Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 309
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochem Biophys Res Commun ; 719: 150043, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38735206

ABSTRACT

In this study, a simple green synthesis of vanadium pentoxide nanoparticles (VNPs) was prepared by the extract of Kaffir lime fruit (Citrus hystrix) as a green reducing and stabilizing agent, along with the investigation of calcination temperature was carried out at 450 and 550 °C. It was affirmed that, at higher temperature (550 °C), the VNPs possessed a high degree crystalline following the construction of (001) lattice diffraction within an increase in crystalline size from 47.12 to 53.51 nm, although the band gap of the materials at 450 °C was lower than that of the VNPs-550 (2.53 versus 2.66 eV, respectively). Besides, the materials were assessed for the potential bioactivities toward antibacterial, antifungal, DNA cleavage, anti-inflammatory, and hemolytic performances. As a result, the antibacterial activity, with minimal inhalation concentration (MIC) < 6.25 µg/mL for both strains, and fungicidal one of the materials depicted the dose-dependent effects. Once, both VNPs exhibited the noticeable efficacy of the DNA microbial damage, meanwhile, the outstanding anti-inflammatory agent was involved with the IC50 of 123.636 and 227.706 µg/mL, accounting for VNPs-450 and VNPs-550, respectively. Furthermore, this study also demonstrated the hemolytic potential of the VNPs materials. These consequences declare the prospects of the VNPs as the smart and alternative material from the green procedure in biomedicine.


Subject(s)
Anti-Bacterial Agents , Citrus , Fruit , Plant Extracts , Vanadium Compounds , Citrus/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Vanadium Compounds/chemistry , Vanadium Compounds/pharmacology , Fruit/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Nanoparticles/chemistry , Microbial Sensitivity Tests , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/chemical synthesis , Temperature , Hemolysis/drug effects , Green Chemistry Technology , Humans
2.
J Mater Sci Mater Med ; 35(1): 42, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073469

ABSTRACT

Studies have shown that the inhibition of phosphatase and tensin homolog deleted on chromosome 10 (PTEN)was neuroprotective against ischemia/reperfusion(I/R) injury. Bisperoxovanadium (bpV), a derivative of vanadate, is a well-established inhibitor of PTEN. However, its function islimited due to its general inadequacy in penetrating cell membranes. Mxene(Ti3C2Tx) is a novel two-dimensional lamellar nanomaterial with an excellent ability to penetrate the cell membrane. Yet, the effects of this nanomaterial on nervous system diseases have yet to be scrutinized. Here, Mxene(Ti3C2Tx) was used for the first time to carry bpV(HOpic), creating a new nanocomposite Mxene-bpV that was probed in a cerebral I/R injury model. The findings showed that this synthetic Mxene-bpV was adequately stable and can cross the cell membraneeasily. We observed that Mxene-bpV treatment significantly increased the survival rate of oxygen glucose deprivation/reperfusion(OGD/R)--insulted neurons, reduced infarct sizes and promoted the recovery of brain function after mice cerebral I/R injury. Crucially, Mxene-bpV treatment was more therapeutically efficient than bpV(HOpic) treatment alone over the same period. Mechanistically, Mxene-bpV inhibited the enzyme activity of PTEN in vitro and in vivo. It also promoted the expression of phospho-Akt (Ser473) by repressing PTEN and then activated the Akt pathway to boost cell survival. Additionally, in PTEN transgenic mice, Mxene-bpV suppressed I/R-induced inflammatory response by promoting M2 microglial polarization through PTEN inhibition. Collectively, the nanosynthetic Mxene-bpV inhibited PTEN' enzymatic activity by activating Akt pathway and promoting M2 microglial polarization, and finally exerted neuroprotection against cerebral I/R injury.


Subject(s)
Microglia , Neuroprotective Agents , PTEN Phosphohydrolase , Proto-Oncogene Proteins c-akt , Reperfusion Injury , Signal Transduction , Vanadium Compounds , Animals , Microglia/drug effects , Microglia/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mice , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Neuroprotective Agents/pharmacology , Vanadium Compounds/pharmacology , Vanadium Compounds/chemistry , PTEN Phosphohydrolase/metabolism , Male , Mice, Inbred C57BL , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Cell Polarity/drug effects , Neurons/drug effects , Neurons/metabolism , Nanocomposites/chemistry
3.
Pak J Pharm Sci ; 37(1): 79-84, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38741403

ABSTRACT

Vanadyl sulfate (VS), is a component of some food supplements and experimental drugs. This study was carried out to present a novel method for induction of Type 2 diabetes in rats, then for the first time in literature, for evaluating the effect of VS on metabolic parameters and gene expression, simultaneously. 40 male wistar rats were distributed between the four groups, equally. High fat diet and fructose were used for diabetes induction. Diabetic rats treated by two different dose of VS for 12 weeks. Metabolic profiles were evaluated by commercial available kits and gene expression were assayed by real time-PCR. Compared to controls, in non-treated diabetic rats, weight, glucose, triglyceride, total cholesterol, insulin and insulin resistance were increased significantly (p-value <0.05) that indicated induction of type 2 diabetes. Further, the results showed that VS significantly reduced weight, insulin secretion, Tumor Necrosis Factor-alpha (TNF-α) genes expression, lipid profiles except HDL that we couldn't find any significant change and increased Peroxisome Proliferator-Activated Receptor- gamma (PPAR-γ) gene expression in VS-treated diabetic animals in comparison with the non-treated diabetics. Our study demonstrated that vanadyl supplementation in diabetic rats had advantageous effects on metabolic profiles and related gene expression.


Subject(s)
Blood Glucose , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , PPAR gamma , Tumor Necrosis Factor-alpha , Vanadium Compounds , Animals , Rats , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Diet, High-Fat/adverse effects , Gene Expression Regulation/drug effects , Hypoglycemic Agents/pharmacology , Insulin/blood , Insulin Resistance , PPAR gamma/drug effects , PPAR gamma/genetics , PPAR gamma/metabolism , Rats, Wistar , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vanadium Compounds/pharmacology
4.
Int J Mol Sci ; 24(21)2023 Oct 27.
Article in English | MEDLINE | ID: mdl-37958659

ABSTRACT

Over the last four decades, vanadium compounds have been extensively studied as potential antidiabetic drugs. With the present review, we aim at presenting a general overview of the most promising compounds and the main results obtained with in vivo studies, reported from 1899-2023. The chemistry of vanadium is explored, discussing the importance of the structure and biochemistry of vanadate and the impact of its similarity with phosphate on the antidiabetic effect. The spectroscopic characterization of vanadium compounds is discussed, particularly magnetic resonance methodologies, emphasizing its relevance for understanding species activity, speciation, and interaction with biological membranes. Finally, the most relevant studies regarding the use of vanadium compounds to treat diabetes are summarized, considering both animal models and human clinical trials. An overview of the main hypotheses explaining the biological activity of these compounds is presented, particularly the most accepted pathway involving vanadium interaction with phosphatase and kinase enzymes involved in the insulin signaling cascade. From our point of view, the major discoveries regarding the pharmacological action of this family of compounds are not yet fully understood. Thus, we still believe that vanadium presents the potential to help in metabolic control and the clinical management of diabetes, either as an insulin-like drug or as an insulin adjuvant. We look forward to the next forty years of research in this field, aiming to discover a vanadium compound with the desired therapeutic properties.


Subject(s)
Diabetes Mellitus , Vanadium Compounds , Animals , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/chemistry , Vanadium Compounds/pharmacology , Vanadium Compounds/therapeutic use , Vanadium Compounds/chemistry , Vanadium/chemistry , Diabetes Mellitus/drug therapy , Insulin/therapeutic use , Insulin, Regular, Human/therapeutic use
5.
J Biol Inorg Chem ; 26(4): 511-531, 2021 06.
Article in English | MEDLINE | ID: mdl-34057639

ABSTRACT

Vanadocene dichloride (VDC), a vanadium containing metallocene dihalide exhibits promising anticancer activity. However, its mechanism of action remains elusive as several diverse targets and pathways have been proposed for its anticancer activity. In this study, we observed that VDC inhibited the proliferation of mammalian cancer cells and induced apoptotic cell death by altering the mitochondrial membrane potential and the expression of bcl2 and bax. Probing further into its anticancer mechanism, we found that VDC caused depolymerization of interphase microtubules and blocked the cells at mitosis with considerable proportion of cells exhibiting monopolar spindles. The reassembly of cold depolymerized microtubules was strongly inhibited in the presence of 10 µM VDC. VDC perturbed the microtubule-kinetochore interactions during mitosis as indicated by the absence of cold stable spindle microtubules in the cells treated with 20 µM VDC. Using goat brain tubulin, we found that VDC inhibited the steady-state polymer mass of microtubules and bound to tubulin at a novel site with a Kd of 9.71 ± 0.19 µM and perturbed the secondary structure of tubulin dimer. In addition, VDC was also found to bind to the mitotic kinesin Eg5 and inhibit its basal as well as microtubule stimulated ATPase activity. The results suggest that disruption of microtubule assembly dynamics and inhibition of the ATPase activity of Eg5 could be a plausible mechanism for the antiproliferative and antimitotic activity of VDC.Graphic abstract.


Subject(s)
Apoptosis/drug effects , Gene Expression Regulation/drug effects , Kinesins/metabolism , Microtubules/drug effects , Vanadium Compounds/pharmacology , Acridine Orange , Cell Proliferation/drug effects , HeLa Cells , Hep G2 Cells , Humans , Kinesins/genetics , Microtubules/metabolism
6.
Inorg Chem ; 60(20): 15291-15309, 2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34597028

ABSTRACT

Five new anionic aqueous dioxidovanadium(V) complexes, [{VO2L1,2}A(H2O)n]α (1-5), with the aroylhydrazone ligands pyridine-4-carboxylic acid (3-ethoxy-2-hydroxybenzylidene)hydrazide (H2L1) and furan-2-carboxylic acid (3-ethoxy-2-hydroxybenzylidene)hydrazide (H2L2) incorporating different alkali metals (A = Na+, K+, Cs+) as countercation were synthesized and characterized by various physicochemical techniques. The solution-phase stabilities of 1-5 were determined by time-dependent NMR and UV-vis, and also the octanol/water partition coefficients were obtained by spectroscopic techniques. X-ray crystallography of 2-4 confirmed the presence of vanadium(V) centers coordinated by two cis-oxido-O atoms and the O, N, and O atoms of a dianionic tridentate ligand. To evaluate the biological behavior, all complexes were screened for their DNA/protein binding propensity through spectroscopic experiments. Finally, a cytotoxicity study of 1-5 was performed against colon (HT-29), breast (MCF-7), and cervical (HeLa) cancer cell lines and a noncancerous NIH-3T3 cell line. The cytotoxicity was cell-selective, being more active against HT-29 than against other cells. In addition, the role of hydrophobicity in the cytotoxicity was explained in that an optimal hydrophobicity is essential for high cytotoxicity. Moreover, the results of wound-healing assays indicated antimigration in case of HT-29 cells. Remarkably, 1 with an IC50 value of 5.42 ± 0.15 µM showed greater activity in comparison to cisplatin against the HT-29 cell line.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , DNA/chemistry , Hydrazones/pharmacology , Serum Albumin, Bovine/chemistry , Vanadium Compounds/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Hydrazones/chemistry , Hydrophobic and Hydrophilic Interactions , Mice , Models, Molecular , Molecular Structure , NIH 3T3 Cells , Solubility , Vanadium Compounds/chemistry , Water/chemistry
7.
Biometals ; 34(1): 161-173, 2021 02.
Article in English | MEDLINE | ID: mdl-33206308

ABSTRACT

Cardiovascular complications are among the leading causes of morbidity and mortality in diabetes mellitus (DM). Despite the anti-hyperglycemic effects of various anti-diabetic therapeutic agents like insulin, some of these drugs are implicated in precipitating cardiovascular dysfunction. There is therefore an imperative need to seek alternative drugs that may ameliorate these complications. Accordingly, the aim of the study was to investigate the effects of a dioxidovanadium (V) complex, cis-[VO2(obz)py]) on selected cardiovascular function markers in STZ-induced diabetic rats. The vanadium complex (40 mg kg) was administered orally twice every 3rd day 5 weeks, non-diabetic and diabetic control groups received distilled water whereas the insulin group received subcutaneous insulin injections twice daily for 5 weeks. Blood glucose concentrations, mean arterial pressure (MAP), heart rate, triglycerides (TG) and total cholesterol concentrations were monitored weekly for 5 weeks. Rats were then euthanised and blood and hearts were collected for biochemical analysis. There was a significant decrease in blood glucose, triglycerides, cholesterol concentrations as well as blood pressure of vanadium treated rats compared to the untreated diabetic animals. Vanadium treatment also attenuated cardiac oxidative stress and decreased the expression of transforming growth factor ß1 (TGFß1) and Smad7. Lastly, the administration of the vanadium complex significantly decreased C reactive protein (CRP) and cardiotropin 1(CT-1) concentrations in the plasma and heart tissues. The administration of the dioxidovanadium(V) complex to diabetic rats culminated into cardio-protective effects. Taken together, these observations suggest that this metal complex exhibit a significant potential as an alternative therapeutic drug for DM management.


Subject(s)
Coordination Complexes/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/pharmacology , Protective Agents/pharmacology , Vanadium Compounds/pharmacology , Animals , Coordination Complexes/chemistry , Diabetes Mellitus, Experimental/chemically induced , Dose-Response Relationship, Drug , Hypoglycemic Agents/chemistry , Male , Models, Molecular , Protective Agents/chemistry , Rats , Rats, Sprague-Dawley , Streptozocin , Vanadium Compounds/chemistry
8.
Molecules ; 26(18)2021 Sep 12.
Article in English | MEDLINE | ID: mdl-34577005

ABSTRACT

Vanadium has a good therapeutic potential, as several biological effects, but few side effects, have been demonstrated. Evidence suggests that vanadium compounds could represent a new class of non-platinum, metal antitumor agents. In the present study, we aimed to characterize the antiproliferative activities of fluorescent vanadyl complexes with acetylacetonate derivates bearing asymmetric substitutions on the ß-dicarbonyl moiety on different cell lines. The effects of fluorescent vanadyl complexes on proliferation and cell cycle modulation in different cell lines were detected by ATP content using the CellTiter-Glo Luminescent Assay and flow cytometry, respectively. Western blotting was performed to assess the modulation of mitogen-activated protein kinases (MAPKs) and relevant proteins. Confocal microscopy revealed that complexes were mainly localized in the cytoplasm, with a diffuse distribution, as in podocyte or a more aggregate conformation, as in the other cell lines. The effects of complexes on cell cycle were studied by cytofluorimetry and Western blot analysis, suggesting that the inhibition of proliferation could be correlated with a block in the G2/M phase of cell cycle and an increase in cdc2 phosphorylation. Complexes modulated mitogen-activated protein kinases (MAPKs) activation in a cell-dependent manner, but MAPK modulation can only partly explain the antiproliferative activity of these complexes. All together our results demonstrate that antiproliferative effects mediated by these compounds are cell type-dependent and involve the cdc2 and MAPKs pathway.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Hydroxybutyrates/chemistry , Pentanones/chemistry , Vanadium Compounds/chemistry , Vanadium Compounds/pharmacology , Biological Transport , CDC2 Protein Kinase/metabolism , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Fluorescent Dyes , Humans , Inhibitory Concentration 50 , Microscopy, Confocal , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Podocytes/drug effects , Podocytes/ultrastructure , Protein Kinase Inhibitors/pharmacology
9.
Histochem Cell Biol ; 154(3): 287-299, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32495040

ABSTRACT

In mammals, progressive activation of primordial follicles is essential for maintenance of the reproductive lifespan. Several reports have demonstrated that mitogen-activated protein kinases 3 and 1 (MAPK3/1)-mammalian target of rapamycin complex 1 (mTORC1) signaling in pre-granulosa cells promotes primordial follicle activation by increasing KIT ligand (KITL) expression and then stimulating phosphatidylinositol 3 kinase signaling in oocytes. However, the mechanism of mTORC1 signaling in the promotion of KITL expression is unclear. Immunofluorescence staining results showed that phosphorylated cyclic AMP response element-binding protein (CREB) was mainly expressed in pre-granulosa cells. The CREB inhibitor KG-501 and CREB knockdown by Creb siRNA significantly suppressed primordial follicle activation, reduced pre-granulosa cell proliferation and dramatically increased oocyte apoptosis. Western blotting results demonstrated that both the MAPK3/1 inhibitor U0126 and mTORC1 inhibitor rapamycin significantly decreased the levels of phosphorylated CREB, indicating that MAPK3/1-mTORC1 signaling is required for CREB activation. Furthermore, CREB could bind to the Kitl promoter region, and KG-501 significantly decreased the expression levels of KITL. In addition, KG-501 and CREB knockdown significantly decreased the levels of phosphorylated Akt, leading to a reduced number of oocytes with Foxo3a nuclear export. KG-501 also inhibited bpV (HOpic)-stimulated primordial follicle activation. Taken together, the results show that CREB is required for MAPK3/1-mTORC1 signaling-promoted KITL expression followed by the activation of primordial follicles.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Ovarian Follicle/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Female , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Inbred ICR , Naphthols/pharmacology , Organophosphates/pharmacology , Ovarian Follicle/drug effects , Phosphorylation , Signal Transduction/genetics , Stem Cell Factor/antagonists & inhibitors , Stem Cell Factor/metabolism , Tissue Culture Techniques , Vanadium Compounds/antagonists & inhibitors , Vanadium Compounds/pharmacology
10.
Molecules ; 25(7)2020 Apr 10.
Article in English | MEDLINE | ID: mdl-32290299

ABSTRACT

Discovering that metals are essential for the structure and function of biomolecules has given a completely new perspective on the role of metal ions in living organisms. Nowadays, the design and synthesis of new metal-based compounds, as well as metal ion binding components, for the treatment of human diseases is one of the main aims of bioinorganic chemistry. One of the areas in vanadium-based compound research is their potential anticancer activity. In this review, we summarize recent molecular and cellular mechanisms in the cytotoxic activity of many different synthetic vanadium complexes as well as inorganic salts. Such mechanisms shall include DNA binding, oxidative stress, cell cycle regulation and programed cell death. We focus mainly on cellular studies involving many type of cancer cell lines trying to highlight some new significant advances.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Vanadium Compounds/chemistry , Vanadium Compounds/pharmacology , Animals , Cell Cycle Checkpoints/drug effects , DNA Damage/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
11.
Growth Factors ; 37(3-4): 178-189, 2019 08.
Article in English | MEDLINE | ID: mdl-31646909

ABSTRACT

The in-vitro development of primordial follicles is critical for improving mammalian fertility and wildlife conservation. This study aimed to optimise the effective doses of bpV (HOpic) and stem cell factor (SCF) for the in-vitro activation of sheep primordial follicles. To do this, sheep ovarian cortex was treated with bpV (1.5, 15, and 150 µM) and SCF (50 and 100 ng/ml). Follicular count indicated that 15 µM bpV and 100 ng/ml SCF significantly increased normal primary follicles compared to other groups (p < 0.05). Also, a significant downregulation of P53 and PTEN, as well as the increased expression of PI3K was observed. The in-vitro maturation was more pronounced when the fragmented tissues were co-treated with selected doses of bpV and SCF. In conclusion, the combination of 15 µM bpV and 100 ng/ml SCF was the most effective treatment strategy for the activation and survival of primordial follicles in sheep ovarian fragments.


Subject(s)
Ovarian Follicle/growth & development , PTEN Phosphohydrolase/antagonists & inhibitors , Stem Cell Factor/pharmacology , Vanadium Compounds/pharmacology , Animals , Cells, Cultured , Female , Phosphatidylinositol 3-Kinase/biosynthesis , Sheep , Signal Transduction/drug effects , Tumor Suppressor Protein p53/antagonists & inhibitors
12.
Hum Reprod ; 34(2): 297-307, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30521029

ABSTRACT

STUDY QUESTION: Does ovarian follicle activation by phosphatase homologue of chromosome-10 (PTEN) inhibition affect DNA damage and repair in bovine oocytes and granulosa cells? SUMMARY ANSWER: PTEN inhibition promotes bovine non-growing follicle activation but results in increased DNA damage and impaired DNA repair capacity in ovarian follicles in vitro. WHAT IS KNOWN ALREADY: Inhibition of PTEN is known to activate primordial follicles but may compromise further developmental potential. In breast cancer cells, PTEN inhibition represses nuclear translocation of breast cancer susceptibility 1 (BRCA1) and Rad51; this impairs DNA repair resulting in an accumulation of damaged DNA, which contributes to cell senescence. STUDY DESIGN, SIZE, DURATION: Bovine ovarian tissue fragments were exposed to control medium alone or containing either 1 or 10 µM bpv(HOpic), a pharmacological inhibitor of PTEN, in vitro for 24 h. A sub-group of tissue fragments were collected for Western blot analysis after bpv(HOpic) exposure. The remainder were incubated in control medium for a further 5 days and then analysed histologically and by immunohistochemistry to detect DNA damage and repair pathways. PARTICIPANTS/MATERIALS, SETTING, METHODS: Bovine ovaries were obtained from abattoir-slaughtered heifers. Tissue fragments were exposed to either control medium alone or medium containing either 1 µM or 10 µM bpv(HOpic) for 24 h. Tissue fragments collected after 24 h were subjected to Akt quantification by Western blotting (six to nine fragments per group per experiment). Follicle stage and morphology were classified in remaining fragments. Immunohistochemical analysis included nuclear exclusion of FOXO3 as a marker of follicle activation, γH2AX as a marker of DNA damage, meiotic recombination 11 (MRE11), ataxia telangiectasia mutated (ATM), Rad51, breast cancer susceptibility 1 (BRCA1) and breast cancer susceptibility 2 (BRCA2) as DNA repair factors. A total of 29 550 follicles from three independent experiments were analysed. MAIN RESULTS AND THE ROLE OF CHANCE: Tissue fragments exposed to bpv(HOpic) had increased Akt phosphorylation at serine 473 (pAkt/Akt ratio, 2.25- and 6.23-fold higher in 1 and 10 µM bpv(HOpic) respectively compared to control, P < 0.05). These tissue fragments contained a significantly higher proportion of growing follicles compared to control (78.6% in 1 µM and 88.7% in 10 µM versus 70.5% in control; P < 0.001). The proportion of morphologically healthy follicles did not differ significantly between 1 µM bpv(HOpic) and control (P < 0.001) but follicle health was lower in 10 µM compared to 1 µM and control in all follicle types (P < 0.05). DNA damage in oocytes, indicated by expression of γH2AX, increased following exposure to 1 µM bpv(HOpic) (non-growing, 83%; primary follicles, 76%) and 10 µM (non-growing, 77%; primary, 84%) compared to control (non-growing, 30% and primary, 59%) (P < 0.05 for all groups). A significant reduction in expression of DNA repair proteins MRE11, ATM and Rad51 was observed in oocytes of non-growing and primary follicles of treatment groups (primary follicles in controls versus 10 µM bpv(HOpic): MRE, 68% versus 47%; ATM, 47% versus 18%; Rad51, 48% versus 24%), P < 0.05 for all groups. Higher dose bpv(HOpic) also resulted in lower expression of BRCA1 compared to control and 1 µM bpv(HOpic) (P < 0.001) in non-growing and primary follicles. BRCA2 expression was increased in oocytes of primary follicles in 1 µM bpv(HOpic) (36%) compared to control (20%, P = 0.010) with a marked decrease in 10 µM (1%, P ≤ 0.001). Granulosa cells of primary and secondary follicles in bpv(HOpic) groups showed more DNA damage compared to control (P < 0.05). However, bpv(HOpic) did not impact granulosa cell DNA repair capacity in secondary follicles, but BRCA1 declined significantly in higher dose bpv(HOpic). LARGE-SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: This study focuses on non-growing follicle activation after 6 days culture and may not reflect DNA damage and repair capacity in later stages of oocyte and follicle growth. WIDER IMPLICATIONS OF THE FINDINGS: In vitro activation of follicle growth may compromise the bidirectional signalling between oocyte and granulosa cells necessary for optimal oocyte and follicle health. This large animal model may be useful in optimising follicle activation protocols with a view to transfer for clinical application. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by Indonesia endowment fund for education. No competing interest. TRIAL REGISTRATION NUMBER: Not applicable.


Subject(s)
DNA Damage/drug effects , DNA Repair/drug effects , Ovarian Follicle/drug effects , PTEN Phosphohydrolase/antagonists & inhibitors , Vanadium Compounds/pharmacology , Animals , Cattle , Female , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Oocytes/drug effects , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , PTEN Phosphohydrolase/metabolism , Tissue Culture Techniques
13.
Biometals ; 32(5): 785-794, 2019 10.
Article in English | MEDLINE | ID: mdl-31552528

ABSTRACT

This study was conducted to investigate the damage caused by vanadium compounds and to explore the protective effects of berberine (BBR) in human umbilical vein endothelial cells (HUVECs). BBR is a biologically active small molecule found in Coptis rhizome, a remedy used in traditional Chinese medicine to treat diabetes. BBR has also been shown to lower blood glucose in diabetic patients. MTT assay was performed to observe the influence of bis(acetylacetonato)-oxidovanadium [VO(acac)2] or sodium metavanadate (NaVO3) and BBR on viability of HUVECs. The monolayer permeability of the HUVECs was assessed by measuring the transendothelial electrical resistance (TER). The endothelial nitric oxide synthase (eNOS) activity was detected by ELISA. Flow cytometry was performed to detect the generation of reactive oxygen species (ROS). The results showed that the viability of HUVECs was decreased by treatment with vanadium compounds 50-400 µM in a concentration-dependent manner, while 0.01-1 µM BBR effectively protected HUVECs from the inhibitory effects of vanadium compounds on cell viability. Also 100 and 200 µM VO(acac)2 induced high permeability and decreased eNOS activity in HUVECs. While 0.01-1 µM BBR showed no improvement in the permeability, and failed to reverse the VO(acac)2-induced changes of eNOS activity, but BBR treatment increased the eNOS activity in control cells. The addition of 200 µM VO(acac)2 significantly induced ROS generation in HUVECs, while 0.01 or 0.1 µM BBR reversed the change of ROS. In summary, BBR has protective effects in HUVECs damage induced by vanadium compounds, which is not mediated by eNOS, but related to reduced intracellular ROS.


Subject(s)
Berberine/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Vanadium Compounds/pharmacology , Cell Survival/drug effects , Humans , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species/metabolism
14.
Biometals ; 32(3): 545-561, 2019 06.
Article in English | MEDLINE | ID: mdl-31209680

ABSTRACT

Oncolytic viruses rewire the immune system and can lead to long-lasting antitumor defenses against primary and metastatic tumors. However, results from clinical studies have shown heterogeneity in responses suggesting that multiplexed approaches may be necessary to consistently generate positive outcomes in patients. To this end, we explored the combination of oncolytic rhabdovirus VSV∆51 with vanadium(V) dipicolinate derivatives, which have already been explored for their antidiabetic properties in animal models. The combination of vanadium-based dipicolinate compounds with VSV∆51 significantly increased viral replication and cytotoxicity in the human renal cell carcinoma cell line 786-0. The effects of three vanadium(V)-dipicolinate coordination complexes ([VO2dipic]-, [VO2dipic-OH]- and [VO2dipic-Cl]- with -OH or -Cl in the para position) were compared to that of the simple salts using spectroscopy and speciation profiles. Like the vanadate salts and the vanadyl cation, all dioxovanadium(V) dipicolinate complexes tested were found to increase viral infection and cytotoxicity when used in combination with VSV∆51. Viral sensitization is dependent on the vanadium since free dipicolinate ligands exerted no effect on viral infection and viability. The ability of these complexes to interact with interfaces and the stability of the complexes were evaluated under physiological conditions. Results indicate that these complexes undergo hydrolysis in cell culture media thereby generating vanadate. The vanadium dipicolinate derivatives in the context of immunovirotherapy shares similarities with previous studies exploring the antidiabetic properties of the compounds. The synergy between vanadium compounds and the oncolytic virus suggests that these compounds may be valuable in the development of novel and effective pharmaco-viral therapies.


Subject(s)
Antiviral Agents/pharmacology , Coordination Complexes/pharmacology , Oncolytic Virotherapy , Oncolytic Viruses/drug effects , Picolinic Acids/pharmacology , Vanadium Compounds/pharmacology , Virus Diseases/therapy , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Dose-Response Relationship, Drug , Humans , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Molecular Structure , Picolinic Acids/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured , Vanadium Compounds/chemistry , Virus Diseases/drug therapy
15.
Mol Ther ; 26(1): 56-69, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29175158

ABSTRACT

Oncolytic viruses (OV) are an emerging class of anticancer bio-therapeutics that induce antitumor immunity through selective replication in tumor cells. However, the efficacy of OVs as single agents remains limited. We introduce a strategy that boosts the therapeutic efficacy of OVs by combining their activity with immuno-modulating, small molecule protein tyrosine phosphatase inhibitors. We report that vanadium-based phosphatase inhibitors enhance OV infection in vitro and ex vivo, in resistant tumor cell lines. Furthermore, vanadium compounds increase antitumor efficacy in combination with OV in several syngeneic tumor models, leading to systemic and durable responses, even in models otherwise refractory to OV and drug alone. Mechanistically, this involves subverting the antiviral type I IFN response toward a death-inducing and pro-inflammatory type II IFN response, leading to improved OV spread, increased bystander killing of cancer cells, and enhanced antitumor immune stimulation. Overall, we showcase a new ability of vanadium compounds to simultaneously maximize viral oncolysis and systemic anticancer immunity, offering new avenues for the development of improved immunotherapy strategies.


Subject(s)
Genetic Vectors/genetics , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Vanadium Compounds/pharmacology , Animals , Biomarkers , Chemokine CXCL9/metabolism , Combined Modality Therapy , Cytokines/metabolism , Disease Models, Animal , Female , Genetic Therapy/methods , Humans , Immunotherapy , Inflammation Mediators/metabolism , Interferon Type I/metabolism , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Mice , Mortality , Reactive Oxygen Species/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
16.
J Integr Neurosci ; 18(4): 401-408, 2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31912698

ABSTRACT

Vascular dementia is the second most common type of dementia, yet no effective treatment for it exists. Akt and Erk1/2 signaling pathways are involved in neuronal survival. It has been reported that bisperoxovanadium (pyridin-2-squaramide), a novel squaramide compound, protects against cerebral ischemia injury via activation of Akt and Erk1/2. Here, the potential neuroprotective effect of bisperoxovanadium is shown for the first time in a model of vascular dementia induced in 6-month-old male Sprague-Dawley rats by two-vessel occlusion injury applied to 6-month-old. Following this lesion, bisperoxovanadium (pyridin-2-squaramide) (1 mg/kg/day) was intragastrically administered for four successive weeks. The Morris water maze test estimated cognitive function. The morphological examination was performed by hematoxylin-eosin staining. Akt and Erk1/2 protein abundance were assessed by Western blot. Results showed that bisperoxovanadium (pyridin-2-squaramide) attenuated not only cognitive dysfunction but also alleviated histopathological changes in rats with vascular dementia. Moreover, bisperoxovanadium (pyridin-2-squaramide) ultimately reduced neuronal apoptosis represented by the Bax/Bcl-2 ratio in the CA1 (cornu ammonis 1) region of the hippocampus. Importantly, the levels of p-Akt(ser473) and p-Erk1/2(Thr202/Tyr204>) were increased after treatment with bisperoxovanadium (pyridin-2-squaramide). It is concluded that the novel squaramide compound bisperoxovanadium (pyridin-2-squaramide) might be effective in the treatment of vascular dementia by activation of Akt and Erk1/2.


Subject(s)
Apoptosis/drug effects , CA1 Region, Hippocampal/drug effects , Cognitive Dysfunction/drug therapy , Dementia, Vascular/drug therapy , MAP Kinase Signaling System/drug effects , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Signal Transduction/drug effects , Vanadium Compounds/pharmacology , Animals , Behavior, Animal/drug effects , Cognitive Dysfunction/etiology , Dementia, Vascular/complications , Disease Models, Animal , Male , Maze Learning/drug effects , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Vanadium Compounds/administration & dosage
17.
Reprod Domest Anim ; 54(8): 1057-1063, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31087697

ABSTRACT

The vanadate-derivative dipotassium bisperoxo (5-hydroxy-pyridine-2-carboxylic) oxovanadate (V) (bpV(HOpic)), a pharmacological inhibitor of phosphatase and tensin homolog (PTEN), has been used in ovarian follicle culture systems for activation of follicular growth in vitro and suggested to be responsible for primordial follicle survival through indirect Akt activation. For pig ovarian tissue, it is still not clear which culture medium needs to be used, as well as which factors and hormones could influence follicular development; this also applies to bpV(HOpic) exposure. Therefore, ovarian cortical strips from pigs were cultured in 1 µM bpV(HOpic) (N = 24) or control medium (N = 24) for 48 hr. Media were then replaced with control medium and all tissue pieces incubated for additional 4 days. The strips were embedded in paraffin for histological determination of follicle proportions at the end of the culture period and compared to histological sections from tissue pieces without cultivation, which had been embedded right after preparation; comparison of healthy follicles for each developmental stage was performed to quantify follicle survival and activation. After 6-day culture, follicle activation occurred in tissue samples from both cultured groups but significantly more follicles showed progression of follicular development in the presence of 1 µM bpV(HOpic). The amount of non-vital follicles was not significantly increased during cultivation. BpV(HOpic) affects pig ovarian follicle development by promoting the initiation of follicle growth and development, similar as in rodent species and humans.


Subject(s)
Ovarian Follicle/drug effects , Swine , Vanadates/pharmacology , Vanadium Compounds/pharmacology , Animals , Culture Media/chemistry , Female , Tissue Culture Techniques
18.
Int J Mol Sci ; 20(2)2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30634697

ABSTRACT

Pancreatic cancer is characterized by one of the lowest five-year survival rates. In search for new treatments, some studies explored several metal complexes as potential anticancer drugs. Therefore, we investigated three newly synthesized oxidovanadium(IV) complexes with 2-methylnitrilotriacetate (bcma3-), N-(2-carbamoylethyl)iminodiacetate (ceida3-) and N-(phosphonomethyl)-iminodiacetate (pmida4-) ligands as potential anticancer compounds using pancreatic cancer cell lines. We measured: Cytotoxicity using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), neutral red (NR) and lactate dehydrogenase (LDH) assay; antiproliferative activity by bromodeoxyuridine BrdU assay; reactive oxygen species (ROS) generation and cell cycle analysis by flow cytometry; protein level by Western blot and cellular morphology by confocal laser scanning microscopy. The results showed that these oxidovanadium(IV) complexes were cytotoxic on pancreatic cancer cells (PANC-1 and MIA PaCa2), but not on non-tumor human immortalized pancreas duct epithelial cells (hTERT-HPNE) over the concentration range of 10⁻25 µM, following 48 h incubation. Furthermore, molecular mechanisms of cytotoxicity of [4-NH2-2-Me(Q)H][VO(bcma)(H2O)]2H2O (T1) were dependent on antiproliterative activity, increased ROS generation, cell cycle arrest in G2/M phase with simultaneous triggering of the p53/p21 pathway, binucleation, and induction of autophagy. Our study indicates that oxidovanadium(IV) coordination complexes containing 2-methylnitrilotriacetate ligand are good candidates for preclinical development of novel anticancer drugs targeting pancreatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carcinoma, Pancreatic Ductal/metabolism , Cell Cycle Checkpoints/drug effects , Pancreatic Neoplasms/metabolism , Vanadium Compounds/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Molecular Structure , Reactive Oxygen Species/metabolism , Vanadium Compounds/chemistry , Pancreatic Neoplasms
19.
Int J Exp Pathol ; 99(4): 180-188, 2018 08.
Article in English | MEDLINE | ID: mdl-30198103

ABSTRACT

Carnosine (ß-alanyl-L-histidine) is synthesized in the olfactory system, has antioxidant activity as a scavenger of free radicals and has been reported to have neuroprotective action in diseases which have been attributed to oxidative damage. In neurodegenerative disorders, such as Parkinson's and Alzheimer's diseases, impairment of olfactory function has been described. Vanadium derivatives are environmental pollutants, and its toxicity has been associated with oxidative stress. Vanadium toxicity on the olfactory bulb was reported previously. This study investigates the neuroprotective effect of carnosine on the olfactory bulb in a mice model of vanadium inhalation. Male mice were divided into four groups: vanadium pentoxide (V2 O5 ) [0.02 mol/L] inhalation for one hour twice a week; V2 O5 inhalation plus 1 mg/kg of carnosine administered daily; carnosine only, and the control group that inhaled saline. The olfactory function was evaluated using the odorant test. Animals were sacrificed four weeks after exposure. The olfactory bulbs were dissected and processed using the rapid Golgi method; cytological and ultrastructural analysis was performed and malondialdehyde (MDA) concentrations were measured. The results showed evidence of olfactory dysfunction caused by vanadium exposure and also an increase in MDA levels, loss of dendritic spines and necrotic neuronal death in the granule cells. But, in contrast, vanadium-exposed mice treated with carnosine showed an increase in dendritic spines and a decrease in neuronal death and in MDA levels when compared with the group exposed to vanadium without carnosine. These results suggest that dendritic spine loss and ultrastructural alterations in the granule cells induced by vanadium are mediated by oxidative stress and that carnosine may modulate the neurotoxic vanadium action, improving the olfactory function.


Subject(s)
Carnosine/pharmacology , Neuroprotective Agents/pharmacology , Olfactory Bulb/drug effects , Spine/pathology , Animals , Disease Models, Animal , Neurotoxicity Syndromes/drug therapy , Olfactory Bulb/pathology , Oxidative Stress/drug effects , Spine/drug effects , Vanadium Compounds/pharmacology
20.
Neurochem Res ; 43(7): 1424-1438, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29882124

ABSTRACT

Bisperoxovanadium (pyridine-2-carboxyl) [bpV(pic)] is a commercially available PTEN inhibitor. Previous studies from us and others have shown that bpV(pic) confers neuroprotection in cerebral ischemia injury. We set up to determine whether ERK 1/2 activation plays a role in bpV(pic)-induced neuroprotective effect in cerebral ischemia injury. We found that the phosphorylation levels of Akt (p-AKT) and ERK1/2 (p-ERK 1/2) were down-regulated after cerebral ischemia-reperfusion injury. The injection of bpV(pic) after injury not only increased the level of p-AKT but also the level of p-ERK 1/2. While the inhibition of PTEN mediated the up-regulatation of p-AKT and p-ERK 1/2 by bpV(pic). Interestingly, the ERK 1/2 activation induced by bpV(pic) was also independent of the inhibition of PTEN. Our results indicate that bpV(pic) protects against OGD-induced neuronal death and promotes the functional recovery of stroke animals through PTEN inhibition and ERK 1/2 activation, respectively. This study suggests that the effect of bpV(pic) on ERK 1/2 signaling should be considered while using bpV(pic) as a PTEN inhibitor.


Subject(s)
Brain Injuries/drug therapy , Brain Ischemia/drug therapy , MAP Kinase Signaling System/drug effects , Reperfusion Injury/drug therapy , Vanadium Compounds/pharmacology , Animals , Disease Models, Animal , Male , Neurons/drug effects , Neurons/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Recovery of Function/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL