Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 610
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Urol ; 31(9): 1052-1060, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38884570

ABSTRACT

OBJECTIVES: To investigate roles of brain carbon monoxide (CO), an endogenous gasotransmitter, in regulation of the rat micturition reflex. METHODS: In urethane-anesthetized (0.8 g/kg, ip) male rats, evaluation of urodynamic parameters was started 1 h before intracerebroventricular administration of CORM-3 (CO donor) or ZnPP (non-selective inhibitor of heme oxygenase, a CO producing enzyme) and continued for 2 h after the administration. We also investigated effects of centrally pretreated SR95531 (GABAA receptor antagonist) or SCH50911 (GABAB receptor antagonist) on the CORM-3-induced response. RESULTS: CORM-3 significantly prolonged intercontraction intervals (ICIs) without changing maximal voiding pressure (MVP), while ZnPP significantly shortened ICI and reduced single-voided volume and bladder capacity without affecting MVP, post-voided residual volume, or voiding efficiency. The ZnPP-induced ICI shortening was reversed by CORM-3. The CORM-3-induced ICI prolongation was significantly attenuated by centrally pretreated SR95531 or SCH50911, respectively. CONCLUSIONS: Brain CO can suppress the rat micturition reflex through brain γ-aminobutyric acid (GABA) receptors.


Subject(s)
Brain , Carbon Monoxide , Rats, Sprague-Dawley , Urinary Bladder , Urination , Animals , Male , Urination/drug effects , Rats , Carbon Monoxide/pharmacology , Brain/drug effects , Brain/physiology , Urinary Bladder/drug effects , Urinary Bladder/physiology , Reflex/drug effects , Organometallic Compounds/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Urodynamics/drug effects , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Receptors, GABA/drug effects , Receptors, GABA/metabolism
2.
Biochemistry ; 60(9): 689-698, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33621054

ABSTRACT

Iron is an essential micronutrient for the survival and virulence of the bacterial pathogen Pseudomonas aeruginosa. To overcome iron withholding and successfully colonize a host, P. aeruginosa uses a variety of mechanisms to acquire iron, including the secretion of high-affinity iron chelators (siderophores) or the uptake and utilization of heme. P. aeruginosa heme oxygenase (HemO) plays pivotal roles in heme sensing, uptake, and utilization and has emerged as a therapeutic target for the development of antipseudomonal agents. Using a high-throughput fluorescence quenching assay combined with minimum inhibitory concentration measurements, we screened the Selleck Bioactive collection of 2100 compounds and identified acitretin, a Food and Drug Administration-approved oral retinoid, as a potent and selective inhibitor of HemO. Acitretin binds to HemO with a KD value of 0.10 ± 0.02 µM and inhibits the growth of P. aeruginosa PAO1 with an IC50 of 70 ± 18 µg/mL. In addition, acitretin showed good selectivity for HemO, which uniquely generates BVIXß/δ, over human heme oxygenase (hHO1) and other BVIXα-producing homologues such as the heme oxygenases from Neisseria meningitidis (nmHO) and Acinetobacter baumannii (abHO). The binding of acitretin within the HemO active site was confirmed by 1H-15N heteronuclear single-quantum coherence nuclear magnetic resonance, and molecular modeling provided further insight into potential interactions of acitretin with residues specific for orienting heme in the ß/δ selective HemO. Moreover, at 20 µM, acitretin inhibited the enzymatic activity of HemO in P. aeruginosa cells by >60% and effectively blocked the ability of P. aeruginosa to sense and acquire heme as demonstrated in the ß-galactosidase transcriptional reporter assay.


Subject(s)
Acitretin/pharmacology , Anti-Bacterial Agents/pharmacology , Drug Repositioning/methods , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Iron/pharmacology , Pseudomonas aeruginosa/drug effects , Gene Expression Regulation, Bacterial , Humans , Keratolytic Agents/pharmacology , Pseudomonas aeruginosa/enzymology
3.
Biochem Biophys Res Commun ; 551: 7-13, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33713981

ABSTRACT

Both the Nuclear factor-erythroid 2 p45-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) antioxidant pathway and Nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) pathway are considered essential for the development of acute lung injury (ALI)/ARDS induced by sepsis. Our aim was to study the role of Nrf2/HO-1 pathway on activation of the NLRP3 in the protective effect of marrow mesenchymal stem cells (BMSCs) on LPS-induced ALI. We found that BMSCs ameliorated ALI as evidenced by 1) decreased histopathological injury, wet/dry ratio, and protein permeability index in lung; 2) decreased reactive oxygen species (ROS), malondialdehyde (MDA), and protein carbonyl content and restored the activity of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) in lung tissue; 3) reduced LPS-induced increase in inflammatory cell count and promotion of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6 levels in bronchoalveolar lavage fluid (BALF); 4) improvement in the four-day survival rate of animals; and 5) enhanced expression of Nrf2 and HO-1 and decreased expression of NOD-like receptor protein 3(NLRP3) and caspase-1 (p20) in lung tissue. Of note, Nrf2 transcription factor inhibitor brusatol and HO-1 inhibitor tin protoporphyrin IX (SnppIX) reversed BMSCs induced down-expression of NLRP3 and caspase-1 (p20), and inhibited the protective effects of BMSCs. These findings demonstrated that the Nrf2-mediated HO-1 signaling pathway plays a critical role in the protective effects of BMSCs on LPS-induced ALI. BMSCs may play an anti-inflammatory effect partly through the Nrf2/HO-1-dependent NLRP3 pathway.


