Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
3.
Tissue Cell ; 81: 102015, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36657254

ABSTRACT

Polycystic ovary syndrome (PCOS) is an endocrine disorder seen in women of reproductive age and has been gradually increasing over the years. The mechanism of the syndrome has still not been clearly understood. In this study, the possible effects of exogenously administrated melatonin on melatonin (MT1) receptor, Growth Differentiation Factor-9 (GDF9), and Bone Morphogenetic Protein-15 (BMP15) in experimental PCOS were investigated. Thirty-two 6-8-week-old Sprague-Dawley rats were randomly divided into four groups (n = 8 in each) as Sham control (Group 1), Melatonin (Group 2), PCOS (Group 3), and PCOS + Melatonin (Group 4) groups. At the end of the 21st day, the experiment was terminated, the ovary tissues were taken, and Hematoxylin-Eosin staining, MT1, GDF9, BMP15 immunohistochemical labeling, western blot, and quantitative real-time polymerase chain reaction (qPCR) analyses were performed. Serum Luteinizing Hormone (LH)/Follicle Stimulating Hormone (FSH) levels and colpo-cytological examinations were also carried out. The results revealed that melatonin administration increased the expression levels of the MT1 receptor, GDF9, and BMP15 in PCOS at protein and mRNA levels. It was determined that melatonin administration reduced the microscopic symptoms of PCOS. Melatonin was found to be effective via the MT1 receptor in the pathogenesis of PCOS, and it suppressed the transport pathways of GDF9 to granulosa cells in antral follicles.


Subject(s)
Infertility , Melatonin , Polycystic Ovary Syndrome , Humans , Rats , Animals , Female , Polycystic Ovary Syndrome/drug therapy , Melatonin/pharmacology , Receptor, Melatonin, MT1 , Rats, Sprague-Dawley
4.
ACS Appl Bio Mater ; 2022 Aug 31.
Article in English | MEDLINE | ID: mdl-36043987

ABSTRACT

Selective detection and effective therapy of brain cancer, specifically, the very aggressive glioblastoma multiforme (GBM), remains one of the paramount challenges in clinical settings. While radiotherapy combined surgery is proposed as the main treatment course, it has several drawbacks such as complexity of the operation and common development of recurrent tumors in this course of patient care. Unique opportunities presented by photodynamic therapy (PDT) offer promising, effective, and precise therapy against GBM cells along with simultaneous imaging opportunities. However, activatable, theranostic molecular systems in PDT modality for GBM remained scarce. Specifically, even though elevated ß-galactosidase (ß-gal) activity in glioblastoma cells is well-documented, targeted, activatable therapeutic PDT agents have not been realized. Herein, we report a ß-galactosidase (ß-gal) activatable phototheranostic agent based on an iodinated resorufin core (RB-1) which was realized in only three steps with commercial reagents in 29% overall yield. RB-1 showed very high singlet oxygen (1O2) quantum yield (54%) accompanied by a remarkable turn-on response in fluorescence upon enzymatic activation. RB-1 was tested in different cell lines and revealed selective photocytotoxicity in U-87MG glioblastoma cells. Additionally, thanks to almost 7% fluorescence quantum yield (ΦF) despite extremely high 1O2 generation yield, RB-1 was also demonstrated as a successful agent for fluorescence imaging of U-87MG cells. Due to significantly lower (ß-gal) activity in healthy cells (NIH/3T3), RB-1 stayed in a passive state and showed minimal photo and dark toxicity. RB-1 marks the first example of a ß-gal activatable phototheranostic agent toward effective treatment of glioblastoma.

5.
ACS Appl Bio Mater ; 5(6): 2754-2767, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35537187

ABSTRACT

Activity-based theranostic photosensitizers are highly attractive in photodynamic therapy as they offer enhanced therapeutic outcome on cancer cells with an imaging opportunity at the same time. However, photosensitizers (PS) cores that can be easily converted to activity-based photosensitizers (aPSs) are still quite limited in the literature. In this study, we modified the dicyanomethylene-4H-chromene (DCM) core with a heavy iodine atom to get two different PSs (DCMO-I, I-DCMO-Cl) that can be further converted to aPS after simple modifications. The effect of iodine positioning on singlet oxygen generation capacity was also evaluated through computational studies. DCMO-I showed better performance in solution experiments and further proved to be a promising phototheranostic scaffold via cell culture studies. Later, a cysteine (Cys) activatable PS based on the DCMO-I core (DCMO-I-Cys) was developed, which induced selective photocytotoxicity along with a fluorescence turn-on response in Cys rich cancer cells.


Subject(s)
Iodine , Neoplasms , Photochemotherapy , Fluorescence , Iodine/therapeutic use , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Singlet Oxygen/therapeutic use
6.
Tissue Cell ; 73: 101585, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34375824

ABSTRACT

The involvement of endometrial IGF-1R/IGF-1/Bcl-2 pathways and the potential regulatory effects of exogenously administrated melatonin on this expression is investigated in the experimental PCOS model in the present study. Thirty-two 6-8 week old Sprague Dawley rats were divided into four groups: the Sham Control Group (1% CMC/day by oral gavage [o.g.]); the Melatonin Group (2 mg/kg/day melatonin by subcutaneous administration [s.c.]); the Experimental PCOS Group (1 mg/kg/day Letrozole by o.g.); and the Experimental PCOS + Melatonin Group (1 mg/kg/day Letrozole by o.g. and 2 mg/kg/day melatonin by s.c. administration). Vaginal smear samples were taken from the 14th day to the end of the experiment for colpocytological measurements. At the end of the 21 day experimental period, uterine tissues were taken; Hematoxylin-Eosin histochemical, IGF-1R/IGF-1/Bcl-2, PCNA immuno-histochemical stainings and western blot analyses were performed for related antibodies. All of the data was supported statistically. The epithelium of endometrium lost its single-layer structure in some parts, separation was observed between the epithelium and the basal membrane junction, intracellular edema was found in the uterine glands by the polycystic ovary-induction. Also this induction increased the expression of IGF-1R/IGF-1, Bcl-2, and PCNA proteins. Morphological degenerations returned to its normal appearance generally by the melatonin administrations and melatonin also regulated the increased expression of endometrial IGF-1R/IGF-1/Bcl-2 and PCNA pathways. It is concluded that additional studies are needed, using melatonin as a supporting agent may be appropriate in cases of PCOS.


Subject(s)
Endometrium/metabolism , Insulin-Like Growth Factor I/metabolism , Melatonin/pharmacology , Polycystic Ovary Syndrome/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Animals , Body Weight/drug effects , Endometrium/drug effects , Endometrium/pathology , Female , Ovary/drug effects , Ovary/pathology , Polycystic Ovary Syndrome/pathology , Proliferating Cell Nuclear Antigen/metabolism , Rats, Sprague-Dawley , Regression Analysis , Staining and Labeling , Vagina/drug effects
7.
Neurochem Res ; 46(12): 3325-3341, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34432181

ABSTRACT

Neuroinflammation and oxidative stress cooperate to compromise the function of the central nervous system (CNS). Colloidal platinum nanoparticles (Pt NPs) are ideal candidates for reducing the deleterious effects of neuroinflammation since they act as free radical scavengers. Here we evaluated the effects of Pt NPs on several markers of lipopolysaccharide (LPS)-induced inflammation in cultured BV-2 microglial cells. BV-2 cells were treated with increased dilutions (1-100 ppm) of Colloidal Pt and/or LPS (1-10 µg/mL) at different exposure times. Three different protocols of exposure were used combining Pt NPs and LPS: (a) conditioning-protective effect (pre-post-treat), (b) therapeutic effect (co-treat) and (c) conditioning-therapeutic effect (pre-co-treat). After exposure to LPS for 24 h, cells were used for assessment of cell viability, reactive oxygen species (ROS) generation, lactate dehydrogenase (LDH) activity, apoptosis and caspase-3 levels, cell proliferation, mitochondrial membrane potential, inducible nitric oxide (iNOS) activity, pro-inflammatory cytokine (IL-1ß, TNF-α and IL-6) levels, and phagocytic activity. Low concentrations (below or equal to 10 ppm) of Colloidal Pt prevented or ameliorated the LPS-induced increase in ROS formation, loss of mitochondrial membrane potential, induction of apoptosis, increase in LDH release, increase in pro-inflammatory cytokines and iNOS, inhibition of phagocytosis linked to microglial persistence in the M1 phase phenotype, loss of cell adhesion, differentiation and/or proliferation, as well as loss of cell viability. These protective effects were evident when cells were preconditioned with Pt NPs prior to LPS treatment. Collectively, the findings demonstrate that at low concentrations, Pt NPs can regulate the function and phenotype of BV-2 cells, activating protective mechanisms to maintain the microglial homeostasis and reduce inflammatory events triggered by the inflammatory insults induced by LPS. These preventive/protective effects on the LPS pro-inflammatory model are linked to the antioxidant properties and phagocytic activity of these NPs.


Subject(s)
Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Metal Nanoparticles/administration & dosage , Microglia/drug effects , Neuroinflammatory Diseases/drug therapy , Oxidative Stress , Phagocytosis , Platinum/pharmacology , Animals , Cytokines/metabolism , Mice , Microglia/metabolism , Microglia/pathology , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism
8.
ACS Med Chem Lett ; 12(5): 752-757, 2021 May 13.
Article in English | MEDLINE | ID: mdl-34055222

ABSTRACT

Iodination of the silicon-fluorescein core revealed a new class of highly cytotoxic, red-shifted and water-soluble photosensitizer (SF-I) which is also fairly emissive to serve as a theranostic agent. Singlet oxygen generation capacity of SF-I was evaluated chemically, and up to 45% singlet oxygen quantum yield was reported in aqueous solutions. SF-I was further tested in triple negative breast (MDA MB-231) and colon (HCT-116) cancer cell lines, which are known to have limited chemotherapy options as well as very poor prognosis. SF-I induced efficient singlet oxygen generation and consequent photocytotoxicity in both cell lines upon light irradiation with a negligible dark toxicity while allowing cell imaging at the same time. SF-I marks the first ever example of a silicon xanthene-based photosensitizer and holds a lot of promise as a small-molecule-based theranostic scaffold.

9.
Biochem Cell Biol ; 99(5): 645-654, 2021 10.
Article in English | MEDLINE | ID: mdl-33930279

ABSTRACT

The discovery of new pharmacological agents is needed to control the progression of osteoarthritis (OA), characterized by joint cartilage damage. Human OA chondrocyte (OAC) cultures were either applied to S-allylcysteine (SAC), a sulfur-containing amino acid derivative, or colchicine, an ancient anti-inflammatory therapeutic, for 24 h. SAC or colchicine did not change viability at 1 nM-10 µM but inhibited p-JNK/pan-JNK. While SAC seems to be more effective, both agents inhibited reactive oxygen species (ROS), 3-nitrotyrosine (3-NT), lipid hydroperoxides (LPO), advanced lipoxidation end-products (ALEs as 4-hydroxy-2-nonenal, HNE), advanced glycation end-products (AGEs), and increased glutathione peroxidase (GPx) and type-II-collagen (COL2). IL-1ß, IL-6, and osteopontin (OPN) were more strongly inhibited by SAC than by colchicine. In contrast, TNF-α was inhibited only by SAC, and COX2 was only inhibited by colchicine. Casp-1/ICE, GM-CSF, receptor for advanced glycation end-products (RAGE), and toll-like receptors (TLR4) were inhibited by both agents, but bone morphogenetic protein 7 (BMP7) was partially inhibited by SAC and induced by colchicine. Nuclear factor erythroid 2-related factor 2 (Nrf2) was induced by SAC; in contrast, it was inhibited by colchicine. Although they exert opposite effects on TNF-α, COX2, BMP7, and Nrf2, SAC and colchicine exhibit anti-osteoarthritic properties in OAC by modulating redox-sensitive inflammatory signaling.


Subject(s)
Chondrocytes/drug effects , Cysteine/analogs & derivatives , Inflammation/drug therapy , Osteoarthritis/drug therapy , Aged , Antigens, Neoplasm/metabolism , Chondrocytes/metabolism , Cysteine/pharmacology , Female , Humans , Inflammation/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Middle Aged , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Osteoarthritis/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism
10.
Neurotox Res ; 39(3): 588-597, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33713301

ABSTRACT

Aldose reductase (AR) catalyzes the conversion of glucose to sorbitol in a NADPH-dependent reaction, thereby increasing the production of reactive oxygen species (ROS). Since AR activation is linked to redox dysregulation and cell damage in neurodegenerative diseases, AR inhibitors (ARIs) constitute promising therapeutic tools for the treatment of these disorders. Among these compounds, the novel substituted triazinoindole derivatives cemtirestat (CMTI) and COTI, as well as the clinically employed epalrestat (EPA) and the pyridoindole-antioxidant stobadine (STB), were tested in both PC12 cells and BV2 microglia exposed to four different neurotoxic models. These include (1) oxidative stress with hydrogen peroxide (H2O2), (2) mitochondrial complex IV inhibition with NaN3, (3) endoplasmic reticulum-stress and lipotoxicity induced by palmitic acid/bovine serum albumin (PAM/BSA), and (4) advanced carbonyl compound lipotoxicity by 4-hydroxynonenal (4-HNE). All toxic compounds decreased cell viability and increased ROS formation in both PC12 and BV2 cells in a concentration-dependent manner (1-1000 µM; NaN3 < H2O2≈PAM/BSA < 4-HNE). In PC12 cells, EPA increased cell viability in all toxic models only at 1 µM, whereas CMTI restored baseline viability in all toxic models. COTI afforded protection against lipotoxicity, while STB only prevented H2O2-induced toxicity. Except for the 4-HNE model, EPA prevented ROS generation in all other toxic models, whereas CMTI, COTI, and STB prevented ROS production in all toxic models. In BV2 cells, EPA and CMTI restored baseline cell viability in all toxic models tested, while COTI and STB did not prevent the loss of viability in the NaN3 model. All ARIs and STB efficiently prevented ROS formation in all toxic models in a concentration-independent manner. The differential protective effects evoked by the novel ARIs and STB on the toxic models tested herein provide novel and relevant comparative evidence for the design of specific therapeutic strategies against neurodegenerative events associated with neurological disorders.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Antioxidants/pharmacology , Carbolines/pharmacology , Enzyme Inhibitors/pharmacology , Microglia/drug effects , Oxidative Stress/drug effects , Rhodanine/analogs & derivatives , Thiazolidines/pharmacology , Aldehyde Reductase/metabolism , Animals , Antioxidants/chemistry , Carbolines/chemistry , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Indoles/chemistry , Indoles/pharmacology , Mice , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oxidative Stress/physiology , PC12 Cells , Pyridones/chemistry , Pyridones/pharmacology , Rats , Rhodanine/chemistry , Rhodanine/pharmacology , Thiazolidines/chemistry
11.
Neurochem Int ; 145: 104993, 2021 05.
Article in English | MEDLINE | ID: mdl-33610590

ABSTRACT

Alzheimer's disease (AD) is the most common dementia causing progressive loss of memory and compromised cognitive functions. Although the neurotoxic mechanisms underlying AD have yet to be fully elucidated, hyperglycemia seems to trigger oxidative and inflammatory responses in the brain of afflicted patients. Removal of free radicals reduces the neurotoxic effects of hyperglycemia in AD models. In this study we investigated the neuroprotective effects of the antioxidant phytoconstituents oleuropein (OLE), rutin (RUT), luteolin (LUT) and S-allylcysteine (SAC) in an experimental model combining the exposure to high glucose (HG, mimicking chronic hyperglycemia) plus amyloid-ß peptide 1-42 (Aß1-42, mimicking AD) in primary hippocampal neurons. Cells were pre-treated with OLE, RUT, LUT or SAC (10-1000 nM), and then co-treated with high glucose (GLU, 150 mM) for 24 h plus 500 nM oligomeric Aß1-42 for 24 h more. Cell viability and reactive oxygen species (ROS) formation were assessed as indices of survival/toxicity and oxidative stress, respectively. Activity/expression of antioxidant enzymes, toxic adducts, inflammatory molecules, mitochondrial membrane potential (ΔΨm) and the pattern of amyloid aggregation were also assessed. The GLU + Aß1-42 treatment significantly decreased cell viability, increased ROS formation, reduced superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase activities, augmented Advanced Glycation End Products- and 4-hydroxynonenal-adducts generation, increased 3-nitrotyrosine and inflammatory outcomes such as inducible nitric oxide synthase, interleukin 1ß and Tumor Necrosis Factor α, decreased MMP and augmented amyloid aggregation. All phytoconstituents reduced in a differential manner all toxic endpoints, with SAC showing the highest efficacy in preventing loss of cell viability and oxidative damage, whereas RUT was most efficacious in mitigating inflammatory endpoints. Combined, the results of this study suggest that protection afforded by these compounds against GLU + Aß1-42-induced cell damage in hippocampal neurons is attributable to their properties as redox modulators, which might act through a concerted mechanism oriented to reduce oxidative stress and neuroinflammation.


Subject(s)
Amyloid beta-Peptides/toxicity , Glucose/toxicity , Hippocampus/metabolism , Hyperglycemia/metabolism , Neurons/metabolism , Peptide Fragments/toxicity , Phytochemicals/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Female , Hippocampus/drug effects , Hyperglycemia/chemically induced , Hyperglycemia/drug therapy , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Neurons/drug effects , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phytochemicals/therapeutic use , Pregnancy , Rats , Rats, Wistar
12.
J Invest Surg ; 34(9): 935-946, 2021 Sep.
Article in English | MEDLINE | ID: mdl-32003261

ABSTRACT

OBJECTIVE: To investigate the protective effect and underlying mechanisms of B. monnieri, a medicinal plant, on kidney and skeletal muscle injury induced by infra-renal abdominal aorta clamping for 2-hours (ischemia) and following removal of the clamp (reperfusion, 2-hours). METHODS: Rats were divided into four groups (n = 6): (I) animals given only saline (sham-control); (II) animals given B. monnieri extract for 10-days (300 mg/kg/day) (Bacopa-treated sham); (III) animals subjected to ischemia/reperfusion (I/R); (IV) animals given B. monnieri extract and then subjected to I/R. Kidneys and lower extremity muscles were examined for GPx, CAT, iNOS, 3-NT, IL-1ß and TNF-α. Apoptosis and injury were evaluated by TUNEL and H&E staining, respectively. RESULTS: I/R resulted in TUNEL positive cells, periarterial edema and glomerular capillary dilatation, decreased GPx activity, unchanged CAT, iNOS, 3-NT, IL-1ß and TNF-α in kidney. B. monnieri minimized renal remote reperfusion injury, and Group IV showed a lower degree of renal histopathology score when compared to the others. B. monnieri mitigated muscle I/R injury, decreased muscle hypertrophy, myofibril abnormalities and apoptosis. Muscle 3-NT and cytokine levels were increased by I/R, and B. monnieri inhibited iNOS and 3-NT both in sham-control and I/R groups. Muscle GPx unaffected by I/R or B. monnieri, but CAT was inhibited only in B. monnieri-treated I/R group. Muscle iNOS, 3-NT, IL-1ß, TNF-α levels and CAT activity of B. monnieri-treated I/R rats were lower than those in sham-control or Bacopa-treated sham. CONCLUSIONS: B. monnieri can protect the directly affected organ as well as distant organs against I/R injury by modulating anti-inflammatory and anti-nitrosative pathways.


Subject(s)
Bacopa , Reperfusion Injury , Animals , Anti-Inflammatory Agents/therapeutic use , Kidney , Rats , Rats, Wistar , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control
13.
Neurotox Res ; 39(2): 210-226, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33146867

ABSTRACT

Cellular redox dysregulation produced by aldose reductase (AR) in the presence of high blood sugar is a mechanism involved in neurodegeneration commonly observed in diabetes mellitus (DM) and Parkinson's disease (PD); therefore, AR is a key target for treatment of both diseases. The substituted triazinoindole derivatives 2-(3-thioxo-2H-[1,2,4]triazino[5,6-b]indol-5(3H)-yl) acetic acid (cemtirestat or CMTI) and 2-(3-oxo-2H-[1,2,4]triazino[5,6-b]indol-5(3H)-yl) acetic acid (COTI) are well-known AR inhibitors (ARIs). The neuroprotective properties of CMTI, COTI, the clinically used epalrestat (EPA), and the pyridoindole antioxidants stobadine and SMe1EC2 were all tested in the neurotoxic models produced by hyperglycemic glucotoxicity (HG, 75 mM D-glucose, 72 h), 6-hydroxydopamine (6-OHDA), and HG+6-OHDA models in PC12 cells. Cell viability decreased in all toxic models, increased by 1-5 µM EPA, and decreased by COTI at ≥ 2.5 µM. In the HG model alone, where compounds were present in the medium for 24 h after a continuous 24-h exposure to HG, cell viability was improved by 100 nM-5 µM EPA, 1-10 µM ARIs, and the antioxidants studied, but decreased by EPA at ≥ 10 µM. In the 6-OHDA model alone, where cells were treated with compounds for 24 h and further exposed to 100 µM 6-OHDA (8 h), only the antioxidants protected cell viability. In the HG+6-OHDA model, where cells were treated with all compounds (1 nM to 50 µM) for 48 h and exposed to 75 mM glucose for 24 h followed by incubation with 6-OHDA for 8 h, cell viability was protected by 100 nM-10 µM ARIs and 100-500 nM EPA, but not by antioxidants. All ARIs inhibited the HG+6-OHDA-induced increase in iNOS, IL-1ß, TNF-α, 3-NT, and total oxidant status at 1-50 µM, while increased SOD, CAT, GPx, and total antioxidant status at 1-10 µM. EPA and CMTI also reduced the HG+6-OHDA-induced increase in the cellular levels of nuclear factor kB (NF-KB). The neuroprotective potential of the novel ARIs and the pyridoindole antioxidants studied constitutes a promising tool for the development of therapeutic strategies against DM-induced and PD-related neurodegeneration.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Glucose/toxicity , Inflammation/metabolism , Neuroprotective Agents/administration & dosage , Nitrosative Stress/drug effects , Oxidopamine/toxicity , Animals , Antioxidants/administration & dosage , Cell Survival/drug effects , Inflammation/chemically induced , PC12 Cells , Rats
14.
J Tissue Eng Regen Med ; 14(12): 1841-1857, 2020 12.
Article in English | MEDLINE | ID: mdl-33010113

ABSTRACT

Osteoarthritic chondrocytes show an over-activity of inflammatory catabolic mediators, and olive products have attracted attention because they were discovered to have some benefits on osteoarthritis patients. We investigated the mechanisms of action of olive leaf polyphenolic compounds in osteoarthritic chondrocytes (OACs) using a standardized leaf extract, ZeyEX, and its main phenolic component, oleuropein, also compared with anti-inflammatory drug ibuprofen. OACs, isolated from joint-cartilages of Grade 4 OA patients, were found to express COMP and MMP-9 throughout their culture period. ZeyEX, oleuropein, and ibuprofen increased cell viability at concentrations of 1-100 nM, did not change at 500 nM-50 µM, but inhibited at ≥100 µM. The adherence profile of OACs increased with 1 µM of ibuprofen or ZeyEX and 10 nM-1 µM oleuropein. Although the markers for oxidative and nitrosative stresses (ROS and 3-NT) generally inhibited by three agents, the inhibitory effect of ZeyEX on 3-NT emerged dramatically (1 nM-10 µM). Lipid-hydroperoxides and HNE-adducts were also inhibited by each agent, but AGE-adducts unchanged by oleuropein while reduced by ZeyEX and ibuprofen. Inflammatory biomarkers, IL-1ß, IL-6, Casp-1/ICE, and TNF-α, were inhibited by three agents, however osteopontin and GM-CSF by only ZeyEX and ibuprofen. A decreased COMP, TLR4, and RAGE expression levels were observed by three agents, but only the effects of ZeyEX was concentration-dependent. In particular, ZeyEX and oleuropein improved COL2, inhibited p-JNK/JNK, and increased GPx. COX2 was only inhibited by ibuprofen. The results indicate that polyphenolic-olive compounds counteract redox-sensitive inflammatory aggressions in osteoarthritic chondrocytes that may stop the progression of pathology and allow regeneration.


Subject(s)
Chondrocytes/pathology , Ibuprofen/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Olea/chemistry , Osteoarthritis/pathology , Phenol/pharmacology , Receptor for Advanced Glycation End Products/metabolism , Toll-Like Receptor 4/metabolism , Aged , Aldehydes/metabolism , Biomarkers/metabolism , Cartilage, Articular/pathology , Cell Adhesion/drug effects , Cell Survival/drug effects , Chondrocytes/drug effects , Female , Glycation End Products, Advanced/metabolism , Humans , Inflammation Mediators/metabolism , Lipid Peroxides/metabolism , Male , Middle Aged , Oxidation-Reduction , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
15.
Neurochem Int ; 140: 104817, 2020 11.
Article in English | MEDLINE | ID: mdl-32781098

ABSTRACT

Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder linked to various converging toxic mechanisms. Evidence suggests that hyperglycemia induces oxidative stress, mitochondrial dysfunction, inflammation and excitotoxicity, all of which play important roles in the onset and progression of AD pathogenesis. The endocannabinoid system (ECS) orchestrates major physiological responses, including neuronal plasticity, neuroprotection, and redox homeostasis, to name a few. The multi-targeted effectiveness of the ECS emerges as a potential approach to treat AD. Here we characterized the protective properties of the endocannabinoids arachidonylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG), the synthetic cannabinoids CP 55-940 and WIN 55,212-2, and the fatty acid amide hydrolase (FAAH) inhibitor URB597, on a combined hyperglycemia + oligomeric amyloid ß peptide (Aß1-42) neurotoxic model in primary hippocampal neurons which exhibit several AD features. Cells were treated with cannabinoid agents at increased concentrations (1 nM-1 µM) for 6 h, and then co-treated with 150 mM glucose (GLU, 24 h), followed by incubation with 500 nM Aß1-42 (24 h). Cell viability/survival, reactive oxygen species (ROS) levels, antioxidant enzyme (SOD, CAT, GPx and GRx) activities, biological products of oxidative damage (AGE and HNE adducts) and nitrosative stress (3-NT), several endpoints of inflammation (iNOS, IL-1ß and TNF-α), amyloid quantification, mitochondrial membrane potential, and the involvement of the Nrf2 pathway, were all evaluated. The combined high glucose + amyloid beta 1-42 (GLU + Aß1-42) condition decreased cell viability and mitochondrial membrane potential, while augmenting oxidative damage and inflammation. All agents tested preserved cell viability and stimulated mitochondrial membrane potential, while reducing all the evaluated toxic endpoints in a differential manner, with URB597 showing the highest efficacy. The neuroprotective efficacy of all cannabinoid agents, except for URB597, led to partial recruitment of specific antioxidant activity and Nrf2 pathway regulation. Our results support the neuroprotective potential of these agents at low concentrations against the damaging effects of GLU + Aß1-42, affording new potential modalities for the design of AD therapies.


Subject(s)
Amyloid beta-Peptides/toxicity , Cannabinoids/pharmacology , Hyperglycemia/metabolism , Inflammation Mediators/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Peptide Fragments/toxicity , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Glucose/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Hyperglycemia/chemically induced , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/physiology , Rats , Rats, Wistar
16.
Drug Chem Toxicol ; 43(1): 96-103, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30207190

ABSTRACT

Rotenone, an environmental toxin, triggers Parkinson's disease (PD)-like pathology through microglia-mediated neuronal death. The effects and molecular mechanisms of flavonoid luteolin against rotenone-induced toxicity was assessed in microglial BV2 cells. Cells were pretreated with luteolin (1-50 µM) for 12 h and then was co-treated with 20 µM of rotenone for an additional 12 h in the presence of luteolin. The viability (MTT), IL-1ß and TNF-α levels and lactate dehydrogenase (LDH) release (ELISA), and Park2, Lrrk2, Pink1, Nrf2 and Trx1 mRNA levels (qRT-PCR) were measured. In rotenone exposed microglia, luteolin increased viability significantly at lower concentrations (1-5 µM) compared to higher concentrations (25-50 µM). Rotenone increased LDH release and IL-1ß levels in a dose-dependent manner (1-20 µM). Luteolin inhibited rotenone-induced LDH release, however the activity decreased in concentration-dependent manner Neither rotenone nor luteolin altered TNF-α levels, but luteolin reduced IL-1ß levels in a concentration dependent manner in rotenone exposed cells. The mRNA levels of Nrf2 and Trx1, which are the master regulators of redox state, were increased by rotenone, as well as by luteolin, which exhibited an inverse relationship between its concentration and effect (1-20 µM). Park2 mRNA levels increased by luteolin, but decreased by rotenone. Pink1 mRNA levels was not altered by rotenone or luteolin. Lrrk2 mRNA levels reduced by luteolin, while it was increased by rotenone. Results suggest that luteolin have favorable effects on regulation of oxidative stress response, genes associated with PD and inflammatory pathways, hence protects microglia against rotenone toxicity in a hormetic manner.


Subject(s)
Luteolin/pharmacology , Microglia/drug effects , Oxidative Stress/drug effects , Parkinsonian Disorders/prevention & control , Animals , Cell Line , Dose-Response Relationship, Drug , Hormesis/drug effects , Inflammation/pathology , Inflammation/prevention & control , L-Lactate Dehydrogenase/drug effects , L-Lactate Dehydrogenase/metabolism , Luteolin/administration & dosage , Mice , Microglia/pathology , Oxidation-Reduction/drug effects , Parkinsonian Disorders/genetics , Parkinsonian Disorders/physiopathology , Rotenone/administration & dosage , Rotenone/toxicity
17.
Ocul Immunol Inflamm ; 28(6): 888-897, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-31429629

ABSTRACT

INTRODUCTION: It was aimed to evaluate the efficacy of two tea tree oil (TTO)-based cleansing gels in chronic blepharitis patients. METHODS: Group-1 (basic gel containing 3%(w/w)-TTO) included 50 eyes of 25 patients and group-2 (advanced gel containing 3%(w/w)-TTO plus essential oils and vitamins) included 48 eyes of 24 patients. Ocular Surface Disease Index (OSDI), tear breakup time (TBUT), ocular surface staining pattern, Schirmer's test, impression cytology, Demodex presence and TNF-α, IL-6, IL-1ß levels were evaluated at the first visit and 1 month after treatment. RESULTS: In both groups, the mean OSDI score decreased (p1:0.001, p2:0.001), TBUT increased (p1:0.002, p2:0.004). In group-1, Demodex presence decreased from 42% to 27.8%; in group-2 from 54.2% to 20.6% (p1:0.302, p2:0.004). IL-1ß and IL-6 decreased in group-2 (p1:0.002, p2:0.050). TNF-α decreased in both groups (p1:0.001, p2:0.001). CONCLUSION: Both formulations improved ocular surface parameters. Group 2 showed more reduction in tear cytokines and Demodex count.


Subject(s)
Anti-Infective Agents, Local/therapeutic use , Blepharitis/drug therapy , Tea Tree Oil/therapeutic use , Adult , Animals , Anti-Infective Agents, Local/adverse effects , Blepharitis/metabolism , Blepharitis/parasitology , Chronic Disease , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Eye Proteins/metabolism , Eyelashes/parasitology , Female , Gels , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Middle Aged , Mites , Pharmaceutical Preparations , Tea Tree Oil/adverse effects , Tears/metabolism , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism
18.
Naunyn Schmiedebergs Arch Pharmacol ; 393(4): 651-661, 2020 04.
Article in English | MEDLINE | ID: mdl-31802170

ABSTRACT

Hyperglycemia is considered a key risk factor for development of diabetic complications including neuropathy. There is strong scientific evidence showing a primary role of aldose reductase, the first enzyme of the polyol pathway, in the cascade of metabolic imbalances responsible for the detrimental effects of hyperglycemia. Aldose reductase is thus considered a significant drug target. We investigated the effects of cemtirestat, a novel aldose reductase inhibitor, in the streptozotocin-induced rat model of uncontrolled type 1 diabetes in a 4-month experiment. Markedly increased sorbitol levels were recorded in the erythrocytes and the sciatic nerve of diabetic animals. Osmotic fragility of red blood cells was increased in diabetic animals. Indices of thermal hypoalgesia were significantly increased in diabetic rats. Tactile allodynia, recorded in diabetic animals in the early stages, turned to mechanical hypoalgesia by the end of the experiment. Treatment of diabetic animals with cemtirestat (i) reduced plasma triglycerides and TBAR levels; (ii) did not affect the values of HbA1c and body weights; (iii) reversed erythrocyte sorbitol accumulation to near control values, while sorbitol in the sciatic nerve was not affected; (iv) ameliorated indices of the erythrocyte osmotic fragility; and (v) attenuated the symptoms of peripheral neuropathy more significantly in the middle of the experiment than at the end of the treatment. Taking into account the lipid metabolism as an interesting molecular target for prevention or treatment of diabetic peripheral neuropathy, the triglyceride-lowering effect of cemtirestat should be considered in future studies. The most feasible mechanisms of triglyceride-lowering action of cemtirestat were suggested.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Diabetes Mellitus, Experimental/drug therapy , Diabetic Neuropathies/drug therapy , Hypoglycemic Agents/therapeutic use , Indoleacetic Acids/therapeutic use , Sulfhydryl Compounds/therapeutic use , Triglycerides/blood , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Experimental/blood , Diabetic Neuropathies/blood , Hypoglycemic Agents/pharmacology , Indoleacetic Acids/pharmacology , Male , Osmotic Fragility/drug effects , Rats, Wistar , Sulfhydryl Compounds/pharmacology
19.
Turk Neurosurg ; 29(6): 887-900, 2019.
Article in English | MEDLINE | ID: mdl-31608966

ABSTRACT

AIM: To investigate the potential protective effects of melatonin on the chronic radiation emitted by third generation mobile phones on the brain. MATERIAL AND METHODS: A total of 24 male Wistar albino rats were divided into four equal groups. Throughout a 90-day experiment, no application was performed on the control group. The second group was exposed to 2100 MHz radiation for 30 minutes. Subcutaneous melatonin was injected into the third group. Subcutaneous melatonin injection was applied 40 minutes before radiation and then the fourth group was exposed to radiation for 30 minutes. At the end of the experiment, brain (cerebrum and cerebellum) tissues were taken from the subjects. Histochemical, immunohistochemical, ultrastructural and western blot analyses were applied. In addition to brain weight, Purkinje cells’ number, immunohistochemical H Score analyses and the results of the Western blot were examined statistically. RESULTS: With the application of radiation, neuronal edema, relatively-decreased numbers of neurons on hippocampal CA1 and CA3 regions, displacement of the Purkinje neurons and dark neurons findings were observed as a result of histochemical stainings. Radiation also activated the NMDA-receptor 2B/Calpain-1/Caspase-12 pathway, NMDA-receptor 2B and Calpain-1 with the findings being supported by western blot analyses. Pre-increased protein synthesis before apoptosis was identified by electron microscopy. CONCLUSION: Mobile phone radiation caused certain (ultra) structural changes on the brain and activated the NMDA-receptor 2B/ Calpain-1/Caspase-12 pathway; in addition, melatonin was found to be effective, but insufficient in demonstrating the protective effects.


Subject(s)
Brain/metabolism , Calpain/metabolism , Caspase 12/metabolism , Electromagnetic Radiation , Melatonin/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Brain/drug effects , Brain/radiation effects , Calpain/radiation effects , Caspase 12/radiation effects , Cell Phone , Male , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/radiation effects , Signal Transduction/drug effects , Signal Transduction/radiation effects
20.
Mol Neurobiol ; 56(2): 844-856, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29802570

ABSTRACT

The endocannabinoid system (ECS) actively participates in several physiological processes within the central nervous system. Among such, its involvement in the downregulation of the N-methyl-D-aspartate receptor (NMDAr) through a modulatory input at the cannabinoid receptors (CBr) has been established. After its production via the kynurenine pathway (KP), quinolinic acid (QUIN) can act as an excitotoxin through the selective overactivation of NMDAr, thus participating in the onset and development of neurological disorders. In this work, we evaluated whether the pharmacological inhibition of fatty acid amide hydrolase (FAAH) by URB597, and the consequent increase in the endogenous levels of anandamide, can prevent the excitotoxic damage induced by QUIN. URB597 (0.3 mg/kg/day × 7 days, administered before, during and after the striatal lesion) exerted protective effects on the QUIN-induced motor (asymmetric behavior) and biochemical (lipid peroxidation and protein carbonylation) alterations in rats. URB597 also preserved the structural integrity of the striatum and prevented the neuronal loss (assessed as microtubule-associated protein-2 and glutamate decarboxylase localization) induced by QUIN (1 µL intrastriatal, 240 nmol/µL), while modified the early localization patterns of CBr1 (CB1) and NMDAr subunit 1 (NR1). Altogether, these findings support the concept that the pharmacological manipulation of the endocannabinoid system plays a neuroprotective role against excitotoxic insults in the central nervous system.


Subject(s)
Amidohydrolases/drug effects , Corpus Striatum/drug effects , Quinolinic Acid/pharmacology , Receptor, Cannabinoid, CB1/drug effects , Animals , Arachidonic Acids/pharmacology , Corpus Striatum/injuries , Endocannabinoids/pharmacology , Lipid Peroxidation/drug effects , Male , Neostriatum/drug effects , Neostriatum/metabolism , Polyunsaturated Alkamides/pharmacology , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...