Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters








Publication year range
1.
Antioxidants (Basel) ; 12(2)2023 Jan 22.
Article in English | MEDLINE | ID: mdl-36829809

ABSTRACT

Besides the clinically proven superior antimalarial activity, artemisinins (ARTs) are also associated with anticancer properties, albeit at much lower potency. Iron and heme have been proposed as possible activators of ARTs against cancer cells. Here we show that zinc protoporphyrin-9 (ZnPPIX), a heme homolog and a natural metabolite for heme synthesis during iron insufficiency, greatly enhanced the anticancer activity of dihydroartemisinin (DHA) in multiple cell lines. Using melanoma B16 and breast cancer 4T1 cells, we demonstrated ZnPPIX dramatically elevated intracellular free heme levels, accompanied by heightened reactive oxidative species (ROS) production. The tumor-suppression activity of ZnPPIX and DHA is mitigated by antioxidant vitamin E or membrane oxidation protectant ferrostatin. In vivo xenograft animal models confirmed that ZnPPIX significantly potentiated the tumor-inhibition capability of DHA while posing no apparent toxicity to the mice. The proliferating index and growth of tumors after the combinatory treatment of DHA and ZnPPIX were evidently reduced. Considering the clinical safety profiles of both DHA and ZnPPIX, their action synergy offers a promising strategy to improve the application of ARTs in our fight against cancer.

2.
Front Immunol ; 13: 879158, 2022.
Article in English | MEDLINE | ID: mdl-35619717

ABSTRACT

The enzyme heme oxygenase-1 (HO-1) has cytoprotective effects by catalyzing the degradation of heme to produce carbon monoxide, iron and biliverdin. Furthermore, HO-1 activity has been associated with successful pregnancy. On the other hand, in the context of certain inflammatory conditions, HO-1 can induce iron overload and cell death. To investigate the role of HO-1 in gestational malaria, pregnant BALB/c mice were infected with Plasmodium berghei ANKA in early, mid and late gestation. We found that malaria affected the pregnancy outcome in the three periods evaluated. However, only poor pregnancy outcomes in early pregnancy were related to HO-1 upregulation, iron overload, lipid peroxidation and necrosis of the decidua, which were prevented by HO-1 inhibition. In conclusion, HO-1 expression must be finely tuned in gestational malaria to avoid the deleterious effect of increased enzyme activity.


Subject(s)
Heme Oxygenase-1 , Malaria , Pregnancy Outcome , Protoporphyrins , Animals , Female , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/metabolism , Iron Overload , Lipid Peroxidation , Malaria/drug therapy , Mice , Plasmodium berghei , Pregnancy , Pregnancy Complications, Parasitic/drug therapy , Protoporphyrins/pharmacology
3.
Mol Cell Proteomics ; 19(12): 1968-1986, 2020 12.
Article in English | MEDLINE | ID: mdl-32912968