Subject(s)
Acute Lung Injury/metabolism , Acute Lung Injury/prevention & control , Bone Marrow Cells/cytology , Endotoxins/adverse effects , Heme Oxygenase (Decyclizing)/metabolism , Mesenchymal Stem Cells/metabolism , NF-E2-Related Factor 2/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Acute Lung Injury/chemically induced , Animals , Antioxidants/metabolism , Capillary Permeability/drug effects , Gene Expression Regulation , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Inflammasomes/drug effects , Inflammasomes/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Lung/blood supply , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mesenchymal Stem Cells/cytology , NF-E2-Related Factor 2/antagonists & inhibitors , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction , Survival Rate
4.
Bioorg Chem ; 117: 105428, 2021 12.
Article in English | MEDLINE | ID: mdl-34710668

ABSTRACT

Up-regulation of HO-1 had been frequently reported in different cases and types of human malignancies. Since poor clinical outcomes are reported in these cases, this enzyme's inhibition is considered a valuable and proven anticancer approach. To identify novel HO-1 inhibitors suitable for drug development, we report a structure-guided fragment-based approach to identify new lead compounds. Different parts of the selected molecules were analyzed, and the different series of novel compounds were virtually evaluated. The growing experiments of the classical HO-1 inhibitors structure led us to different hit-compounds. A synthetic pathway for six selected molecules was designed, and the compounds were synthesized. The biological activity revealed that molecules 10 and 12 inhibit the HO-1 activity with an IC50 of 1.01 and 0.90 µM, respectively. This study suggested that our growing approach was successful, and these results are ongoing for further development.


Subject(s)
Enzyme Inhibitors/pharmacology , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Imidazoles/pharmacology , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Heme Oxygenase (Decyclizing)/metabolism , Imidazoles/chemical synthesis , Imidazoles/chemistry , Ligands , Models, Molecular , Molecular Structure , Rats , Structure-Activity Relationship
5.
Molecules ; 26(24)2021 Dec 19.
Article in English | MEDLINE | ID: mdl-34946766

ABSTRACT

The development of the field of nanotechnology has revolutionized various aspects in the fields of modern sciences. Nano-medicine is one of the primary fields for the application of nanotechnology techniques. The current study sheds light on the reno-protective impacts of gold nano-particles; nanogold (AuNPs) against 5-flurouracil (5-FU)-induced renal toxicity. Indeed, the use of 5-FU has been associated with kidney injury which greatly curbs its therapeutic application. In the current study, 5-FU injection was associated with a significant escalation in the indices of renal injury, i.e., creatinine and urea. Alongside this, histopathological and ultra-histopathological changes confirmed the onset of renal injury. Both gene and/or protein expression of nuclear factor erythroid 2-related factor 2 (Nrf-2) and downstream antioxidant enzymes revealed consistent paralleled anomalies. AuNPs administration induced a significant renal protection on functional, biochemical, and structural levels. Renal expression of the major sensor of the cellular oxidative status Nrf-2 escalated with a paralleled reduction in the renal expression of the other contributor to this axis, known as Kelch-like ECH-associated protein 1 (Keap-1). On the level of the effector downstream targets, heme oxygenase 1 (HO-1) and gamma-glutamylcysteine synthetase (γ-GCS) AuNPs significantly restored their gene and protein expression. Additionally, combination of AuNPs with 5-FU showed better cytotoxic effect on MCF-7 cells compared to monotreatments. Thus, it can be inferred that AuNPs conferred reno-protective impact against 5-FU with an evident modulatory impact on Nrf-2/Keap-1 and its downstream effectors, HO-1 and γ-GCS, suggesting its potential use in 5-FU regimens to improve its therapeutic outcomes and minimize its underlying nephrotoxicity.


Subject(s)
Fluorouracil/antagonists & inhibitors , Gold/pharmacology , Kidney/drug effects , Metal Nanoparticles/chemistry , Animals , Disease Models, Animal , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Gold/administration & dosage , Gold/chemistry , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/metabolism , Injections, Intraperitoneal , Kidney/injuries , Kidney/pathology , Metal Nanoparticles/administration & dosage , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Nanotechnology , Particle Size , Rats , Rats, Sprague-Dawley , gamma-Glutamylcyclotransferase/antagonists & inhibitors , gamma-Glutamylcyclotransferase/metabolism
6.
Biochem Biophys Res Commun ; 532(1): 151-158, 2020 10 29.
Article in English | MEDLINE | ID: mdl-32838965

ABSTRACT

Following renal ischemia-reperfusion injury (RIRI), because of the decrease in oxygen supply to the kidney, a large amount of oxygen-free radicals is generated, and in severe cases, tissue cells will undergo apoptosis or even die. Normobaric hyperoxia (NBHO) is a very common clinical adjuvant treatment. It restores the oxygen supply after renal ischemia and combats oxidative stress in tissues, thus playing a protective role. In this study, our aim is to elucidate the protective mechanism of NBHO inhalation in a rat RIRI model. We performed a surgical excision of the left kidney of the rat and established a right kidney solitary kidney model. Later, the right renal pedicle of the rat was clamped using a non-invasive vascular clamp for 45 min. After the vascular clamp was released and reperfused for 24 h, the rat was placed in a closed oxygen chamber. It was subjected to inhalation of high-concentration oxygen (50%-55%), 2 h daily, for 7 days.RIRI induces postoperative weight loss, impaired renal function, increased oxygen free radicals, reduced antioxidant substances, increased histopathological damage, and increased levels of apoptosis. These effects were significantly improved after treatment with NBHO. At the same time, NBHO significantly increased the expression levels of Nrf2 and HO-1 in the tissues after RIRI. To verify whether HO-1 induced by Nrf2 is involved in the resistance to oxidative stress, after the rat RIRI and before inhaling NBHO, we intraperitoneally injected HO-1 specific inhibitor zinc protoporphyrin (ZnPP) (45 µmol/Kg). However, we found that ZnPP reversed the protective effect of NBHO on RIRI in rats. Combining all the results, we have demonstrated the protective effect of NBHO on RIRI, which can be at least partially attributed to the activation of the Nrf2/HO-1 antioxidative stress pathway.


Subject(s)
Heme Oxygenase (Decyclizing)/metabolism , Hyperoxia/metabolism , Kidney/injuries , Kidney/metabolism , NF-E2-Related Factor 2/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Animals , Antioxidants/metabolism , Apoptosis , Atmospheric Pressure , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Male , Oxidative Stress , Protoporphyrins/pharmacology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Signal Transduction
7.
Circ Res ; 122(12): 1689-1702, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29545367

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PH) is a life-threatening condition associated with immune dysregulation and abnormal regulatory T cell (Treg) activity, but it is currently unknown whether and how abnormal Treg function differentially affects males and females. OBJECTIVE: To evaluate whether and how Treg deficiency differentially affects male and female rats in experimental PH. METHODS AND RESULTS: Male and female athymic rnu/rnu rats, lacking Tregs, were treated with the VEGFR2 (vascular endothelial growth factor receptor 2) inhibitor SU5416 or chronic hypoxia and evaluated for PH; some animals underwent Treg immune reconstitution before SU5416 administration. Plasma PGI2 (prostacyclin) levels were measured. Lung and right ventricles were assessed for the expression of the vasoprotective proteins COX-2 (cyclooxygenase 2), PTGIS (prostacyclin synthase), PDL-1 (programmed death ligand 1), and HO-1 (heme oxygenase 1). Inhibitors of these pathways were administered to athymic rats undergoing Treg immune reconstitution. Finally, human cardiac microvascular endothelial cells cocultured with Tregs were evaluated for COX-2, PDL-1, HO-1, and ER (estrogen receptor) expression, and culture supernatants were assayed for PGI2 and IL (interleukin)-10. SU5416-treatment and chronic hypoxia produced more severe PH in female than male athymic rats. Females were distinguished by greater pulmonary inflammation, augmented right ventricular fibrosis, lower plasma PGI2 levels, decreased lung COX-2, PTGIS, HO-1, and PDL-1 expression and reduced right ventricular PDL-1 levels. In both sexes, Treg immune reconstitution protected against PH development and raised levels of plasma PGI2 and cardiopulmonary COX-2, PTGIS, PDL-1, and HO-1. Inhibiting COX-2, HO-1, and PD-1 (programmed death 1)/PDL-1 pathways abrogated Treg protection. In vitro, human Tregs directly upregulated endothelial COX-2, PDL-1, HO-1, ERs and increased supernatant levels of PGI2 and IL-10. CONCLUSIONS: In 2 animal models of PH based on Treg deficiency, females developed more severe PH than males. The data suggest that females are especially reliant on the normal Treg function to counteract the effects of pulmonary vascular injury leading to PH.


