Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Hum Exp Toxicol ; 40(12_suppl): S530-S539, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34715756

ABSTRACT

BACKGROUND: Mitochondria are the main target organelles through which drugs and chemicals exert their toxic effect on cardiomyocytes. The mitochondria-related mechanisms of celecoxib-induced cardiotoxicity have been extensively studied. Accumulated evidence shows natural molecules targeting mitochondria have proven to be effective in preventing cardiotoxicity. PURPOSE: In the present study, we examined the ameliorative effect of gallic acid (GA) against celecoxib-induced cellular and mitochondrial toxicity in isolated cardiomyocytes and mitochondria. RESEARCH DESIGN: The isolated cardiomyocytes and mitochondria were divided into various group, namely, control, celecoxib, celecoxib + GA (10, 50, and 100 µM). Several cellular and mitochondrial parameters such as cell viability, lipid peroxidation, succinate dehydrogenase (SDH) activity, reactive oxygen species (ROS) formation, mitochondrial membrane potential (MMP) collapse, and mitochondrial swelling were assessed in isolated cardiomyocytes and mitochondria. RESULTS: Our results showed that administration of celecoxib (16 µg/ml) induced cytotoxicity and mitochondrial dysfunction at 6 h and 1 h, respectively, which is associated with lipid peroxidation intact cardiomyocytes, mitochondrial ROS formation, MMP collapse, and mitochondrial swelling. The cardiomyocytes and mitochondria treated with celecoxib + GA (10, 50, and 100 µM) significantly and dose-dependently restore the altered levels of cellular and mitochondrial parameters. CONCLUSIONS: We concluded that GA through antioxidant potential and inhibition of mitochondrial permeability transition (MPT) pore exerted ameliorative role in celecoxib-induced toxicity in isolated cardiomyocytes and mitochondria. The data of the current study suggested that GA supplementation may reduce celecoxib-induced cellular and mitochondrial toxicity during exposure and may provide a potential prophylactic and defensive candidate for coxibs-induced mitochondrial dysfunction, oxidative stress, and cardiotoxicity.


Subject(s)
Celecoxib/toxicity , Gallic Acid/pharmacology , Mitochondrial Transmembrane Permeability-Driven Necrosis/drug effects , Animals , Cells, Cultured , Embryonic Stem Cells/drug effects , Male , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Rats , Rats, Wistar
2.
Matrix Biol ; 95: 1-14, 2021 01.
Article in English | MEDLINE | ID: mdl-33065248

ABSTRACT

The tumor microenvironment encompasses a complex cellular network that includes cancer-associated fibroblasts, inflammatory cells, neo-vessels, and an extracellular matrix enriched in angiogenic growth factors. Decorin is one of the main components of the tumor stroma, but it is not expressed by cancer cells. Lack of this proteoglycan correlates with down-regulation of E-cadherin and induction of ß-catenin signaling. In this study, we investigated the role of a decorin-deficient tumor microenvironment in colon carcinoma progression and metastasis. We utilized an established model of colitis-associated cancer by administering Azoxymethane/Dextran sodium sulfate to adult wild-type and Dcn-/- mice. We discovered that after 12 weeks, all the animals developed intestinal tumors independently of their genotype. However, the number of intestinal neoplasms was significantly higher in the Dcn-/- microenvironment vis-à-vis wild-type mice. Mechanistically, we found that under unchallenged basal conditions, the intestinal epithelium of the Dcn-/- mice showed a significant increase in the protein levels of epithelial-mesenchymal transition associated factors including Snail, Slug, Twist, and MMP2. In comparison, in the colitis-associated cancer evoked in the Dcn-/- mice, we found that intercellular adhesion molecule 1 (ICAM-1) was also significantly increased, in parallel with epithelial-mesenchymal transition signaling pathway-related factors. Furthermore, a combined Celecoxib/decorin treatment revealed a promising therapeutic efficacy in treating human colorectal cancer cells, in decorin-deficient animals. Collectively, our results shed light on colorectal cancer progression and provide a protein-based therapy, i.e., treatment using recombinant decorin, to target the tumor microenvironment.


Subject(s)
Cadherins/genetics , Colonic Neoplasms/drug therapy , Decorin/genetics , Proteoglycans/genetics , Animals , Azoxymethane/toxicity , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Celecoxib/toxicity , Colitis-Associated Neoplasms/chemically induced , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Decorin/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Extracellular Matrix/drug effects , Humans , Mice , Neoplasm Metastasis , Tumor Microenvironment/drug effects , beta Catenin/genetics
3.
Free Radic Biol Med ; 156: 70-82, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32561319

ABSTRACT

Deoxynivalenol is a trichothecene mycotoxin which naturally contaminates small grain, cereals intended for human and animal consumption. Investigations for dermal toxicity of DON has been needed and highlighted by WHO. Previous studies on dermal toxicity suggest that DON has DNA damaging potential leading to skin tumor initiation in mice skin. However, considering its toxicological manifestations arising after dermal exposure, strategies for its prevention/protection are barely available in literatute. Collectively, our study demonstrated that N-acetylcysteine (NAC), precursor of glutathione, significantly alters the genotoxic potential of DON. Further NAC in combination with Celecoxib (CXB) inhibits tumor growth by altering antioxidant status and increasing autophagy in DON initiated Swiss mice. Despite the broad spectrum use of CXB, its use is limited by the concerns about its adverse effects on the cardiovascular system. Serum parameters and histology analysis revealed that CXB (2 mg) when applied topically for 24 weeks did not impart any cardiovascular toxicity which could be because skin permeation potential of CXB was quite low when analyzed through HPLC analysis. Although the anticancer effects of CXB and NAC have been studied, however, the combination of NAC and CXB has yet not been explored for any cancer treatment. Therefore our observations provide additional insights into the therapeutic effects of combinatorial treatment of CXB and NAC against skin tumor prevention. This approach might form a novel alternative strategy for skin cancer treatment as well as skin associated toxicities caused by mycotoxins such as DON. This combinatorial approach can overcome the limitations associated with the use of CXB for long term as topical application of the same seems to be safe in comparison to the oral mode of administration.


Subject(s)
Acetylcysteine , Skin Neoplasms , Animals , Autophagy , Celecoxib/toxicity , Mice , Trichothecenes
4.
Bioorg Chem ; 97: 103670, 2020 04.
Article in English | MEDLINE | ID: mdl-32088417

ABSTRACT

Selective COX-2 inhibitor celecoxib was found directly inhibiting the growth of tested phytopathogenic fungi with the inhibitory rate ranging from 30 to 40% at 100 µg/ml. Lead optimization of celecoxib led to the identification of compound 12 among its derivatives as the most active antifungal candidate. The antifungal effect of compound 12 was supposed to be independent of COX-2 inhibition. Transcriptome profiling analysis of Fusarium graminearium (PH-1) treated with compound 12 brought about 406 up-regulated and 572 down-regulated differentially express genes (DEGs) respectively.


Subject(s)
Celecoxib/analogs & derivatives , Crop Protection/methods , Cyclooxygenase 2 Inhibitors/chemistry , Fungicides, Industrial/chemistry , Fusarium/drug effects , Plant Diseases/prevention & control , Celecoxib/toxicity , Crops, Agricultural/microbiology , Cyclooxygenase 2 Inhibitors/toxicity , Fungicides, Industrial/toxicity , Fusarium/genetics , Gene Expression Regulation, Fungal/drug effects , Plant Diseases/microbiology , Transcriptome/drug effects
5.
Cardiovasc Pathol ; 46: 107194, 2020.
Article in English | MEDLINE | ID: mdl-31982687

ABSTRACT

BACKGROUND: Celecoxib, a selective cyclooxygenase-2 inhibitor, was recently associated with increased incidence of aortic stenosis and found to produce a valvular calcification risk in vitro. Several cyclooxygenase-2 independent celecoxib derivatives have been developed and identified as possible therapies for inflammatory diseases due to their cadherin-11 inhibitory functions. Potential cardiovascular toxicities associated with these cyclooxygenase-2 independent celecoxib derivatives have not yet been investigated. Furthermore, the mechanism by which celecoxib produces valvular toxicity is not known. METHODS AND RESULTS: Celecoxib treatment produces a 2.8-fold increase in calcification in ex vivo porcine aortic valve leaflets and a more than 2-fold increase in calcification in porcine aortic valve interstitial cells cultured in osteogenic media. Its cyclooxygenase-2 independent derivative, 2,5-dimethylcelecoxib, produces a similar 2.5-fold increase in calcification in ex vivo leaflets and a 13-fold increase in porcine aortic valve interstitial cells cultured in osteogenic media. We elucidate that this offtarget effect depends on the presence of either of the two media components: dexamethasone, a synthetic glucocorticoid used for osteogenic induction, or cortisol, a natural glucocorticoid present at basal levels in the fetal bovine serum. In the absence of glucocorticoids, these inhibitors effectively reduce calcification. By adding glucocorticoids or hydrocortisone to a serum substitute lacking endogenous glucocorticoids, we show that dimethylcelecoxib conditionally induces a 3.5-fold increase in aortic valve calcification and osteogenic expression. Treatment with the Mitogen-activated protein kinase kinase inhibitor, U0126, rescues the offtarget effect, suggesting that celecoxib and dimethylcelecoxib conditionally augment Mitogen-activated protein kinase kinase/extracellular-signal-regulated kinase activity in the presence of glucocorticoids. CONCLUSION: Here we identify glucocorticoids as a possible source of the increased valvular calcification risk associated with celecoxib and its cyclooxygenase-2 independent derivatives. In the absence of glucocorticoids, these inhibitors effectively reduce calcification. Furthermore, the offtarget effects are not due to the drug's intrinsic properties as dual cyclooxygenase-2 and cadherin-11 inhibitors. These findings inform future design and development of celecoxib derivatives for potential clinical therapy.


Subject(s)
Aortic Valve/drug effects , Calcinosis/chemically induced , Celecoxib/toxicity , Cyclooxygenase 2 Inhibitors/toxicity , Dexamethasone/toxicity , Glucocorticoids/toxicity , Heart Valve Diseases/chemically induced , Hydrocortisone/toxicity , Osteogenesis/drug effects , Pyrazoles/toxicity , Sulfonamides/toxicity , Animals , Aortic Valve/metabolism , Aortic Valve/pathology , Cadherins/genetics , Cadherins/metabolism , Calcinosis/genetics , Calcinosis/metabolism , Calcinosis/pathology , Celecoxib/analogs & derivatives , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart Valve Diseases/genetics , Heart Valve Diseases/metabolism , Heart Valve Diseases/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Signal Transduction , Sus scrofa , Tissue Culture Techniques
6.
Commun Biol ; 2: 288, 2019.
Article in English | MEDLINE | ID: mdl-31396568

ABSTRACT

Tuberculosis, caused by infection with Mycobacterium tuberculosis (Mtb), kills over 1.6 million people each year despite availability of antibiotics. The increase in drug resistant Mtb strains is a major public health emergency and host-directed therapy as adjunct to antibiotic treatment has gained increased interest. Cyclooxygenase inhibitors (COXi) are frequently used drugs to alleviate tuberculosis related symptoms. Mouse studies of acute intravenous Mtb infection have suggested a potential benefit of COXi for host-directed therapy. Here we show that COXi treatment (ibuprofen and celecoxib) is detrimental to Mtb control in different mouse models of respiratory infection. This effect links to impairments of the Type-1 helper (Th1) T-cell response as CD4 T-cells in COXi-treated animals have significantly decreased Th1 differentiation, reduced IFNγ expression and decreased protective capacity upon adoptive transfer. If confirmed in clinical trials, these findings could have major impact on global health and question the use of COXi for host-directed therapy.


Subject(s)
Celecoxib/toxicity , Cyclooxygenase Inhibitors/toxicity , Ibuprofen/toxicity , Lung/drug effects , Mycobacterium tuberculosis/pathogenicity , Th1 Cells/drug effects , Tuberculosis, Pulmonary/microbiology , Adoptive Transfer , Aerosols , Animals , Bacterial Load , Cell Differentiation/drug effects , Cyclooxygenase 2 Inhibitors/toxicity , Disease Models, Animal , Disease Progression , Female , Host-Pathogen Interactions , Inhalation Exposure , Interferon-gamma/immunology , Lung/immunology , Lung/microbiology , Lymphocyte Activation/drug effects , Mice, Inbred C3H , Mycobacterium tuberculosis/immunology , Th1 Cells/immunology , Th1 Cells/microbiology , Th1 Cells/transplantation , Tuberculosis, Pulmonary/immunology
7.
Tissue Eng Part C Methods ; 24(9): 514-523, 2018 09.
Article in English | MEDLINE | ID: mdl-30101668

ABSTRACT

INTRODUCTION: The inflammatory cascade and production of prostaglandins may play a role in the pathogenesis of arthrofibrosis, a debilitating condition after joint replacement and other orthopedic procedures. Cyclooxygenase 2 (COX-2) inhibitors may mitigate the inflammatory response and formation of arthrofibrosis, but oral delivery is associated with risk of systemic side effects in many patients. The nonsteroidal anti-inflammatory drug, celecoxib, may have therapeutic benefits for arthrofibrosis, but current methods for its local delivery (e.g., biologically derived microspheres) are not translatable to immediate clinical use. Therefore, we investigated the use of a drug scaffold for sustainable intra-articular delivery of therapeutic doses of celecoxib. MATERIALS AND METHODS: Celecoxib was eluted from clinically approved biodegradable collagen membranes over 7 days as measured by UV spectroscopy and high-performance liquid chromatography/mass spectroscopy. Eluted concentrations of celecoxib were examined for toxicity (live/dead staining) and profibrotic gene expression (real-time-quantitative polymerase chain reaction) in rabbit knee capsular fibroblasts in vitro. RESULTS: Sustained concentrations of celecoxib eluted from the membrane over 7 days from both a wet and dry scaffold, with a burst release (30-45%) of celecoxib in the first 2 h. Rabbit cells treated with eluted concentrations experienced a toxic response to the burst release doses, and inhibitory effects on profibrotic genes were seen in response to the sustained doses eluted from the scaffold. CONCLUSIONS: This study characterized the novel use of collagen scaffolds for intra-articular drug delivery to treat arthrofibrosis. Scaffolds exhibit celecoxib release through an initial burst release followed by sustained release of antifibrotic doses over 7 days. Thus, collagen scaffolds are promising for clinician-directed treatment of arthrofibrosis.


Subject(s)
Celecoxib/pharmacology , Joints/pathology , Tissue Scaffolds/chemistry , Animals , Celecoxib/toxicity , Cell Death/drug effects , Cell Line , Fibrosis , Gene Expression Regulation/drug effects , Joints/drug effects , Rabbits
8.
Biochem Biophys Res Commun ; 501(2): 329-335, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29505793

ABSTRACT

Celecoxib is a non-steroidal anti-inflammatory drug that selectively inhibits cyclooxygenase-2 and is prescribed for severe pain and inflammation. The excellent therapeutic effects of celecoxib mean that it is frequently used clinically, including for women of child-bearing age. However, the prenatal effects of this compound have not been studied extensively in vertebrates. The present study examined the developmental toxicity of celecoxib using a frog embryo teratogenic assay-Xenopus (FETAX). In addition, we examined its effects on cell migration using co-cultures of human umbilical vein endothelial cells and 10T1/2 cells. These studies revealed that celecoxib induced concentration-dependent mortality and various malformations of the Xenopus internal organs, including gut miscoiling, haemorrhage, and oedema. Celecoxib also downregulated the expression of vascular wall markers (Msr and alpha smooth muscle actin) and other organ-specific markers (Nkx2.5, Cyl104 and IFABP). In vitro co-culture studies revealed that celecoxib inhibited pericyte migration and differentiation into vascular smooth muscle cells. In conclusion, celecoxib was both toxic and teratogenic in Xenopus embryos, where it produced serious heart and vessel malformation by inhibiting vascular wall maturation and vascular network formation.


Subject(s)
Celecoxib/toxicity , Teratogens/toxicity , Xenopus laevis/embryology , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Biomarkers , Blood Vessels/abnormalities , Blood Vessels/drug effects , Blood Vessels/embryology , Celecoxib/administration & dosage , Cell Movement/drug effects , Dose-Response Relationship, Drug , Embryo, Nonmammalian/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Male , Xenopus laevis/physiology
9.
Cell Biochem Funct ; 35(7): 407-413, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28975647

ABSTRACT

Mesenchymal stem cells (MSCs) have therapeutic potential for treatment of diabetes. However, in vitro behavior of MSCs in high glucose condition as well as presence of glucose lowering agents is not fully understood. Because MSCs have an important role in tissue repair, we examined the effects of metformin and celecoxib on viability of MSCs in different glucose conditions. MSCs, from umbilical cord blood, were cultured in normoglycemic (glucose 5.5 mM), midglycemic (glucose 10 mM), and hyperglycemic (glucose 25 mM) conditions, and the cell viability was evaluated by MTT assay. The cytotoxicity and secretion of GDF-15 were further tested in MSCs treated with metformin and celecoxib in various glucose concentrations. Our results showed that high glucose condition lowered viability of MSCs. Metformin treatment also inhibited proliferation of MSCs, but its toxicity was not changed in high glucose condition. Celecoxib induced cytotoxicity in MSCs, and the toxicity was increased in high glucose condition. Metformin and celecoxib induced release from MSCs; however, high glucose inhibited the metformin-induced GDF-15 release. These findings suggested that metformin did not increase the cytotoxicity of high glucose condition in MSCs. Moreover, celecoxib treatment in diabetic condition can reduce the viability of MSCs to proliferate and regenerate perhaps via change in release of GDF-15.


Subject(s)
Celecoxib/toxicity , Cell Proliferation/drug effects , Glucose/pharmacology , Growth Differentiation Factor 15/metabolism , Metformin/toxicity , Cell Line , Cell Survival/drug effects , Fetal Blood/cytology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism
10.
World J Gastroenterol ; 23(29): 5304-5312, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28839430

ABSTRACT

AIM: To counteract/reveal celecoxib-induced toxicity and NO system involvement. METHODS: Celecoxib (1 g/kg b.w. ip) was combined with therapy with stable gastric pentadecapeptide BPC 157 (known to inhibit these lesions, 10 µg/kg, 10 ng/kg, or 1 ng/kg ip) and L-arginine (100 mg/kg ip), as well as NOS blockade [N(G)-nitro-L-arginine methyl ester (L-NAME)] (5 mg/kg ip) given alone and/or combined immediately after celecoxib. Gastrointestinal, liver, and brain lesions and liver enzyme serum values in rats were assessed at 24 h and 48 h thereafter. RESULTS: This high-dose celecoxib administration, as a result of NO system dysfunction, led to gastric, liver, and brain lesions and increased liver enzyme serum values. The L-NAME-induced aggravation of the lesions was notable for gastric lesions, while in liver and brain lesions the beneficial effect of L-arginine was blunted. L-arginine counteracted gastric, liver and brain lesions. These findings support the NO system mechanism(s), both NO system agonization (L-arginine) and NO system antagonization (L-NAME), that on the whole are behind all of these COX phenomena. An even more complete antagonization was identified with BPC 157 (at both 24 h and 48 h). A beneficial effect was evident on all the increasingly negative effects of celecoxib and L-NAME application and in all the BPC 157 groups (L-arginine + BPC 157; L-NAME + BPC 157; L-NAME + L-arginine + BPC 157). Thus, these findings demonstrated that BPC 157 may equally counteract both COX-2 inhibition (counteracting the noxious effects of celecoxib on all lesions) and additional NOS blockade (equally counteracting the noxious effects of celecoxib + L-NAME). CONCLUSION: BPC 157 and L-arginine alleviate gastrointestinal, liver and brain lesions, redressing NSAIDs' post-surgery application and NO system involvement.


Subject(s)
Anti-Ulcer Agents/therapeutic use , Arginine/therapeutic use , Brain/drug effects , Chemical and Drug Induced Liver Injury/drug therapy , Cyclooxygenase 2 Inhibitors/toxicity , Stomach Ulcer/drug therapy , Animals , Antidotes/therapeutic use , Brain/pathology , Celecoxib/administration & dosage , Celecoxib/toxicity , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Cyclooxygenase 2 Inhibitors/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination/methods , Humans , Liver/drug effects , Liver/pathology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Peptide Fragments/therapeutic use , Proteins/therapeutic use , Rats , Rats, Wistar , Stomach/drug effects , Stomach/pathology , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology
11.
Life Sci ; 185: 95-102, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28754619

ABSTRACT

Celecoxib is widely used in pregnant women but its influence on fetal brain neurogenesis is largely unknown. The objective of the present study was to examine the influence of celecoxib to fetal brain development and to investigate whether curcumin could ameliorate celecoxib-induced neurotoxicity. Pregnant mice, cultured neurons and cultured neural progenitor cells were all treated with celecoxib with or without curcumin. The change in proliferation, differentiation and the activity of Wnt/ß-catenin signaling pathway were then assessed. Here, we report that prenatal celecoxib exposure inhibited the activity of Wnt/ß-catenin pathway and disrupted the proliferation of neuronal progenitor cells, leading to a decrease of newborn neurons in fetal frontal cortex. Treatment with curcumin significantly could attenuate the celecoxib-induced deficits in proliferation through activating the Wnt/ß-Catenin pathway. Our study for the first time showed that maternal celecoxib administration caused detrimental effects to fetal brain development and provided an evidence of the therapeutic role of curcumin in celecoxib-induced neurotoxicity.


Subject(s)
Brain/drug effects , Celecoxib/toxicity , Curcumin/pharmacology , Neurogenesis/drug effects , Neurotoxicity Syndromes/drug therapy , Animals , Animals, Newborn , Brain/embryology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Frontal Lobe/drug effects , Frontal Lobe/embryology , Mice , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Neurons/drug effects , Neurotoxicity Syndromes/etiology , Pregnancy , Wnt Signaling Pathway/drug effects
12.
Life Sci ; 185: 15-22, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28739304

ABSTRACT

AIMS: The hypertensive effect of the immunosuppressant drug cyclosporine (CSA) is paralleled, and probably triggered, by impaired arterial baroreceptor sensitivity (BRS). Here we asked if these effects of CSA are influenced by co-administration of nonsteroidal antiinflammatory drugs (NSAIDs) and if the oxidative NADPH-oxidase (NADPHox)/Rho-kinase (ROCK) pathway mediates this interaction. MATERIALS AND METHODS: Female rats were treated for 10days with CSA (25mg/kg/day), diclofenac (DIC, COX-1/COX-2 inhibitor, 1mg/kg/day), celecoxib (COX-2 inhibitor, 10mg/kg/day), or their combinations. Baroreflex curves relating changes in heart rate (HR) to increases or decreases in blood pressure (BP) evoked by phenylephrine (PE) and sodium nitroprusside (SNP), respectively, were constructed and slopes of the curves were taken as measures of BRS. KEY FINDINGS: Compared with control rats, CSA increased BP and reduced reflex chronotropic responses as indicated by the significantly smaller BRSPE and BRSSNP values. Similar effects were observed in rats treated with diclofenac alone or combined with CSA. Whereas CSA hypertension was maintained after selective COX-2 inhibition by celecoxib, the concomitant BRS reductions were largely eliminated. NADPHox inhibition by diphenyleneiodonium (DPI) blunted the CSA/DIC-evoked increases and decreases in BP and BRSPE, respectively. By contrast, fasudil (ROCK inhibitor) had no effect on CSA/DIC hypertension but reversed the associated reductions in both BRSPE and BRSSNP. SIGNIFICANCE: Depending on the nature of the cardiovascular response, NADPHox and ROCK contribute variably to the worsened cardiovascular profile in CSA/DIC-treated rats. Further, celecoxib rather than diclofenac could be a better choice as an add-on therapy to CSA in autoimmune arthritic conditions.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Celecoxib/toxicity , Cyclosporine/toxicity , Diclofenac/toxicity , Hypertension/chemically induced , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Baroreflex/drug effects , Blood Pressure/drug effects , Celecoxib/administration & dosage , Cyclosporine/administration & dosage , Diclofenac/administration & dosage , Drug Interactions , Female , NADPH Oxidases/metabolism , Phenylephrine/pharmacology , Rats , Rats, Wistar , rho-Associated Kinases/metabolism
13.
J Physiol Pharmacol ; 68(2): 265-272, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28614776

ABSTRACT

The sphincters failure is a part of NSAIDs-toxicity that can be accordingly counteracted. We used a safe stable gastric pentadecapeptide BPC 157 (GEPPPGKPADDAGLV, MW 1419), LD1 not achieved, since successful in inflammatory bowel disease trials, and counteracts esophagitis, sphincters failure, gastrointestinal ulcer and skin ulcer, external and internal fistulas in rats, and particularly counteracts all NSAIDs-lesions. We assessed lower esophageal sphincter and pyloric sphincter pressure (cmH2O) in rats treated with various NSAIDs regimens, at corresponding time points, known to produce stomach, small intestine lesions, hepatotoxicity and encephalopathy. Assessment was after diclofenac (12.5 mg/kg, 40 mg/kg intraperitoneal challenge), ibuprofen (400 mg/day/kg intraperitoneally for 4 weeks), paracetamol (5.0 g/kg intraperitoneal challenge), aspirin (400 mg/kg intraperitoneally or intragastrically), celecoxib (0.5 mg/kg, 1.0 mg/kg intraperitoneally). BPC 157 (10 µg/kg, 10 ng/kg) was given immediately after NSAIDs (intraperitoneally or intragastrically) or given in drinking water. Regularly, in all control NSAIDs fall of pressure occurred in both sphincters rapidly and then persisted. By contrast, in all NSAIDs-rats that received BPC 157, initial fall of pressure was minimized and pressure values restored to normal values. All tested NSAIDs decrease pressure in both sphincters, whilst BPC 157 counteracts their effects and restored both sphincters function.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Anti-Ulcer Agents/pharmacology , Esophageal Sphincter, Lower/drug effects , Peptide Fragments/pharmacology , Proteins/pharmacology , Pylorus/drug effects , Acetaminophen/toxicity , Animals , Aspirin/toxicity , Celecoxib/toxicity , Diclofenac/toxicity , Ibuprofen/toxicity , Male , Pressure , Rats, Wistar
14.
Pharm Biol ; 55(1): 1295-1303, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28274156

ABSTRACT

CONTEXT: The cardiotoxic effect of selective cyclo-oxygenase-2 inhibitors is well known. While rofecoxib and valdecoxib have been withdrawn, celecoxib remains on the market. Folic acid, a naturally occurring vitamin, has been shown to reduce myocardial ischemia and post-reperfusion injury in rats. OBJECTIVE: This study examined the cardiac effects of celecoxib and folic acid on doxorubicin-induced cardiomyopathy in rats. MATERIALS AND METHODS: Cardiomyopathy was induced in male Wistar rats with six intraperitoneal injections of 2.5 mg/kg doxorubicin over a period of two weeks. The effect of 28 days of celecoxib (100 mg/kg/day) and its combination with folic acid (10 mg/kg/day) was studied on doxorubicin-induced cardiomyopathy according to serum lactate dehydrogenase (LDH), creatine kinase (CK-MB), troponin-T (Tn-T), tumor necrosis factor alpha (TNF-α), cardiac thiobarbituric acid reactive substance (TBARS), and glutathione (GSH) levels as well as systolic blood pressure (SBP), heart rate (HR) and ultrastructural studies. RESULTS: Celecoxib cardiotoxicity was manifested by significant increases in the LDH, Tn-T, TNF-α, CK-MB, SBP, HR (p < 0.001) and TBARS (p < 0.01) levels and a significant decrease in the GSH (p < 0.05) level when used alone or administered with doxorubicin. However, the combination of folic acid with celecoxib caused a significant reversal of these parameters and reduced the cardiotoxicity of celecoxib that was aggravated by doxorubicin. The ultrastructural study also revealed myocardial protection with this combination. DISCUSSION AND CONCLUSION: Folic acid protects against the cardiotoxic effects of celecoxib, which are aggravated in the presence of doxorubicin. Folic acid may act as a useful adjunct in patients who are taking celecoxib.


Subject(s)
Cardiotoxicity/prevention & control , Celecoxib/toxicity , Disease Models, Animal , Folic Acid/therapeutic use , Heart Failure/chemically induced , Heart Failure/prevention & control , Animals , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Heart Failure/metabolism , Male , Random Allocation , Rats , Rats, Wistar
15.
PLoS One ; 11(10): e0164250, 2016.
Article in English | MEDLINE | ID: mdl-27723829

ABSTRACT

Although drug resistance is often observed in metastatic recurrence of breast cancer, little is known about the intrinsic drug resistance in such metastases. In the present study, we found, for the first time, that MDA-MB-231BR, a brain metastatic variant of a human breast cancer cell line, was refractory to treatment with 5-fluorouracil (5-FU) even without chronic drug exposure, compared to its parent cell line, MDA-MB-231, and a bone metastatic variant, MDA-MB-231SCP2. Both the mRNA and protein levels of COX-2 and BCL2A1 in MDA-MB-231BR were significantly higher than those in MDA-MB-231 or MDA-MB-231SCP2. Neither the COX-2 inhibitor celecoxib nor the NF-κB inhibitor BAY11-7082 could sensitize MDA-MB-231BR to 5-FU, indicating that COX-2 plays little, if any, role in the resistance of MDA-MB-231BR to 5-FU. Although BCL2-family inhibitor ABT-263 failed to sensitize MDA-MB-231BR to 5-FU at a dose at which ABT-263 is considered to bind to BCL2, BCL2-xL, and BCL2-w, but not to BCL2A1, ABT-263 did sensitize MDA-MB-231BR to 5-FU to a level comparable to that in MDA-MB-231 at a dose of 5 µM, at which ABT-263 may disrupt intracellular BCL2A1 protein interactions. More importantly, BCL2A1 siRNA sensitized MDA-MB-231BR to 5-FU, whereas the overexpression of BCL2A1 conferred 5-FU-resistance on MDA-MB-231. These results indicate that BCL2A1 is a key contributor to the intrinsic 5-FU-resistance in MDA-MB-231BR. It is interesting to note that the drug sensitivity of MDA-MB-231BR was distinct from that of MDA-MB-231SCP2 even though they have the same origin (MDA-MB-231). Further investigations pertinent to the present findings may provide valuable insight into the breast cancer brain metastasis.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Fluorouracil/toxicity , Aniline Compounds/toxicity , Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Celecoxib/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/toxicity , Female , Fluorouracil/therapeutic use , Gene Expression/drug effects , Humans , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitriles/toxicity , Protein Interaction Maps/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Sulfonamides/toxicity , Sulfones/toxicity
16.
Environ Toxicol Pharmacol ; 45: 227-34, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27327526

ABSTRACT

Pathophysiological changes in diabetes like hyperglycemia, oxidative stress, insulin resistance and compensatory hyperinsulinemia predispose cells to malignant transformation and damage DNA repair mechanism. This study was designed to explore the potential synergistic toxic effects of anti-diabetic drug (Metformin), and an analgesic drug (Celecoxib) at cellular level. MTT assay run on Vero cell line revealed that the combinations of Metformin and Celecoxib augment the anti-proliferative effects, whereas Single cell gel electrophoresis spotlighted that Metformin produce non-significant DNA damage with the threshold concentration of 400µg/ml in peripheral blood mononuclear cells (lymphocytes and monocytes), while Celecoxib produced significant (P<0.05) DNA damage (class III comets) above the concentration of 75µg/ml, however the DNA damage or DNA tail protrusions by combinations of both drugs were less than what was observed with Celecoxib alone. Metformin or Celecoxib did not appear mutagenic against any mutant strains (TA 100 and TA 98) but their combination exhibited slight mutagenicity at much higher concentration. The results obtained at concentrations higher than the therapeutic level of drugs and reflect that Metformin in combination with Celecoxib synergistically inhibits the cell proliferation in a concentration dependent pattern. Since, this increase in cytotoxicity did not confer an increase in DNA damage; this combination could be adopted to inhibit the growth of malignant cell without producing any genotoxic or mutagenic effects at cellular level.


Subject(s)
Celecoxib/toxicity , Cell Proliferation/drug effects , DNA Damage , Metformin/toxicity , Animals , Celecoxib/administration & dosage , Cell Survival/drug effects , Chlorocebus aethiops , Comet Assay , Dose-Response Relationship, Drug , Drug Synergism , Metformin/administration & dosage , Microscopy, Fluorescence , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Vero Cells
17.
Toxicol In Vitro ; 32: 26-40, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26689325

ABSTRACT

Cyclooxygenase-2 (COX-2) inhibitors (coxibs) are non-steroidal anti-inflammatory drugs (NSAIDs) designed to selectively inhibit COX-2. However, drugs of this therapeutic class are associated with drug induced liver injury (DILI) and mitochondrial injury is likely to play a role. The effects of selective COX-2 inhibitors on inhibition of oxidative phosphorylation (ATP synthesis) in rat liver mitochondria were investigated. The order of potency of inhibition of ATP synthesis was: lumiracoxib (IC50: 6.48 ± 2.74 µM)>celecoxib (IC50: 14.92 ± 6.40 µM)>valdecoxib (IC50: 161.4 ± 28.6 µM)>rofecoxib (IC50: 238.4 ± 79.2 µM)>etoricoxib (IC50: 405.1 ± 116.3 µM). Mechanism based inhibition of ATP synthesis (Kinact 0.078 min(-1) and KI 21.46 µM and Kinact/KI ratio 0.0036 min(-1)µM(-1)) was shown by lumiracoxib and data suggest that the opening of the MPT pore may not be the mechanism of toxicity. A positive correlation (with r(2)=0.921) was observed between the potency of inhibition of ATP synthesis and the log P values. The in vitro metabolism of coxibs in rat liver mitochondria yielded for each drug substance a major single metabolite and identified a hydroxy metabolite with each of the coxibs and these metabolites did not alter the inhibition profile of ATP synthesis of the parent compound. The results suggest that coxibs themselves could be involved in the hepatotoxic action through inhibition of ATP synthesis.


Subject(s)
Adenosine Triphosphate/metabolism , Cyclooxygenase 2 Inhibitors/toxicity , Mitochondria, Liver/drug effects , Animals , Celecoxib/toxicity , Diclofenac/analogs & derivatives , Diclofenac/toxicity , Etoricoxib , Isoxazoles/toxicity , Lactones/toxicity , Male , Mitochondria, Liver/metabolism , Oxidative Phosphorylation/drug effects , Pyridines/toxicity , Rats, Sprague-Dawley , Sulfonamides/toxicity , Sulfones/toxicity
18.
Drug Deliv ; 23(2): 420-8, 2016.
Article in English | MEDLINE | ID: mdl-24870199

ABSTRACT

Uniform mesoporous carbon spheres (UMCS) were used as a carrier to improve the bioavailability of the model drug, celecoxib (CEL). Furthermore, we investigated the mechanism responsible for the improved bioavailability of CEL. The association, adhesion and uptake of UMCS by intestinal epithelial cells were studied by transmission electron microscopy (TEM), fluorescence-activated cell sorting (FACS) and laser confocal scanning microscopy (LCSM). UMCS was found to promote cellular uptake of CEL. Drug transport in Caco-2 cell monolayers proved that UMCS can significantly reduce the rate of drug efflux and improve CEL permeability. The dissolution rate of CEL from drug-loaded samples was markedly improved compared with pure crystalline CEL; moreover, oral bioavailability of CEL loaded into UMCS was also markedly improved compared with that of commercially available capsules. UMCS indicates the advantages and potential of this method to achieve improved oral absorption by increasing the dissolution rate, cellular uptake and permeability of the drug.


Subject(s)
Carbon/chemistry , Celecoxib/administration & dosage , Cyclooxygenase 2 Inhibitors/administration & dosage , Drug Carriers , Intestinal Absorption , Intestinal Mucosa/metabolism , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Calorimetry, Differential Scanning , Carbon/toxicity , Celecoxib/chemistry , Celecoxib/pharmacokinetics , Celecoxib/toxicity , Crystallography, X-Ray , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/pharmacokinetics , Cyclooxygenase 2 Inhibitors/toxicity , Dogs , Drug Compounding , Flow Cytometry , Humans , Microscopy, Confocal , Microscopy, Electron, Transmission , Permeability , Porosity , Powder Diffraction , Solubility , Technology, Pharmaceutical/methods
19.
Drug Des Devel Ther ; 9: 4105-13, 2015.
Article in English | MEDLINE | ID: mdl-26251576

ABSTRACT

Celecoxib, a selective cyclooxygenase-2 inhibitor, is potentially useful for the treatment of colonic diseases such as colorectal cancer and colitis. However, the cardiovascular toxicity of celecoxib limits its routine use in the clinic. Generally, colon-specific delivery of a drug both increases the therapeutic availability in the large intestine and decreases the systemic absorption of the drug, most likely resulting in enhanced therapeutic effects against colonic diseases such as colitis and reduced systemic side effects. To develop a colon-specific prodrug of celecoxib that could reduce its cardiovascular toxicity and improve its therapeutic activity, dextran-glutamic acid-celecoxib conjugate (glutam-1-yl celecoxib-dextran ester [G1CD]) was prepared and evaluated. While stable in pH 1.2 and 6.8 buffer solutions and small-intestinal contents, G1CD efficiently released celecoxib in cecal contents. Oral administration of G1CD to rats delivered a larger amount of celecoxib to the large intestine than free celecoxib. G1CD prevented the systemic absorption of celecoxib and did not decrease the serum level of 6-ketoprostaglandin F1α, an inverse indicator of cardiovascular toxicity of celecoxib. Collectively, G1CD may be a polymeric colon-specific celecoxib prodrug with therapeutic and toxicological advantages.


Subject(s)
Celecoxib/metabolism , Colon/metabolism , Cyclooxygenase 2 Inhibitors/metabolism , Dextrans/metabolism , Drug Carriers , Glutamates/metabolism , Intestinal Absorption , Prodrugs/metabolism , 6-Ketoprostaglandin F1 alpha/blood , Administration, Oral , Animals , Buffers , Cecum/metabolism , Celecoxib/administration & dosage , Celecoxib/analogs & derivatives , Celecoxib/chemical synthesis , Celecoxib/toxicity , Chemistry, Pharmaceutical , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/toxicity , Dextrans/administration & dosage , Dextrans/chemical synthesis , Dextrans/toxicity , Drug Stability , Glutamates/administration & dosage , Glutamates/chemical synthesis , Glutamates/toxicity , Hydrogen-Ion Concentration , Intestine, Small/metabolism , Male , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Prodrugs/toxicity , Rats, Sprague-Dawley , Technology, Pharmaceutical/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...