ABSTRACT

Mallory-Denk-bodies (MDBs) are hepatic protein aggregates associated with inflammation both clinically and in MDB-inducing models. Similar protein aggregation in neurodegenerative diseases also triggers inflammation and NF-κB activation. However, the precise mechanism that links protein aggregation to NF-κB-activation and inflammatory response remains unclear. Herein we find that treating primary hepatocytes with MDB-inducing agents (N-methylprotoporphyrin (NMPP), protoporphyrin IX (PPIX), or Zinc-protoporphyrin IX (ZnPP)) elicited an IκBα-loss with consequent NF-κB activation. Four known mechanisms of IκBα-loss i.e. the canonical ubiquitin-dependent proteasomal degradation (UPD), autophagic-lysosomal degradation, calpain degradation and translational inhibition, were all probed and excluded. Immunofluorescence analyses of ZnPP-treated cells coupled with 8 M urea/CHAPS-extraction revealed that this IκBα-loss was due to its sequestration along with IκBß into insoluble aggregates, thereby releasing NF-κB. Through affinity pulldown, proximity biotinylation by antibody recognition, and other proteomic analyses, we verified that NF-κB subunit p65, which stably interacts with IκBα under normal conditions, no longer binds to it upon ZnPP-treatment. Additionally, we identified 10 proteins that interact with IκBα under baseline conditions, aggregate upon ZnPP-treatment, and maintain the interaction with IκBα after ZnPP-treatment, either by cosequestering into insoluble aggregates or through a different mechanism. Of these 10 proteins, the nucleoporins Nup153 and Nup358/RanBP2 were identified through RNA-interference, as mediators of IκBα-nuclear import. The concurrent aggregation of IκBα, NUP153, and RanBP2 upon ZnPP-treatment, synergistically precluded the nuclear entry of IκBα and its consequent binding and termination of NF-κB activation. This novel mechanism may account for the protein aggregate-induced inflammation observed in liver diseases, thus identifying novel targets for therapeutic intervention. Because of inherent commonalities this MDB cell model is a bona fide protoporphyric model, making these findings equally relevant to the liver inflammation associated with clinical protoporphyria.


Subject(s)
I-kappa B Proteins/metabolism , Inflammation/pathology , Liver/metabolism , Liver/pathology , NF-kappa B/metabolism , Protein Aggregates , Active Transport, Cell Nucleus/drug effects , Animals , Autophagy/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , HEK293 Cells , HeLa Cells , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Nuclear Pore Complex Proteins/metabolism , Protein Aggregates/drug effects , Protein Binding/drug effects , Protein Multimerization/drug effects , Protoporphyrins/pharmacology , RNA, Small Interfering/metabolism , Sequestosome-1 Protein/metabolism , Solubility
4.
Can J Physiol Pharmacol ; 97(11): 1073-1079, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31536393

ABSTRACT

This study aimed to investigate the effects of propofol through evaluating its interaction with nitric oxide (NO), hydrogen sulfide (H2S), and carbon monoxide (CO). Wistar male rats were divided in 4 groups: (1) bolus injection of propofol (1% 10 mg/mL, 100 mg/kg bw, i.p.); (2) Nω-nitro-l-arginine methyl ester (L-NAME; NO synthase inhibitor, 60 mg/kg bw, i.p.) + bolus injection of propofol (1% 10 mg/mL, 100 mg/kg bw, i.p.); (3) DL-propargylglycine (DL-PAG; H2S synthase inhibitor, 50 mg/kg bw, i.p.) + bolus injection of propofol (1% 10 mg/mL, 100 mg/kg bw, i.p.); (4) zinc protoporphyrin IX (ZnPPIX; CO synthase inhibitor, 50 µmol/kg bw, i.p.) + bolus injection of propofol (1% 10 mg/mL, 100 mg/kg bw, i.p.). Increased levels of albumins, low-density lipoproteins, alkaline phosphatase, amylase, high-sensitivity Troponin T, and fibrinogen were found in L-NAME + propofol group. Platelet crit, platelet count, total cholesterol, and high-density lipoproteins were elevated in ZnPPIX + propofol group. Hydrogen peroxide was increased in all groups treated with gasotransmitters inhibitors. Reduced glutathione was reduced in all groups, superoxide dismutase activity only in L-NAME + propofol. The effect of propofol on various biochemical, haematological, and oxidative stress markers may be at least in part mediated through interaction with 3 estimated gasotransmitters.