Subject(s)
Hypertension, Pulmonary/prevention & control , Sex Factors , T-Lymphocytes, Regulatory/physiology , Angiogenesis Inhibitors/pharmacology , Animals , B7-H1 Antigen/analysis , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Chronic Disease , Cyclooxygenase 2/analysis , Cyclooxygenase 2/metabolism , Cytochrome P-450 Enzyme System/analysis , Cytochrome P-450 Enzyme System/metabolism , Epoprostenol/antagonists & inhibitors , Epoprostenol/blood , Epoprostenol/metabolism , Female , Heme Oxygenase (Decyclizing)/analysis , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/metabolism , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/etiology , Hypoxia/complications , Indoles/pharmacology , Intramolecular Oxidoreductases/analysis , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Lung/metabolism , Male , Prostaglandins I/biosynthesis , Pyrroles/pharmacology , Rats , Rats, Nude , Receptors, Estrogen/analysis , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , T-Lymphocytes, Regulatory/immunology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
8.
Bioorg Chem ; 104: 104310, 2020 11.
Article in English | MEDLINE | ID: mdl-33010625

ABSTRACT

The enzymatic family of heme oxygenase (HO) is accountable for heme breakdown. Among the two isoforms characterized to date, HO-2 is poorly investigated due to the lack of potent HO-2 chemical modulators and the greater attentiveness towards HO-1 isoform. In the present paper, we report the rational design and synthesis of HO-2 inhibitors achieved by modulating the volume of known HO-1 inhibitors. The inhibition preference has been moved from HO-1 to HO-2 by merely increasing the volume of the substituent in the western region of the inhibitors. Docking studies demonstrated that new derivatives soak differently in the two binding pockets, probably due to the presence of a Tyr187 residue in HO-2. These findings could be useful for the design of new selective HO-2 compounds.


Subject(s)
Enzyme Inhibitors/pharmacology , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Nitriles/pharmacology , Algorithms , Animals , Brain/enzymology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Heme Oxygenase (Decyclizing)/metabolism , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation , Molecular Structure , Nitriles/chemical synthesis , Nitriles/chemistry , Rats , Spleen/enzymology , Structure-Activity Relationship
9.
Bioorg Chem ; 99: 103777, 2020 06.
Article in English | MEDLINE | ID: mdl-32222619

ABSTRACT

Heme oxygenase-1 (HO-1) has been recognized as extensively involved in the development and aggravation of cancer, cell propagation and at in the mechanism of chemoresistance development. Low micromolar HO-1 inhibitors selective towards HO-2 has been recently reported, wherein the azole core and the hydrophobic residues are linked through a phenylethanolic spacer bearing a chiral center. Since less information are known about the stereoselective requirements for HO-1 inhibition, here we report the enantiomeric resolution of 1-(biphenyl-3-yl)-2-(1H-imidazol-1-yl)ethanol (1) and 1-[4-[(4-bromobenzyl)oxy]phenyl]-2-(1H-imidazol-1-yl)ethanol (2), two among the most potent and selective HO-1 inhibitors known thus far when tested as racemates. The absolute configuration was established for 1 by a combination of experimental and in silico derived electronic circular dichroism spectra, while docking approaches were useful in the case of compound 2. Biological evaluation of pure enantiomers highlighted higher HO-1 inhibitory activity of (R)-enantiomers. Docking studies demonstrated the importance of hydrogen bond interaction, more pronounced for the (R)-enantiomers, with a consensus water molecule within the binding pocket. The present study demonstrates that differences in three-dimensional structure amongst compounds 1 and 2 enantiomers affect significantly the selectivity of these HO-1 inhibitors.


Subject(s)
Azoles/pharmacology , Enzyme Inhibitors/pharmacology , Phenylethyl Alcohol/pharmacology , Animals , Azoles/chemistry , Density Functional Theory , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/metabolism , Male , Molecular Docking Simulation , Molecular Structure , Phenylethyl Alcohol/chemistry , Rats , Rats, Sprague-Dawley , Spleen/enzymology , Stereoisomerism , Structure-Activity Relationship
10.
Biotechnol Appl Biochem ; 67(3): 317-322, 2020 May.
Article in English | MEDLINE | ID: mdl-31746064

ABSTRACT

Epigallocatechin 3-gallate (EGCG) is a polyphenol that has been shown to have antioxidant and anti-inflammatory effects. In this study, collagen-induced arthritis (CIA) model, in Wistar albino rats, was used to elucidate the effect of EGCG on pathogenetic pathways in inflammatory arthritis. The levels of serum TNF-α, IL-17, malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx); the expression levels of tissue heme oxygenase-1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2); histopathologically, perisynovial inflammation and cartilage-bone destruction were examined. In the sham group, serum TNF-α, IL-17, and MDA levels increased, while SOD, CAT, GPx levels, and the expressions of Nrf2 and HO-1 decreased. On the other hand, in the EGCG administered groups, serum TNF-α, IL-17, and MDA levels improved, while SOD, CAT, GPx levels and the expressions of Nrf2 and HO-1 increased. Moreover, histopathological analysis has shown that perisynovial inflammation and cartilage-bone destruction decreased in the EGCG administered groups. These results suggest that EGCG has an antiarthritic effect by regulating the oxidative-antioxidant balance and cytokine levels in the CIA model, which is a surrogate experimental model of rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Catechin/analogs & derivatives , Cytokines/antagonists & inhibitors , Disease Models, Animal , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , NF-E2-Related Factor 2/antagonists & inhibitors , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Catechin/pharmacology , Collagen Type II , Cytokines/biosynthesis , Female , Heme Oxygenase (Decyclizing)/biosynthesis , NF-E2-Related Factor 2/biosynthesis , Rats , Rats, Wistar
11.
Int J Mol Sci ; 22(1)2020 Dec 30.
Article in English | MEDLINE | ID: mdl-33396647

ABSTRACT

The heme oxygenase (HO) system involves three isoforms of this enzyme, HO-1, HO-2, and HO-3. The three of them display the same catalytic activity, oxidating the heme group to produce biliverdin, ferrous iron, and carbon monoxide (CO). HO-1 is the isoform most widely studied in proinflammatory diseases because treatments that overexpress this enzyme promote the generation of anti-inflammatory products. However, neonatal jaundice (hyperbilirubinemia) derived from HO overexpression led to the development of inhibitors, such as those based on metaloproto- and meso-porphyrins inhibitors with competitive activity. Further, non-competitive inhibitors have also been identified, such as synthetic and natural imidazole-dioxolane-based, small synthetic molecules, inhibitors of the enzyme regulation pathway, and genetic engineering using iRNA or CRISPR cas9. Despite most of the applications of the HO inhibitors being related to metabolic diseases, the beneficial effects of these molecules in immune-mediated diseases have also emerged. Different medical implications, including cancer, Alzheimer´s disease, and infections, are discussed in this article and as to how the selective inhibition of HO isoforms may contribute to the treatment of these ailments.


Subject(s)
Enzyme Inhibitors/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Animals , Dioxolanes/metabolism , Dioxolanes/pharmacology , Enzyme Inhibitors/pharmacology , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase-1/antagonists & inhibitors , Humans , Imidazoles/metabolism , Imidazoles/pharmacology , Neoplasms/metabolism , Neoplasms/prevention & control
12.
Microb Pathog ; 129: 64-67, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30716393

ABSTRACT

OBJECTIVES: This study was aimed to develop small molecule inhibitors of the P. aeruginosa heme oxygenase (pa-HemO) as potential treatment of infections caused by P. aeruginosa. METHODS: New compounds were designed based on the crystal structure of pa-HemO. The binding affinities (KD) were determined using intrinsic fluorescence quenching assays. The anti-microbial effects of the new compounds was evaluated by minimal inhibitory concentration 50% (MIC50). RESULTS: Eleven compounds were synthesized as potential pa-HemO inhibitors. New compounds demonstrated KD values ranging from 1.5 to 180 µM, and MIC50 values ranging from 26 to 260 µg/mL. The compounds had good affinity with HemO and promising anti-microbial effects on P. aeruginosa. CONCLUSIONS: The new inhibitors described herein can inhibit the growth of P. aeruginosa via the inhibition of pa-HemO. There may be broad prospects for HemO inhibitors to treat P. aeruginosa related infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Heme Oxygenase (Decyclizing)/chemistry , Heme Oxygenase (Decyclizing)/metabolism , Microbial Sensitivity Tests , Molecular Docking Simulation , Protein Binding , Protein Conformation , Pseudomonas aeruginosa/enzymology
13.
Arch Biochem Biophys ; 671: 130-142, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31276659

ABSTRACT

Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. The enzyme exerts multiple cytoprotective functions associated with the promotion of angiogenesis and counteraction of the detrimental effects of cellular stress which are crucial for the survival of both normal and tumor cells. Accordingly, in many tumor types, high expression of HO-1 correlates with poor prognosis and resistance to treatment, i.e. chemotherapy, suggesting inhibition of HO-1 as a possible antitumor approach. At the same time, the lack of selective and well-profiled inhibitors of HO-1 determines the unmet need for new modulators of this enzyme, with the potential to be used in either adjuvant therapy or as the stand-alone targeted therapeutics. In the current study, we provided novel inhibitors of HO-1 and validated the effect of pharmacological inhibition of HO activity by the imidazole-based inhibitor (SLV-11199) in human pancreatic (PANC-1) and prostate (DU-145) cancer cell lines. We demonstrated potent inhibition of HO activity in vitro and showed associated anticancer effectiveness of SLV-11199. Treatment with the tested compound led to decreased cancer cell viability and clonogenic potential. It has also sensitized the cancer cells to chemotherapy. In PANC-1 cells, diminished HO activity resulted in down-regulation of pro-angiogenic factors like IL-8. Mechanistic investigations revealed that the treatment with SLV-11199 decreased cell migration and inhibited MMP-1 and MMP-9 expression. Moreover, it affected mesenchymal phenotype by regulating key modulators of the epithelial to mesenchymal transition (EMT) signalling axis. Finally, F-actin cytoskeleton and focal contacts were destabilized by the reported compound. Overall, the current study suggests a possible relevance of the tested novel inhibitor of HO activity as a potential anticancer compound. To support such utility, further investigation is still needed, especially in in vivo conditions.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase-1/antagonists & inhibitors , Imidazoles/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans
14.
Int J Mol Sci ; 20(13)2019 Jun 29.
Article in English | MEDLINE | ID: mdl-31261892

ABSTRACT

BACKGROUND: Angiotensin II (Ang II), released by the renin-angiotensin-aldosterone system (RAAS), contributes to the modulatory role of the RAAS in adipose tissue dysfunction. Investigators have shown that inhibition of AngII improved adipose tissue function and insulin resistance in mice with metabolic syndrome. Heme Oxygenase-1 (HO-1), a potent antioxidant, has been demonstrated to improve oxidative stress and adipocyte phenotype. Molecular effects of high oxidative stress include suppression of sirtuin-1 (SIRT1), which is amenable to redox manipulations. The mechanisms involved, however, in these metabolic effects of the RAAS remain incompletely understood. HYPOTHESIS: We hypothesize that AngII-induced oxidative stress has the potential to suppress adipocyte SIRT1 via down regulation of HO-1. This effect of AngII will, in turn, upregulate mineralocorticoid receptor (MR). The induction of HO-1 will rescue SIRT1, hence improving oxidative stress and adipocyte phenotype. METHODS AND RESULTS: We examined the effect of AngII on lipid accumulation, oxidative stress, and inflammatory cytokines in mouse pre-adipocytes in the presence and absence of cobalt protoporphyrin (CoPP), HO-1 inducer, tin mesoporphyrin (SnMP), and HO-1 inhibitor. Our results show that treatment of mouse pre-adipocytes with AngII increased lipid accumulation, superoxide levels, inflammatory cytokine levels, interleukin-6 (IL-6) and tumor necrosis factor α (TNFα), and adiponectin levels. This effect was attenuated by HO-1 induction, which was further reversed by SnMP, suggesting HO-1 mediated improvement in adipocyte phenotype. AngII-treated pre-adipocytes also showed upregulated levels of MR and suppressed SIRT1 that was rescued by HO-1. Subsequent treatment with CoPP and SIRT1 siRNA in mouse pre-adipocytes increased lipid accumulation and fatty acid synthase (FAS) levels, suggesting that beneficial effects of HO-1 are mediated via SIRT1. CONCLUSION: Our study demonstrates for the first time that HO-1 has the ability to restore cellular redox, rescue SIRT1, and prevent AngII-induced impaired effects on adipocytes and the systemic metabolic profile.


Subject(s)
Adipocytes/metabolism , Angiotensin II/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Oxidative Stress , Sirtuin 1/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Cytokines/metabolism , Fatty Acid Synthases/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Lipid Metabolism , Mice , Receptors, Mineralocorticoid/metabolism
15.
Nephrol Dial Transplant ; 33(11): 1927-1941, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29522116

ABSTRACT

Background: P21, a cyclin kinase inhibitor, is upregulated by renal 'ischemic preconditioning' (IPC), and induces a 'cytoresistant' state. However, P21-induced cell cycle inhibition can also contribute to cellular senescence, a potential adverse renal event. Hence, this study assessed whether: (i) IPC-induced P21 upregulation is associated with subsequent renal senescence; and (ii) preconditioning can be established 'independent' of P21 induction and avoid a post-ischemic senescent state? Methods: CD-1 mice were subjected to either IPC (5-15 min) or to a recently proposed 'oxidant-induced preconditioning' (OIP) strategy (tin protoporphyrin-induced heme oxygenase inhibition +/- parental iron administration). P21 induction [messenger RNA (mRNA)/protein], cell proliferation (KI-67, phosphohistone H3 nuclear staining), kidney senescence (P16ink4a; P19Arf mRNAs; senescence-associated beta-galactosidase levels) and resistance to ischemic acute kidney injury were assessed. Results: IPC induced dramatic (10-25×) and persistent P21 activation and 'downstream' tubular senescence. Conversely, OIP did not upregulate P21, it increased, rather than decreased, cell proliferation markers, and it avoided a senescence state. OIP markedly suppressed ischemia-induced P21 up-regulation, it inhibited the development of post-ischemic senescence and it conferred near-complete protection against ischemic acute renal failure (ARF). To assess OIP's impact on a non-P21-dependent cytoprotective pathway, its ability to activate Nrf2, the so-called 'master regulator' of endogenous cell defenses, was assessed. Within 4 h, OIP activated each of three canonical Nrf2-regulated genes (NQO1, SRXN1, GCLC; 3- to 5-fold mRNA increases). Conversely, this gene activation pathway was absent in Nrf2-/- mice, confirming Nrf2 specificity. Nrf2-/- mice also did not develop significant OIP-mediated protection against ischemic ARF. Conclusions: OIP (i) activates the cytoprotective Nrf2, but not the P21, pathway; (ii) suppresses post-ischemic P21 induction and renal senescence; and (iii) confers marked protection against ischemic ARF. In sum, these findings suggest that OIP may be a clinically feasible approach for safely activating the Nrf2 pathway, and thereby confer protection against clinical renal injury.


Subject(s)
Acute Kidney Injury/prevention & control , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation/drug effects , Ischemic Preconditioning/methods , NF-E2-Related Factor 2/physiology , Oxidants/pharmacology , Animals , Cell Cycle Checkpoints/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Male , Mice , Mice, Knockout , Protoporphyrins/pharmacology
16.
Bioorg Chem ; 81: 334-339, 2018 12.
Article in English | MEDLINE | ID: mdl-30189413

ABSTRACT

HO-1 inhibition is considered a valuable anticancer approach. In fact, up-regulation of HO-1 had been repeatedly reported in many types of human malignancies, and in these clinical cases, poor outcomes are reported. To identify novel HO-1 inhibitors suitable for drug development, a scaffold-hopping strategy calculation was utilized to design novel derivatives. Different parts of the selected molecule were analyzed and the different series of novel compounds were virtually evaluated. The calculation for the linker moiety of the classical HO-1 inhibitors structure led us to compounds 5 and 6. A synthetic pathway for the two molecules was designed and the compounds were synthesized. The biological activity revealed an HO-1 inhibition of 0.9 and 54 µM for molecules 5 and 6 respectively. This study suggested that our scaffold-hopping approach was successful and these results are ongoing for further development.


Subject(s)
Enzyme Inhibitors/pharmacology , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Bilirubin/metabolism , Computer Simulation , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Liver/enzymology , Male , Models, Molecular , Molecular Structure , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Quantitative Structure-Activity Relationship , Rats, Sprague-Dawley , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Spleen/enzymology
17.
Microvasc Res ; 111: 12-19, 2017 05.
Article in English | MEDLINE | ID: mdl-28025064

ABSTRACT

OBJECTIVE: We explored the effects of Nuclear Factor-E2-related factor 2 (Nrf2) and Heme Oxygenase 1 (HO-1) on splanchnic hemodynamics in portal hypertensive rats. METHODS: Experimental cirrhosis with portal hypertension was induced by intraperitoneal injection of carbon tetrachloride. The expression of proteins was examined by immunoblotting. Hemodynamic studies were performed by radioactive microspheres. The vascular perfusion system was used to measure the contractile response of mesentery arterioles in rats. RESULTS: Nrf2 expression in the nucleus and HO-1 expression in cytoplasm was significantly enhanced in portal hypertensive rats. Portal pressure, as well as regional blood flow, increased significantly in portal hypertension and can be blocked by tin protoporphyrin IX. The expression of endogenous nitric oxide synthase and vascular endothelial growth factors increased significantly compared to normal rats, while HO-1 inhibition decreased the expression of these proteins significantly. The contractile response of mesenteric arteries decreased in portal hypertension, but can be partially recovered through tin protoporphyrin IX treatment. CONCLUSIONS: The expression of Nrf2/HO-1 increased in mesenteric arteries of portal hypertensive rats, which was related to oxidative stress. HO-1was involved in increased portal pressure and anomaly splanchnic hemodynamics in portal hypertensive rats.


Subject(s)
Arterioles/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Hemodynamics , Hypertension, Portal/enzymology , Liver Cirrhosis, Experimental/enzymology , Mesentery/blood supply , NF-E2-Related Factor 2/metabolism , Splanchnic Circulation , Animals , Arterioles/drug effects , Arterioles/physiopathology , Carbon Tetrachloride , Enzyme Inhibitors/pharmacology , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Hemodynamics/drug effects , Hypertension, Portal/chemically induced , Hypertension, Portal/drug therapy , Hypertension, Portal/physiopathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/physiopathology , Male , Metalloporphyrins/pharmacology , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Portal Pressure , Protoporphyrins/pharmacology , Rats, Sprague-Dawley , Signal Transduction , Splanchnic Circulation/drug effects , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , Vasoconstriction
18.
Nitric Oxide ; 65: 60-67, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28257997

ABSTRACT

Carbon monoxide (CO), an end product of heme oxygenase (HO) that is involved in the regulation of vascular tonus, may show a compensatory effect in nitric oxide (NO) deficiency. This study aimed to assess the effect of the HO/CO system on the vascular tone in exercise-trained rats with hypertension induced by chronic NO synthase (NOS) inhibition. Hypertension was induced by N-nitro-l-arginine methyl ester (25 mg/kg/day in drinking water), and exercise training comprised swimming 1 h/day, 5 days/week, for 6 weeks. Systolic blood pressure (BP) was measured weekly using a tail-cuff method. The effects of hypertension and/or exercise-training on the constriction and relaxation responses of the thoracic aorta and resistance arteries of the mesenteric and gastrocnemius vascular beds were evaluated. NOS inhibition produced a gradually developed hypertension, and the magnitude of the increase in BP was significantly attenuated by exercise training. Although phenylephrine (Phe)-induced contraction responses of aorta incubated with an HO-1 inhibitor were reduced in hypertensive animals, there was no difference in the hypertensive-exercise group. However, thoracic aortas in the hypertensive-exercise group exhibited significantly more relaxation in response to a CO donor. There was no change in Phe-induced contraction with or without HO inhibition CO donor relaxation responses in both resistance arteries. These results suggest that the HO/CO system does not contribute to diminishing BP by exercise training in a NOS inhibition-induced hypertension model.


Subject(s)
Carbon Monoxide/metabolism , Muscle Tonus/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Physical Conditioning, Animal/physiology , Animals , Aorta, Thoracic/physiopathology , Blood Pressure/drug effects , Citrate (si)-Synthase/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/metabolism , Hypertension/chemically induced , Hypertension/physiopathology , Male , Mesenteric Arteries/physiopathology , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle Tonus/physiology , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , NG-Nitroarginine Methyl Ester/pharmacology , Rats, Wistar , Vascular Resistance
19.
J Cardiovasc Pharmacol ; 69(5): 286-297, 2017 May.
Article in English | MEDLINE | ID: mdl-28195947

ABSTRACT

Alpha-melanocyte-stimulating hormone (α-MSH) is a protein with known capacity for protection against cardiovascular ischemia-reperfusion (I/R) injury. This investigation evaluates the capacity of α-MSH to mitigate I/R effects in an isolated working rat heart model and determine the dependency of these alterations on the activity of heme oxygenase-1 (HO-1, hsp-32), a heat shock protein that functions as a major antioxidant defense molecule. Healthy male Sprague Dawley rats were used for all experiments. After treatment with selected doses of α-MSH, echocardiographic examinations were performed on live, anesthetized animals. Hearts were harvested from anesthetized rats pretreated with α-MSH and/or the HO-1 inhibitor SnPP, followed by cardiac function assessment on isolated working hearts, which were prepared using the Langendorff protocol. Induction of global ischemia was performed, followed by during reperfusion assessment of cardiac functions. Determination of incidence of cardiac arrhythmias was made by electrocardiogram. Major outcomes include echocardiographic data, suggesting that α-MSH has mild effects on systolic parameters, along with potent antiarrhythmic effects. Of particular significance was the specificity of dilatative effects on coronary vasculature, and similar outcomes of aortic ring experiments, which potentially allow different doses of the compound to be used to selectively target various portions of the vasculature for dilation.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/prevention & control , Heme Oxygenase (Decyclizing)/metabolism , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , alpha-MSH/pharmacology , Animals , Arrhythmias, Cardiac/enzymology , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Cytoprotection , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Isolated Heart Preparation , Male , Metalloporphyrins/pharmacology , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Protoporphyrins/pharmacology , Rats, Sprague-Dawley , Signal Transduction/drug effects
20.
Nature ; 477(7363): 225-8, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849978

ABSTRACT

Fumarate hydratase (FH) is an enzyme of the tricarboxylic acid cycle (TCA cycle) that catalyses the hydration of fumarate into malate. Germline mutations of FH are responsible for hereditary leiomyomatosis and renal-cell cancer (HLRCC). It has previously been demonstrated that the absence of FH leads to the accumulation of fumarate, which activates hypoxia-inducible factors (HIFs) at normal oxygen tensions. However, so far no mechanism that explains the ability of cells to survive without a functional TCA cycle has been provided. Here we use newly characterized genetically modified kidney mouse cells in which Fh1 has been deleted, and apply a newly developed computer model of the metabolism of these cells to predict and experimentally validate a linear metabolic pathway beginning with glutamine uptake and ending with bilirubin excretion from Fh1-deficient cells. This pathway, which involves the biosynthesis and degradation of haem, enables Fh1-deficient cells to use the accumulated TCA cycle metabolites and permits partial mitochondrial NADH production. We predicted and confirmed that targeting this pathway would render Fh1-deficient cells non-viable, while sparing wild-type Fh1-containing cells. This work goes beyond identifying a metabolic pathway that is induced in Fh1-deficient cells to demonstrate that inhibition of haem oxygenation is synthetically lethal when combined with Fh1 deficiency, providing a new potential target for treating HLRCC patients.


Subject(s)
Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Genes, Lethal/genetics , Genes, Tumor Suppressor , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Mutation/genetics , Animals , Bilirubin/metabolism , Cell Line , Cells, Cultured , Citric Acid Cycle , Computer Simulation , Fumarate Hydratase/deficiency , Fumarates/metabolism , Glutamine/metabolism , Heme/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Leiomyomatosis/congenital , Leiomyomatosis/drug therapy , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , Mice , Mitochondria/metabolism , NAD/metabolism , Neoplastic Syndromes, Hereditary , Skin Neoplasms , Uterine Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL