetradecane) complex. Interestingly, the half-life (t1/2) for NO release was 1.8 min upon light irradiation, vs 5.4 h upon thermal activation at 37 °C. Importantly, spectroscopic evidence supported the generation of HNO rather than NO induced by glutathione. Moreover, we observed significant inhibition of NO donor- or hypoxia-induced HIF-1α (hypoxia-inducible factor 1α) accumulation in breast cancer cells, as well as reduced vascular tube formation by endothelial cells pretreated with the trans-[Fe(cyclam)(NO)Cl]Cl2 complex. Together, these studies provide the first example of an iron-nitrosyl complex with anti-angiogenic activity as well as the potential dual activity of this compound as a NO/HNO releasing agent, which warrants further pharmacological investigation.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Coordination Complexes/pharmacology , Nitric Oxide Donors/pharmacology , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/radiation effects , Animals , Cell Line, Tumor , Coordination Complexes/chemical synthesis , Coordination Complexes/radiation effects , Glutathione/chemistry , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Iron/chemistry , Iron/radiation effects , Mice , Nitric Oxide/metabolism , Nitric Oxide Donors/chemical synthesis , Nitric Oxide Donors/radiation effects , Nitrogen Oxides/metabolism , Rats , Temperature , Ultraviolet Rays , Vasodilator Agents/chemical synthesis , Vasodilator Agents/pharmacology , Vasodilator Agents/radiation effects
8.
Biofouling ; 36(4): 442-454, 2020 04.
Article in English | MEDLINE | ID: mdl-32447980

ABSTRACT

The aim of this study was to investigate the antibacterial activity, antibiotic-associated synergy, and anti-biofilm activity of the ruthenium complex, cis-[RuCl2 (dppb) (bqdi)]2+ (RuNN). RuNN exhibited antimicrobial activity against Gram-positive bacteria with minimum inhibitory concentration (MIC) values ranging from 15.6 to 62.5 µg ml-1 and minimum bactericidal concentration (MBC) values ranging from 62.5 to 125 µg ml-1. A synergistic effect against Staphylococcus spp. was observed when RuNN was combined with ampicillin, and the range of associated fractional inhibitory concentration index (FICI) values was 0.187 to 0.312. A time-kill curve indicated the bactericidal activity of RuNN in the first 1-5 h. In general, RuNN inhibited biofilm formation and disrupted mature biofilms. Furthermore, RuNN altered the cellular morphology of S. aureus biofilms. Further, RuNN did not cause hemolysis of erythrocytes. The results of this study provide evidence that RuNN is a novel therapeutic candidate to treat bacterial infections caused by Staphylococcus biofilms.


Subject(s)
Anti-Bacterial Agents , Biofilms , Ruthenium , Staphylococcus aureus , Microbial Sensitivity Tests
9.
Res Vet Sci ; 130: 153-160, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32193002

ABSTRACT

The aim of this study was to investigate the antihypertensive properties of cis-[Ru(bpy)2ImN(NO)]3+ (FOR0811) in normotensive and in Nω-nitro-L-arginine methyl ester (L-NAME)-induced hypertensive rats. Vasorelaxant effects were analyzed by performing concentration response curve to FOR0811 in rat aortic rings in the absence or presence of 1H-[1,2,4]-oxadiazolo-[4,3,-a]quinoxalin-1-one (ODQ), L-cysteine or hydroxocobalamin. Normotensive and L-NAME-hypertensive rats were treated with FOR0811 and the effects in blood pressure and heart rate variability in the frequency domain (HRV) were followed. FOR0811 induced relaxation in rat aortic rings. Neither endothelium removal nor L-cysteine altered the FOR0811 effects. However, the incubation with ODQ and hydroxocobalamin completely blunted FOR0811 effects. FOR0811 administered intravenously by bolus infusion (0.01-1 mg/bolus) or chronically by using subcutaneous implanted osmotic pumps significantly reduced the mean arterial blood pressure. The effect was long lasting and did not induce reflex tachycardia. FOR0811 prevented both LF and VLF increases in L-NAME hypertensive rats and has antihypertensive properties. This new ruthenium complex compound might be a promising nitric oxide donor to treat cardiovascular diseases.


Subject(s)
Antihypertensive Agents/pharmacology , Hypertension/drug therapy , Nitric Oxide Donors/pharmacology , Organometallic Compounds/pharmacology , Ruthenium/pharmacology , Vasodilator Agents/pharmacology , Animals , Blood Pressure/drug effects , Heart Rate/drug effects , Hypertension/chemically induced , Male , NG-Nitroarginine Methyl Ester/administration & dosage , Rats , Rats, Wistar
10.
J Biol Inorg Chem ; 25(3): 419-428, 2020 05.
Article in English | MEDLINE | ID: mdl-32172453

ABSTRACT

The cis-[Ru(bpy)2(Met)](PF6)2 complex, where Met = L-methionine and bpy = 2,2'-bipyridine, was prepared and fully characterized. This complex was subjected to blue and green light photolysis (453 and 505 nm, respectively) in aqueous solution, leading to the release of methionine and formation of the cis-[Ru(bpy)2(H2O)2]2+ ion. This latter photoproduct was shown to subsequently interact with DNA, while DNA photocleavage was noticed. In agreement with these reactivities, this compound exhibited an exciting antibacterial action, particularly against Gram-positive bacteria Staphylococcus aureus and Staphylococcus epidermidis, which was enhanced upon blue light irradiation. Altogether, these results showed that our strategy was successful in producing light-triggered DNA-binding agents with pharmacological potential and a likely blocking reagent for efficient peptide chemistry formation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Coordination Complexes/pharmacology , Methionine/pharmacology , Ruthenium/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus epidermidis/drug effects , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , DNA/drug effects , DNA Cleavage , Light , Male , Methionine/chemistry , Microbial Sensitivity Tests , Photochemical Processes , Ruthenium/chemistry , Salmon , Spermatozoa/chemistry
11.
J Inorg Biochem ; 206: 111048, 2020 05.
Article in English | MEDLINE | ID: mdl-32151873

ABSTRACT

Inflammation is a physiological process triggered in response to tissue damage, and involves events related to cell recruitment, cytokines release and reactive oxygen species (ROS) production. Failing to control the process duration lead to chronification and may be associated with the development of various pathologies, including autoimmune diseases and cancer. Considering the pharmacological potential of metal-based compounds, two new ruthenium complexes were synthesized: cis-[Ru(NO2)(bpy)2(5NIM)]PF6 (1) and cis-[RuCl(bpy)2(MTZ)]PF6 (2), where bpy = 2,2'-bipyridine, 5NIM = 5-nitroimidazole and MTZ = metronidazole. Both products were characterized by spectroscopic techniques, followed by Density Functional Theory (DFT) calculations in order to support experimental findings. Afterwards, their in vitro cytotoxic, antioxidant and anti-inflammatory activities were investigated. Compounds 1 and 2 presented expressive in vitro antioxidant activity, reducing lipid peroxidation and decreasing intracellular ROS levels with comparable effectiveness to the standard steroidal drug dexamethasone or α-tocopherol. These complexes showed no noticeable cytotoxicity on the tested cancer cell lines. Bactericidal assay against metronidazole-resistant Helicobacter pylori, a microorganism able to disrupt oxidative balance, unraveled compound 1 moderate activity over that strain. Besides this, it was able to inhibit interleukin-6 (IL-6) and tumor necrosis factor-α (TNF- α) production as well as interleukin-1ß (IL-1ß) and cyclooxygenase-2 (COX-2) expression in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. This latter activity is remarkable, which has not been reported for other ruthenium-based complexes. Altogether, these results suggest cis-[Ru(NO2)(bpy)2(5NIM)]PF6 complex has potential pharmacological application as an anti-inflammatory agent that deserve further biological investigation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Coordination Complexes/pharmacology , Imidazoles/chemistry , Ruthenium/chemistry , A549 Cells , Animals , Anti-Bacterial Agents/chemistry , Anti-Inflammatory Agents/chemistry , Antineoplastic Agents/chemistry , Antioxidants/chemistry , Bacteria/drug effects , Cell Proliferation , Coordination Complexes/chemistry , Humans , Lipid Peroxidation , MCF-7 Cells , Mice , Molecular Structure , RAW 264.7 Cells , Superoxides/metabolism
12.
Acta Cir Bras ; 34(12): e201901201, 2020.
Article in English | MEDLINE | ID: mdl-32022101

ABSTRACT

PURPOSE: To evaluate the effect of Rut-bpy (Cis-[Ru(bpy)2(SO3)(NO)]PF 6), a novel nitric oxide donor, able to modulate the histological changes caused by the NASID (meloxicam). METHODS: Wistar rats were assigned into three groups (n=6 rats/group): Sham group (saline solution), NSAID group (meloxicam - 15 mg/kg) and Rut-bpy group (100 mg/kg of Rut-bpy associated with 15mg/kg of meloxicam). At the end of experiments, kidneys were removed for histological study, fractal dimension and lacunarity in all animals. RESULTS: At the histological examination, all animals (six animals - 100 %) in the NSAID group had membrane thickening and other changes (necrosis, acute tubular congestion and vascular congestion); on the other hand, only one animal (16.6 %) of the Rut-bpy group had congestion. The fractal dimension and lacunarity were greater in the control and Rut-bpy group than in NSAIDs group (p<0.05). CONCLUSION: Rut-bpy may prevent renal histological changes in rats caused by meloxicam.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Meloxicam/adverse effects , Nitric Oxide Donors/pharmacology , Organometallic Compounds/pharmacology , Ruthenium/pharmacology , 2,2'-Dipyridyl/analogs & derivatives , Animals , Fractals , Kidney Diseases/pathology , Male , Random Allocation , Rats, Wistar , Reproducibility of Results
13.
Acta Trop ; 192: 61-65, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30689977

ABSTRACT

Leishmaniasis is a parasitic disease caused by protozoa of the genus Leishmania. There are many complications presented by the current treatment, as high toxicity, high cost and parasite resistance, making the development of new therapeutic agents indispensable. The present study aims to evaluate the leishmanicidal potential of ruthenium nitrosyl complex cis-[Ru(bpy)2(SO3)(NO)](PF6) against Leishmania (Viannia) braziliensis. The effect of this metal complex on parasite-host interaction was evaluated by in vitro efficacy test in dermal fibrobast cells in the presence of different concentrations (1, 10, 50 and 100 µM) and by in vivo efficacy tests performed in the presence of two different concentrations of complex (100 µg/kg/day or 300 µg/kg/day) evaluating its effect on the size of the lesion and the number of parasites present in the draining lymph nodes in hamsters. Even at the lowest concentration of 1 µM of ruthenium complex, it was observed a significant decrease of the infected cells, after 24 h exposure in vitro, with total reduction at 50 µM of the ruthenium complex. In the in vivo cutaneous infection model, administration of daily doses of 300 µg/kg/day of complex reduced significantly lesion size by 51% (p < 0.05), with a 99.9% elimination of the parasites found in the lymph nodes (p < 0.001). The results suggest a promising leishmanicidal effect by that ruthenium nitrosyl complex against L. (V.) braziliensis.


Subject(s)
Leishmania braziliensis/drug effects , Ruthenium Compounds/pharmacology , Animals , Cricetinae , Dose-Response Relationship, Drug , Host-Parasite Interactions , Skin
14.
FEBS J ; 286(3): 479-494, 2019 02.
Article in English | MEDLINE | ID: mdl-30570222

ABSTRACT

A major challenge to the control and eventual eradication of Mycobacterium tuberculosis infection is this pathogen's prolonged dormancy. The heme-based oxygen sensor protein DevS (DosS) plays a key role in this phenomenon, because it is a major activator of the transcription factor DevR. When DevS is active, its histidine protein kinase region is ON and it phosphorylates and activates DevR, which can induce the transcription of the dormancy regulon genes. Here, we have investigated the mechanism by which the ligation of molecular oxygen to a heme-binding domain in DevS switches OFF its histidine protein kinase region. To shed light on the oligomerization states of this protein and possible protein-surfaces of interaction, we used analytical gel filtration, together with dynamic light scattering, fluorescence spectroscopy and chemical crosslinking. We found that DevS exists as three major species: an octamer, a tetramer and a dimer. These three states were observed for the concentration range between 0.5 and 20 µm DevS, but not below 0.1 µm. Levels of DevS in M. tuberculosis are expected to range from 5 to 26 µm. When this histidine protein kinase was OFF, the DevS was mainly tetrameric and dimeric; by contrast, when the kinase was ON, the protein was predominantly octameric. The changes in quaternary structure were rapid upon binding to the physiological signal. This finding represents a novel strategy for switching the activity of a two-component heme-based sensor. An enhanced understanding of this process might potentially lead to the design of novel regulatory agents that target the multimer interfaces for treatment of latent tuberculosis.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Heme/chemistry , Mycobacterium tuberculosis/drug effects , Oxygen/pharmacology , Protamine Kinase/chemistry , Protein Kinases/genetics , Bacterial Proteins/metabolism , Chromatography, Gel , Cloning, Molecular , DNA-Binding Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Heme/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Latent Tuberculosis/microbiology , Latent Tuberculosis/pathology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/isolation & purification , Mycobacterium tuberculosis/metabolism , Oxygen/chemistry , Oxygen/metabolism , Phosphorylation , Protamine Kinase/genetics , Protamine Kinase/metabolism , Protein Interaction Domains and Motifs , Protein Kinases/metabolism , Protein Multimerization , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Regulon , Signal Transduction , Spectrometry, Fluorescence , Transcription, Genetic/drug effects
15.
Acta cir. bras ; 34(12): e201901201, 2019. graf
Article in English | LILACS | ID: biblio-1054690

ABSTRACT

Abstract Purpose To evaluate the effect of Rut-bpy (Cis-[Ru(bpy)2(SO3)(NO)]PF 6), a novel nitric oxide donor, able to modulate the histological changes caused by the NASID (meloxicam). Methods Wistar rats were assigned into three groups (n=6 rats/group): Sham group (saline solution), NSAID group (meloxicam - 15 mg/kg) and Rut-bpy group (100 mg/kg of Rut-bpy associated with 15mg/kg of meloxicam). At the end of experiments, kidneys were removed for histological study, fractal dimension and lacunarity in all animals. Results At the histological examination, all animals (six animals - 100 %) in the NSAID group had membrane thickening and other changes (necrosis, acute tubular congestion and vascular congestion); on the other hand, only one animal (16.6 %) of the Rut-bpy group had congestion. The fractal dimension and lacunarity were greater in the control and Rut-bpy group than in NSAIDs group (p<0.05). Conclusion Rut-bpy may prevent renal histological changes in rats caused by meloxicam.


Subject(s)
Animals , Male , Organometallic Compounds/pharmacology , Ruthenium/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Nitric Oxide Donors/pharmacology , Meloxicam/adverse effects , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Random Allocation , Reproducibility of Results , Rats, Wistar , Fractals , Kidney Diseases/pathology
16.
J Inorg Biochem ; 167: 12-20, 2017 02.
Article in English | MEDLINE | ID: mdl-27893989

ABSTRACT

Heme-based sensors have emerged during the last 20years as being a large family of proteins that occur in all kingdoms of life. A myriad of biological adaptations are associated with these sensors, which include vasodilation, bacterial virulence, dormancy, chemotaxis, biofilm formation, among others. Due to the key activities regulated by these proteins along with many other systems that use similar output domains, there is a growing interest in developing small molecules as their regulators. Here, we review the development of potential activators and inhibitors for many of these systems, including human soluble guanylate cyclase, c-di-GMP-related enzymes, Mycobacterium tuberculosis DevR/DevS/DosT (differentially expressed in virulent strain response regulator/sensor/dormancysurvival sensorT), the Rev-erb-α and ß nuclear receptor, among others. The possible roles of these molecules as biochemical tools, therapeutic agents, and novel antibiotics are critically examined.


Subject(s)
Anti-Bacterial Agents , Biosensing Techniques/methods , Drug Discovery/methods , Mycobacterium tuberculosis , Soluble Guanylyl Cyclase/chemistry , Animals , Anti-Bacterial Agents/analysis , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Heme , Humans
17.
PLoS One ; 10(5): e0125101, 2015.
Article in English | MEDLINE | ID: mdl-25973801

ABSTRACT

The fact that drugs currently used in the treatment of Leishmania are highly toxic and associated with acquired resistance has promoted the search for new therapies for treating American tegumentary leishmaniasis (ATL). In this study, BALB/c mice were injected in the hind paw with Leishmania (Leishmania) amazonensis and subsequently treated with a combination of nitric oxide (NO) donor (cis-[Ru(bpy) 2imN(NO)](PF6)3) (Ru-NO), given by intraperitoneal injection, and oral Brazilian propolis for 30 days. Ru-NO reached the center of the lesion and increased the NO level in the injured hind paw without lesion exacerbation. Histological and immunological parameters of chronic inflammation showed that this combined treatment increased the efficacy of macrophages, determined by the decrease in the number of parasitized cells, leading to reduced expression of proinflammatory and tissue damage markers. In addition, these drugs in combination fostered wound healing, enhanced the number of fibroblasts, pro-healing cytokines and induced collagen synthesis at the lesion site. Overall, our findings suggest that the combination of the NO donor Ru-NO and Brazilian propolis alleviates experimental ATL lesions, highlighting a new therapeutic option that can be considered for further in vivo investigations as a candidate for the treatment of cutaneous leishmaniasis.


Subject(s)
Leishmania/drug effects , Leishmaniasis, Cutaneous/drug therapy , Nitric Oxide Donors/chemistry , Nitric Oxide/pharmacology , Propolis/pharmacology , Administration, Oral , Animals , Cell Movement/drug effects , Collagen/biosynthesis , Cytokines/biosynthesis , Drug Synergism , Drug Therapy, Combination , Female , Fibroblasts/drug effects , Fibroblasts/parasitology , Fibroblasts/pathology , Hindlimb , Injections, Intraperitoneal , Leishmania/growth & development , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/pathology , Macrophages/drug effects , Macrophages/parasitology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Nitric Oxide Donors/pharmacology , Wound Healing/drug effects
18.
J Inorg Biochem ; 140: 236-44, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25189361

ABSTRACT

Tuberculosis has re-emerged as a worldwide threat, which has motivated the development of new drugs. The antituberculosis complex Na3[Fe(CN)5(isoniazid)] (IQG607) in particular is of interest on account of its ability to overcome resistance. IQG607 has the potential for redox-mediated-activation, in which an acylpyridine (isonicotinoyl) radical could be generated without assistance from the mycobacterial KatG enzyme. Here, we have investigated the reactivity of IQG607 toward hydrogen peroxide and superoxide, well-known intracellular oxidizing agents that could play a key role in the redox-mediated-activation of this compound. HPLC, NMR and electronic spectroscopy studies showed a very fast oxidation rate for bound isoniazid, over 460-fold faster than free isoniazid oxidation. A series of EPR spin traps were used for detection of isonicotinoyl and derived radicals bound to iron. This is the first report for an isonicotinoyl radical bound to a metal complex, supported by (14)N and (1)H hyperfine splittings for the POBN and PBN trapped radicals. POBN and PBN exhibited average hyperfine coupling constants of aN=15.6, aH=2.8 and aN=15.4, aH=4.7, respectively, which are in close agreement to the isonicotinoyl radical. Radical generation is thought to play a major role in the mechanism of action of isoniazid and this work provides strong evidence for its production within IQG607, which, along with biological and chemical oxidation data, support a redox-mediated activation mechanism. More generally the concept of redox activation of metallo prodrugs could be applied more widely for the design of therapeutic agents with novel mechanisms of action.


Subject(s)
Antitubercular Agents/therapeutic use , Ferrous Compounds/therapeutic use , Isoniazid/analogs & derivatives , Tuberculosis/drug therapy , Antitubercular Agents/chemistry , Electron Spin Resonance Spectroscopy , Ferrous Compounds/chemistry , Humans , Isoniazid/chemistry , Isoniazid/therapeutic use , Magnetic Resonance Spectroscopy , Oxidation-Reduction , Superoxides/chemistry
19.
Neurochem Res ; 37(4): 749-58, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22160748

ABSTRACT

Rut-bpy is a novel nitrosyl-ruthenium complex releasing NO into the vascular system. We evaluated the effect of Rut-bpy (100 mg/kg) on a rat model of brain stroke. Forty rats were assigned to four groups (Saline solution [SS], Rut-bpy, SS+ischemia-reperfusion [SS+I/R] and Rut-bpy+ischemia-reperfusion [Rut-bpy+I/R]) with their mean arterial pressure (MAP) continuously monitored. The groups were submitted (SS+I/R and Rut-bpy+I/R) or not (SS and Rut-bpy) to incomplete global brain ischemia by occlusion of the common bilateral carotid arteries during 30 min followed by reperfusion for further 60 min. Thirty minutes before ischemia, rats were treated pairwise by intraperitoneal injection of saline solution or Rut-bpy. At the end of experiments, brain was removed for triphenyltetrazolium chloride staining in order to quantify the total ischemic area. In a subset of rats, hippocampus was obtained for histopathology scoring, nitrate and nitrite measurements, immunostaining and western blotting of the nuclear factor- κB (NF-κB). Rut-bpy pre-treatment decreased MAP variations during the transition from brain ischemia to reperfusion and decreased the fractional injury area. Rut-bpy pre-treatment reduced NF-κB hippocampal immunostaining and protein expression with improved histopathology scoring as compared to the untreated operated control. In conclusion, Rut-bpy improved the total brain infarction area and hippocampal neuronal viability in part by inhibiting NF-κB signaling and helped to stabilize the blood pressure during the transition from ischemia to reperfusion.


Subject(s)
Anesthesia , Brain Ischemia/prevention & control , Ischemic Preconditioning/methods , Nitric Oxide Donors/administration & dosage , Reperfusion Injury/prevention & control , Ruthenium Compounds/administration & dosage , Anesthesia/methods , Animals , Brain/blood supply , Brain/drug effects , Brain/metabolism , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Male , Neuroprotective Agents/administration & dosage , Rats , Rats, Wistar , Reactive Nitrogen Species/metabolism , Reperfusion Injury/metabolism
20.
J Biol Inorg Chem ; 17(2): 275-83, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21952749

ABSTRACT

For over a decade, tuberculosis (TB) has been the leading cause of death among infectious diseases. Since the 1950s, isoniazid has been used as a front-line drug in the treatment of TB; however, resistant TB strains have limited its use. The major route of isoniazid resistance relies on KatG enzyme disruption, which does not promote an electron transfer reaction. Here, we investigated the reactivity of isoniazid metal complexes as prototypes for novel self-activating metallodrugs against TB with the aim to overcome resistance. Reactivity studies were conducted with hydrogen peroxide, hexacyanoferrate(III), and aquopentacyanoferrate(III). The latter species showed a preference for the inner-sphere electron transfer reaction pathway. Additionally, electron transfer reaction performed with either free isoniazid or (isoniazid)pentacyanoferrate(II) complex resulted in similar oxidized isoniazid derivatives as observed when the KatG enzyme was used. However, upon metal coordination, a significant enhancement in the formation of isonicotinic acid was observed compared with that of isonicotinamide. These results suggest that the pathway of a carbonyl-centered radical might be favored upon coordination to the Fe(II) owing to the π-back-bonding effect promoted by this metal center; therefore, the isoniazid metal complex could serve as a potential metallodrug. Enzymatic inhibition assays conducted with InhA showed that the cyanoferrate moiety is not the major player involved in this inhibition but the presence of isoniazid is required in this process. Other isoniazid metal complexes, [Ru(CN)(5)(izd)](3-) and [Ru(NH(3))(5)(izd)](2+) (where izd is isoniazid), were also unable to inhibit InhA, supporting our proposed self-activating mechanism of action. We propose that isoniazid reactivity can be rationally modulated by metal coordination chemistry, leading to the development of novel anti-TB metallodrugs.


Subject(s)
Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Drug Design , Isoniazid/chemistry , Isoniazid/pharmacology , Mycobacterium tuberculosis/drug effects , Oxidoreductases/antagonists & inhibitors , Drug Resistance, Bacterial , Ferric Compounds/chemistry , Humans , Mycobacterium tuberculosis/enzymology , Oxidation-Reduction , Tuberculosis/drug therapy , Tuberculosis/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...