Subject(s)
Anesthetics/pharmacology , Gasotransmitters/antagonists & inhibitors , Hematologic Tests , Oxidative Stress/drug effects , Propofol/pharmacology , Animals , Biomarkers/blood , Hemostasis/drug effects , Homocysteine/blood , Male , Rats , Rats, Wistar
5.
Can J Physiol Pharmacol ; 97(9): 850-856, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31116954

ABSTRACT

It has been assumed that the cardioprotective effects of propofol are due to its non-anesthetic pleiotropic cardiac and vasodilator effects, in which gasotransmitters (NO, H2S, and CO) as well as calcium influx could be involved. The study on isolated rat heart was performed using 4 experimental groups (n = 7 in each): (1) bolus injection of propofol (100 mg/kg body mass, i.p.); (2) L-NAME (NO synthase inhibitor, 60 mg/kg body mass, i.p.) + propofol; (3) DL-PAG (H2S synthase inhibitor, 50 mg/kg body mass, i.p.) + propofol; (4) ZnPPIX (CO synthase inhibitor, 50 µmol/kg body mass, i.p.) + propofol. Before and after the verapamil (3 µmol/L) administration, cardiodynamic parameters were recorded (dp/dtmax, dp/dtmin, systolic left ventricular pressure, diastolic left ventricular pressure, heart rate, coronary flow), as well as coronary and cardiac oxidative stress parameters. The results showed significant increases of diastolic left ventricular pressure following NO and CO inhibition, but also increases of coronary flow following H2S and CO inhibition. Following verapamil administration, significant decreases of dp/dtmax were noted after NO and CO inhibition, then increase of diastolic left ventricular pressure following CO inhibition, and increase of coronary flow following NO, H2S, or CO inhibition. Oxidative stress markers were increased but catalase activity was significantly decreased in cardiac tissue. Gasotransmitters and calcium influx are involved in pleiotropic cardiovascular effects of propofol in male Wistar rats.


Subject(s)
Anesthetics/pharmacology , Calcium/metabolism , Gasotransmitters/biosynthesis , Heart/drug effects , Myocardium/metabolism , Oxidative Stress/drug effects , Propofol/pharmacology , Animals , Biological Transport/drug effects , Cardiotonic Agents/pharmacology , Heart/physiology , Male , Rats , Rats, Wistar
6.
Int J Mol Sci ; 19(10)2018 Oct 10.
Article in English | MEDLINE | ID: mdl-30308949

ABSTRACT

Two main isoforms of the Translocator Protein (TSPO) have been identified. TSPO1 is ubiquitous and is mainly present at the outer mitochondrial membrane of most eukaryotic cells, whereas, TSPO2 is specific to the erythroid lineage, located at the plasma membrane, the nucleus, and the endoplasmic reticulum. The design of specific tools is necessary to determine the molecular associations and functions of TSPO, which remain controversial nowadays. We recently demonstrated that TSPO2 is involved in a supramolecular complex of the erythrocyte membrane, where micromolar doses of the classical TSPO ligands induce ATP release and zinc protoporphyrin (ZnPPIX) transport. In this work, three newly-designed ligands (NCS1016, NCS1018, and NCS1026) were assessed for their ability to modulate the functions of various erythrocyte's and compare them to the TSPO classical ligands. The three new ligands were effective in reducing intraerythrocytic Plasmodium growth, without compromising erythrocyte survival. While NCS1016 and NCS1018 were the most effective ligands in delaying sorbitol-induced hemolysis, NCS1016 induced the highest uptake of ZnPPIX and NCS1026 was the only ligand inhibiting the cholesterol uptake. Differential effects of ligands are probably due, not only, to ligand features, but also to the dynamic interaction of TSPO with various partners at the cell membrane. Further studies are necessary to fully understand the mechanisms of the TSPO's complex activation.


Subject(s)
Adenosine Triphosphate/metabolism , Cholesterol/metabolism , Erythrocytes/metabolism , Protoporphyrins/metabolism , Receptors, GABA/metabolism , Biological Transport , Hemolysis , Humans , Ligands , Plasmodium falciparum/drug effects , Protein Binding , Reactive Oxygen Species , Sorbitol/pharmacology
7.
Sensors (Basel) ; 18(6)2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29857594

ABSTRACT

A label-free sensing platform is developed based on switching the structure of aptamer for highly sensitive and selective fluorescence detection of ochratoxin A (OTA). OTA induces the structure of aptamer, transforms into G-quadruplex and produces strong fluorescence in the presence of zinc(II)-protoporphyrin IX probe due to the specific bind to G-quadruplex. The simple method exhibits high sensitivity towards OTA with a detection limit of 0.03 nM and excellent selectivity over other mycotoxins. In addition, the successful detection of OTA in real samples represents a promising application in food safety.


Subject(s)
Aptamers, Nucleotide/chemistry , Biosensing Techniques , Mycotoxins/isolation & purification , Ochratoxins/isolation & purification , Fluorescent Dyes/chemistry , Food Contamination/analysis , Humans , Mycotoxins/chemistry , Mycotoxins/toxicity , Ochratoxins/toxicity
8.
Brain Inj ; 30(1): 36-42, 2016.
Article in English | MEDLINE | ID: mdl-26595800

ABSTRACT

PRIMARY OBJECTIVE: The purpose of this study was to investigate the effects of heme oxygenase-1 (HO-1) on astrocyte injury induced by hemin. RESEARCH DESIGN: Primary astrocytes were isolated from Sprague Dawley rat pups and cultured in vitro. The expression of HO-1 was induced by hemin in a quantitative fashion and the effects of HO-1 on hemin-induced astrocyte injury were estimated by cell viability, cell membrane permeability and apoptosis. METHODS AND PROCEDURES: Astrocytes were divided into control group, hemin 5 µM group, hemin 5 µM + Zn-PPIX group, hemin 30 µM group and hemin 30 µM + Zn-PPIX group. Survival quality of astrocyte was measured by WST-8 assay, LDH assay, Hoechst 33258 Staining and annexin V-FITC/PI assay and apoptotic-related proteins were measured using Western blotting. MAIN OUTCOME AND RESULTS: Hemin could dose-dependently up-regulate the expression of HO-1. HO-1 exerted a protective role on astrocyte damage induced by 5 µM hemin, including increased cell survival rate and anti-apoptotic proteins expression (Bcl-2 and p-AKT), as well as decreased LDH release, apoptosis ratio and apoptotic protein expression (Bax, p-ERK and cleaved-caspase3). However, the effect of HO-1 on astrocyte injury between 30 µM hemin-treated groups was opposite of the protective role in 5 µM hemin-treated groups. CONCLUSIONS: There were dual effects of HO-1 in 5 µM and 30 µM hemin-induced astrocyte injuries.


Subject(s)
Astrocytes/drug effects , Astrocytes/enzymology , Heme Oxygenase-1/biosynthesis , Hemin/pharmacology , Animals , Apoptosis/drug effects , Astrocytes/cytology , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Heme Oxygenase-1/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
9.
Biomed Pharmacother ; 71: 240-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25960243

ABSTRACT

Our previous study suggests that heme oxygenase-1 (HO-1) may play an important role in the metastasis of gastric cancer. Zinc protoporphyrin IX (ZnPPIX) is a special HO-1 inhibitor that inhibits the angiogenesis of pancreatic and lung cancer. In this study, we employed ZnPPIX to investigate the role of HO-1 in peritoneal metastasis of gastric cancer (PMGC) and explored the potential mechanism. We established animal model of PMGC by orthotopic implantation into nude mice of human gastric cancer cell line GC9811-P with high peritoneal metastasis potential. The mice were injected intraperitoneally with saline, CTX or ZnPPIX. Tumor microvessel density (MVD) in peritoneal metastatic nodules was determined by immunohistochemistry, and vascular endothelial growth factor (VEGF) level was determined by ELISA. We found that the number, volume, weight of peritoneal metastatic nodules and volume of seroperitoneum in ZnPPIX (4 mg/kg) group decreased remarkably compared with control group. MVD value and VEGF level of peritoneal metastatic tumor in ZnPPIX (4 mg/kg) group also decreased significantly, while the survival rate and survival time of the mice were higher than control group. ZnPPIX dose-dependently suppressed VEGF and GC9811-P induced angiogenesis. Furthermore, ZnPPIX suppressed VEGF induced reactive oxygen species production and ERK phosphorylation in human umbilical vein endothelial cells. In conclusion, our results suggest that HO-1 plays an important role in PMGC and ZnPPIX is an effective antitumor and antiangiogenic agent for PMGC.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/secondary , Protoporphyrins/therapeutic use , Stomach Neoplasms/pathology , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chickens , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Extracellular Signal-Regulated MAP Kinases , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kaplan-Meier Estimate , Mice, Nude , Phosphorylation/drug effects , Protoporphyrins/pharmacology , Reactive Oxygen Species/metabolism , Stomach Neoplasms/drug therapy , Survival Analysis , Vascular Endothelial Growth Factor A/metabolism
10.
Biosens Bioelectron ; 66: 547-53, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25506903

ABSTRACT

Site-specific identification of DNA methylation and assay of MTase activity are imperative for determining specific cancer types, provide insights into the mechanism of gene repression, and develop novel drugs to treat methylation-related diseases. Herein, we developed a highly sensitive fluorescence assay of DNA methyltransferase by methylation-sensitive cleavage-based primer generation exponential isothermal amplification (PG-EXPA) coupled with supramolecular fluorescent Zinc(II)-protoporphyrin IX (ZnPPIX)/G-quadruplex. In the presence of DNA adenine methylation (Dam) MTase, the methylation-responsive sequence of hairpin probe is methylated and cleaved by the methylation-sensitive restriction endonuclease Dpn I. The cleaved hairpin probe then functions as a signal primer to initiate the exponential isothermal amplification reaction (EXPAR) by hybridizing with a unimolecular DNA containing three functional domains as the amplification template, producing a large number of G-quadruplex nanostructures by utilizing polymerases and nicking enzymes as mechanical activators. The G-quadruplex nanostructures act as host for ZnPPIX that lead to supramolecular complexes ZnPPIX/G-quadruplex, which provides optical labels for amplified fluorescence detection of Dam MTase. While in the absence of Dam MTase, neither methylation/cleavage nor PG-EXPA reaction can be initiated and no fluorescence signal is observed. The proposed method exhibits a wide dynamic range from 0.0002 to 20U/mL and an extremely low detection limit of 8.6×10(-5)U/mL, which is superior to most conventional approaches for the MTase assay. Owing to the specific site recognition of MTase toward its substrate, the proposed sensing system was able to readily discriminate Dam MTase from other MTase such as M.SssI and even detect the target in a complex biological matrix. Furthermore, the application of the proposed sensing strategy for screening Dam MTase inhibitors was also demonstrated with satisfactory results. This novel method not only provides a promising platform for monitoring activity and inhibition of DNA MTases, but also shows great potentials in biological process researches, drugs discovery and clinical diagnostics.


Subject(s)
Biosensing Techniques/methods , DNA Methylation , DNA Modification Methylases/analysis , G-Quadruplexes , Biosensing Techniques/statistics & numerical data , DNA Primers , Deoxyribonucleases, Type II Site-Specific/analysis , Deoxyribonucleases, Type II Site-Specific/antagonists & inhibitors , Deoxyribonucleases, Type II Site-Specific/metabolism , Fluorescence , Humans , Nucleic Acid Amplification Techniques/methods , Protoporphyrins , Site-Specific DNA-Methyltransferase (Adenine-Specific)/analysis , Site-Specific DNA-Methyltransferase (Adenine-Specific)/antagonists & inhibitors , Site-Specific DNA-Methyltransferase (Adenine-Specific)/blood
11.
Biosens Bioelectron ; 61: 351-6, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-24912035

ABSTRACT

A cascadic sensing system was developed for detection of DNA target at ultralow concentration by a combination of magnetic nanoparticles (MNPs) and exonuclease III (Exo III)-induced cascade two-stage isothermal amplification in the study. An ingeniously designed capture hairpin probe (CHP) that integrates target-binding and signal transduction sequences within one multifunctional design was assembled on MNPs. Upon sensing of the analyte nucleic acid, the hairpin probe on MNPs could be opened and stepwise removed by Exo III accompanied by the releasing of target DNA for the successive hybridization and cleavage process and the generation of bare signal transduction sequences of CHP as a new trigger for next circular reaction. The new DNA triggers initiate hybridizing with hairpin DNA probe that contains a partially "caged" G-quadruplex sequence (GHP), forming a duplex structure and liberating the active G-quadruplex structure. Then, Exo III digests the resulting duplex domain, leading to the recycling of new DNA trigger and simultaneously generating numerous ZnPPIX/G-quadruplex supramolecular complexes with the help of the zinc (II)-protoporphyrin IX (ZnPPIX), as an optical label for amplified fluorescence sensing event. Finally, numerous liberated cascade ZnPPIX/G-quadruplex supramolecular complexes give a remarkable fluorescence response. Because of two-stage autocatalytic recycling amplification and the specifically catalyzed formation of ZnPPIX/G-quadruplex supramolecular complexes, this newly designed protocol provides a high sensitivity with a detection limit of 0.75 fM, can discriminate mismatched DNA from perfectly matched target DNA, and gives low matrix effect due to using MNPs as the separation and amplification elements in the real samples. Therefore, it holds great potential for early diagnosis in gene-related diseases.


Subject(s)
Biosensing Techniques/methods , DNA/analysis , Exodeoxyribonucleases/metabolism , G-Quadruplexes , Nucleic Acid Amplification Techniques/methods , Protoporphyrins/metabolism , DNA/metabolism , Fluorescence , Limit of Detection , Magnetite Nanoparticles/chemistry , Spectrometry, Fluorescence/methods
12.
Biochim Biophys Acta ; 1830(10): 4848-59, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23816986

ABSTRACT

BACKGROUND: Diallyl disulfide (DADS) is a garlic-derived organosulfur compound. The current study is designed to evaluate the protective effects of DADS against ethanol-induced oxidative stress, and to explore the underlying mechanisms by examining the HO-1/Nrf-2 pathway. METHODS: We investigated whether or not DADS could activate the HO-1 in normal human liver cell LO2, and then evaluated the protective effects of DADS against ethanol-induced damage in LO2 cells and in acute ethanol-intoxicated mice. The biochemical parameters were measured using commercial kits. HO-1 mRNA level was determined by RT-PCR. Histopathology and immunofluorescence assay were performed with routine methods. Protein levels were measured by western blot. RESULTS: DADS significantly increased the mRNA and protein levels of HO-1, stimulated the nuclear translocation of Nrf-2 and increased the phosphorylation of MAPK in LO2 cells. The nuclear translocation of Nrf-2 was abrogated by MAPK inhibitors. DADS significantly suppressed ethanol-induced elevation of lactate dehydrogenase (LDH) and aspartate transaminase (AST) activities, decrease of glutathione (GSH) level, increase of malondialdehyde (MDA) levels, and apoptosis of LO2 cells, which were all blocked by ZnPPIX. In mice, DADS effectively suppressed acute ethanol-induced elevation of aminotransferase activities, and improved liver histopathological changes, which might be associated with HO-1 activation. CONCLUSION: These results demonstrate that DADS could induce the activation of HO-1/Nrf-2 pathway, which may contribute to the protective effects of DADS against ethanol-induced liver injury. GENERAL SIGNIFICANCE: DADS may be beneficial for the prevention and treatment of ALD due to significant activation of HO-1/Nrf-2 pathway.


Subject(s)
Allyl Compounds/pharmacology , Disulfides/pharmacology , Ethanol/toxicity , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Animals , Base Sequence , Cell Line , DNA Primers , Humans , MAP Kinase Signaling System , Male , Mice , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL