Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Publication year range
1.
Mol Biol Rep ; 51(1): 859, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39066934

ABSTRACT

BACKGROUND: Doxorubicin (DOX) is a potent chemotherapy widely used in treating various neoplastic diseases. However, the clinical use of DOX is limited due to its potential toxic effect on the cardiovascular system. Thus, identifying the pathway involved in this toxicity may help minimize chemotherapy risk and improve cancer patients' quality of life. Recent studies suggest that Endothelial-to-Mesenchymal transition (EndMT) and endothelial toxicity contribute to the pathogenesis of DOX-induced cardiovascular toxicity. However, the molecular mechanism is yet unknown. Given that arachidonic acid and associated cytochrome P450 (CYP) epoxygenase have been involved in endothelial and cardiovascular function, we aimed to examine the effect of suppressing CYP epoxygenases on DOX-induced EndMT and cardiovascular toxicity in vitro and in vivo. METHODS AND RESULTS: To test this, human endothelial cells were treated with DOX, with or without CYP epoxygenase inhibitor, MSPPOH. We also investigated the effect of MSPPOH on the cardiovascular system in our zebrafish model of DOX-induced cardiotoxicity. Our results showed that MSPPOH exacerbated DOX-induced EndMT, inflammation, oxidative stress, and apoptosis in our endothelial cells. Furthermore, we also show that MSPPOH increased cardiac edema, lowered vascular blood flow velocity, and worsened the expression of EndMT and cardiac injury markers in our zebrafish model of DOX-induced cardiotoxicity. CONCLUSION: Our data indicate that a selective CYP epoxygenase inhibitor, MSPPOH, induces EndMT and endothelial toxicity to contribute to DOX-induced cardiovascular toxicity.


Subject(s)
Cardiotoxicity , Cytochrome P-450 Enzyme System , Doxorubicin , Epithelial-Mesenchymal Transition , Oxidative Stress , Zebrafish , Doxorubicin/adverse effects , Animals , Humans , Cardiotoxicity/metabolism , Cardiotoxicity/etiology , Epithelial-Mesenchymal Transition/drug effects , Cytochrome P-450 Enzyme System/metabolism , Oxidative Stress/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Apoptosis/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism
2.
Cell Mol Biol Lett ; 29(1): 33, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38448800

ABSTRACT

Gut microbiota regulates various aspects of human physiology by producing metabolites, metabolizing enzymes, and toxins. Many studies have linked microbiota with human health and altered microbiome configurations with the occurrence of several diseases, including cancer. Accumulating evidence suggests that the microbiome can influence the initiation and progression of several cancers. Moreover, some microbiotas of the gut and oral cavity have been reported to infect tumors, initiate metastasis, and promote the spread of cancer to distant organs, thereby influencing the clinical outcome of cancer patients. The gut microbiome has recently been reported to interact with environmental factors such as diet and exposure to environmental toxicants. Exposure to environmental pollutants such as polycyclic aromatic hydrocarbons (PAHs) induces a shift in the gut microbiome metabolic pathways, favoring a proinflammatory microenvironment. In addition, other studies have also correlated cancer incidence with exposure to PAHs. PAHs are known to induce organ carcinogenesis through activating a ligand-activated transcriptional factor termed the aryl hydrocarbon receptor (AhR), which metabolizes PAHs to highly reactive carcinogenic intermediates. However, the crosstalk between AhR and the microbiome in mediating carcinogenesis is poorly reviewed. This review aims to discuss the role of exposure to environmental pollutants and activation of AhR on microbiome-associated cancer progression and explore the underlying molecular mechanisms involved in cancer development.


Subject(s)
Environmental Pollutants , Microbiota , Neoplasms , Humans , Receptors, Aryl Hydrocarbon , Carcinogenesis , Tumor Microenvironment
3.
Chem Res Toxicol ; 36(3): 552-560, 2023 03 20.
Article in English | MEDLINE | ID: mdl-36877625

ABSTRACT

Patients with sepsis are at a high risk of morbidity and mortality due to multiple organ injuries caused by pathological inflammation. Although sepsis is accompanied by multiple organ injuries, acute renal injury is a significant contributor to sepsis morbidity and mortality. Thus, dampening inflammation-induced renal injury may limit severe consequences of sepsis. As several studies have suggested that 6-formylindolo(3,2-b)carbazole (FICZ) is beneficial for treating various inflammatory diseases, we aimed to examine the potential protective effect of FICZ on the acute endotoxin-induced sepsis model of kidney injury. To test this, male C57Bl/6N mice were injected with FICZ (0.2 mg/kg) or vehicle 1 h prior to an injection of either lipopolysaccharides (LPS) (10 mg/kg), to induce sepsis, or phosphate-buffered saline for 24 h. Thereafter, gene expression of kidney injury and pro-inflammatory markers, circulating cytokines and chemokines, and kidney morphology were assessed. Our results show that FICZ reduced LPS-induced acute injury in kidneys from LPS-injected mice. Furthermore, we found that FICZ dampens both renal and systemic inflammation in our sepsis model. Mechanistically, our data indicated that FICZ significantly upregulates NAD(P)H quinone oxidoreductase 1 and heme oxygenase 1 via aryl hydrocarbon receptor (AhR) and nuclear factor erythroid 2-related factor 2 (Nrf2) in the kidneys to lessen inflammation and improve septic acute kidney injury. Overall, the data of our study show that FICZ possesses a beneficial reno-protective effect against sepsis-induced renal injury via dual activation of AhR/Nrf2.


Subject(s)
Acute Kidney Injury , Sepsis , Animals , Male , Mice , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Carbazoles/pharmacology , Endotoxins , Inflammation/chemically induced , Inflammation/drug therapy , Kidney/metabolism , Lipopolysaccharides , NF-E2-Related Factor 2 , Receptors, Aryl Hydrocarbon/metabolism , Sepsis/chemically induced , Sepsis/drug therapy
4.
Circulation ; 143(22): 2188-2204, 2021 06.
Article in English | MEDLINE | ID: mdl-33832341

ABSTRACT

BACKGROUND: SGLT2 (sodium/glucose cotransporter 2) inhibitors exert robust cardioprotective effects against heart failure in patients with diabetes, and there is intense interest to identify the underlying molecular mechanisms that afford this protection. Because the induction of the late component of the cardiac sodium channel current (late-INa) is involved in the etiology of heart failure, we investigated whether these drugs inhibit late-INa. METHODS: Electrophysiological, in silico molecular docking, molecular, calcium imaging, and whole heart perfusion techniques were used to address this question. RESULTS: The SGLT2 inhibitor empagliflozin reduced late-INa in cardiomyocytes from mice with heart failure and in cardiac Nav1.5 sodium channels containing the long QT syndrome 3 mutations R1623Q or ΔKPQ. Empagliflozin, dapagliflozin, and canagliflozin are all potent and selective inhibitors of H2O2-induced late-INa (half maximal inhibitory concentration = 0.79, 0.58, and 1.26 µM, respectively) with little effect on peak sodium current. In mouse cardiomyocytes, empagliflozin reduced the incidence of spontaneous calcium transients induced by the late-INa activator veratridine in a similar manner to tetrodotoxin, ranolazine, and lidocaine. The putative binding sites for empagliflozin within Nav1.5 were investigated by simulations of empagliflozin docking to a three-dimensional homology model of human Nav1.5 and point mutagenic approaches. Our results indicate that empagliflozin binds to Nav1.5 in the same region as local anesthetics and ranolazine. In an acute model of myocardial injury, perfusion of isolated mouse hearts with empagliflozin or tetrodotoxin prevented activation of the cardiac NLRP3 (nuclear-binding domain-like receptor 3) inflammasome and improved functional recovery after ischemia. CONCLUSIONS: Our results provide evidence that late-INa may be an important molecular target in the heart for the SGLT2 inhibitors, contributing to their unexpected cardioprotective effects.


Subject(s)
Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Sodium Channels/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Animals , Benzhydryl Compounds/therapeutic use , Glucosides/therapeutic use , Humans , Male , Mice , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
5.
Cell Mol Biol Lett ; 27(1): 103, 2022 Nov 22.
Article in English | MEDLINE | ID: mdl-36418969

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcriptional factor that mediates the toxicities of several environmental pollutants. Decades of research have been carried out to understand the role of AhR as a novel mechanism for disease development. Its involvement in the pathogenesis of cancer, cardiovascular diseases, rheumatoid arthritis, and systemic lupus erythematosus have long been known. One of the current hot research topics is investigating the role of AhR activation by environmental pollutants on glucose homeostasis and insulin secretion, and hence the pathogenesis of diabetes mellitus. To date, epidemiological studies have suggested that persistent exposure to environmental contaminants such as dioxins, with subsequent AhR activation increases the risk of specific comorbidities such as obesity and diabetes. The importance of AhR signaling in various molecular pathways highlights that the role of this receptor is far beyond just xenobiotic metabolism. The present review aims at providing significant insight into the physiological and pathological role of AhR and its regulated enzymes, such as cytochrome P450 1A1 (CYP1A1) and CYP1B1 in both types of diabetes. It also provides a comprehensive summary of the current findings of recent research studies investigating the role of the AhR/CYP1A1 pathway in insulin secretion and glucose hemostasis in the pancreas, liver, and adipose tissues. This review further highlights the molecular mechanisms involved, such as gluconeogenesis, hypoxia-inducible factor (HIF), oxidative stress, and inflammation.


Subject(s)
Diabetes Mellitus , Environmental Pollutants , Insulin Resistance , Humans , Receptors, Aryl Hydrocarbon/genetics , Cytochrome P-450 CYP1A1 , Glucose , Homeostasis
6.
Am J Physiol Heart Circ Physiol ; 320(1): H432-H446, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33185110

ABSTRACT

Following cardiac injury, increased adrenergic drive plays an important role in compensating for reduced cardiac function. However, chronic excess adrenergic stimulation can be detrimental to cardiac pathophysiology and can also affect other organs including adipose tissue, leading to increased lipolysis. Interestingly, inhibition of adipose triglyceride lipase (ATGL), a rate-limiting enzyme in lipolysis, in adipocytes ameliorates cardiac dysfunction in a heart failure model. Thus, we investigated whether inhibition of adipocyte ATGL can mitigate the adverse cardiac effects of chronic adrenergic stimulation and explored the underlying mechanisms. To do this, isoproterenol (ISO) was continuously administered to C57Bl/6N mice for 2 wk with or without an ATGL inhibitor (Atglistatin). We found that Atglistatin alleviated ISO-induced cardiac remodeling and reduced ISO-induced upregulation of galectin-3, a marker of activated macrophages and a potent inducer of fibrosis, in white adipose tissue (WAT), heart, and the circulation. To test whether the beneficial effects of Atglistatin occur via inhibition of adipocyte ATGL, adipocyte-specific ATGL knockout (atATGL-KO) mice were utilized for similar experiments. Subsequently, the same cardioprotective effects of atATGL-KO following ISO administration were observed. Furthermore, Atglistatin and atATGL-KO abolished ISO-induced galectin-3 secretion from excised WAT. We further demonstrated that activation of cardiac fibroblasts by the conditioned media of ISO-stimulated WAT is galectin-3-dependent. In conclusion, the inhibition of adipocyte ATGL ameliorated ISO-induced cardiac remodeling possibly by reducing galectin-3 secretion from adipose tissue. Thus, inhibition of adipocyte ATGL might be a potential target to prevent some of the adverse effects of chronic excess adrenergic drive.NEW & NOTEWORTHY The reduction of lipolysis by adipocyte ATGL inhibition ameliorates cardiac remodeling induced by chronic ß-adrenergic stimulation likely via reducing galectin-3 secretion from adipose tissue. Our findings highlight that suppressing lipolysis in adipocytes may be a potential therapeutic target for patients with heart failure whose sympathetic nervous system is activated. Furthermore, galectin-3 might be involved in the mechanisms by which excessive lipolysis in adipose tissues influences remote cardiac pathologies and thus warrants further investigation.


Subject(s)
Adipose Tissue, White/drug effects , Anti-Inflammatory Agents/pharmacology , Enzyme Inhibitors/pharmacology , Heart Diseases/prevention & control , Inflammation Mediators/metabolism , Isoproterenol , Lipase/antagonists & inhibitors , Phenylurea Compounds/pharmacology , Ventricular Remodeling/drug effects , Adipose Tissue, White/enzymology , Animals , Cells, Cultured , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Galectin 3/metabolism , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/physiopathology , Lipase/metabolism , Lipolysis/drug effects , Male , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Paracrine Communication , Signal Transduction
7.
Arch Toxicol ; 95(1): 1-9, 2021 01.
Article in English | MEDLINE | ID: mdl-32852568

ABSTRACT

Doxorubicin (DOX) is a powerful broad-spectrum anti-neoplastic anthracycline antibiotic. However, DOX may cause a dose-dependent cardiotoxicity that can eventually progress to congestive heart failure and death. Numerous molecular mechanisms have been implicated in the cardiotoxic effect of DOX including topoisomerase IIß and generation of free radicals. However, targeting these pathways appears to be insufficient to mitigate the cardiotoxic effects of DOX and/or ultimately reduces the anti-tumor activity of DOX. Thus, there remains a crucial need to identify novel pharmacological targets that can alleviate the cardiotoxic effects of DOX without reducing its anti-tumor activity. Recent studies have suggested that the Nucleotide-Binding Domain-Like Receptor Protein 3 (NLRP3) inflammasome is implicated in tumor progression and the chemoresistance of cancer cells to DOX. Of interest, reducing NLRP3 inflammasome activity alleviates DOX-induced cardiotoxicity. Therefore, we postulate that strategies that target the NLRP3 inflammasome can help mitigate the cardiotoxic effects of DOX while maintaining and/or even enhancing its anti-cancer activity. Herein, we review the current knowledge about the potential implication of the NLRP3 inflammasome in the anti-cancer and cardiotoxic effects of DOX.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibiotics, Antineoplastic/adverse effects , Doxorubicin/adverse effects , Heart Diseases/prevention & control , Inflammasomes/antagonists & inhibitors , Myocytes, Cardiac/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Animals , Cardiotoxicity , Drug Resistance, Neoplasm , Heart Diseases/chemically induced , Heart Diseases/immunology , Heart Diseases/metabolism , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Molecular Targeted Therapy , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction
8.
Inflammopharmacology ; 29(1): 269-279, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32564182

ABSTRACT

BACKGROUND: Sepsis-induced systemic inflammation response syndrome is the leading cause of morbidity and mortality among patients in intensive care units in North America. While sepsis is associated with multiple organ damage, acute renal injury represents a hallmark of sepsis. Since systemic and renal inflammation is known to play a vital role in morbidity and mortality associated with sepsis, identifying a potent anti-inflammatory agent may help minimize morbidity and mortality associated with acute septic kidney injury. Since recent work has suggested that empagliflozin, a renal sodium-glucose cotransporter 2 (SGLT2) inhibitor, may assist in the treatment of inflammatory diseases, our objective was to examine the effect of empagliflozin on acute sepsis-induced renal injury. METHOD: Mice were treated with three daily doses of empagliflozin or vehicle, with lipopolysaccharide (LPS) administered on the third day, at the same time as the third dose of empagliflozin or vehicle. In another cohort, mice were injected with a single dose of LPS 3 h before a dose of empagliflozin. RESULTS: Our results show that empagliflozin improves survival in a mouse model of LPS-induced septic shock. We further demonstrate that the beneficial effects of empagliflozin are likely mediated via reducing LPS-induced acute renal injury. Moreover, our data indicate that empagliflozin significantly reduces systemic and renal inflammation to contribute to the improvements observed in an LPS-model of acute septic renal injury. CONCLUSION: Overall, the findings of this study suggest that empagliflozin could be repurposed to reduce morbidity and mortality in patients with acute septic renal injury. TRIAL REGISTRATION: Not applicable.


Subject(s)
Acute Kidney Injury/prevention & control , Anti-Inflammatory Agents/pharmacology , Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Inflammation/drug therapy , Acute Kidney Injury/etiology , Animals , Anti-Inflammatory Agents/administration & dosage , Benzhydryl Compounds/administration & dosage , Disease Models, Animal , Glucosides/administration & dosage , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Sepsis/complications , Sepsis/drug therapy , Shock, Septic/complications , Shock, Septic/drug therapy , Sodium-Glucose Transporter 2 Inhibitors/administration & dosage , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
9.
BMC Cancer ; 20(1): 751, 2020 Aug 12.
Article in English | MEDLINE | ID: mdl-32787791

ABSTRACT

BACKGROUND: The survival rates of women with breast cancer have improved significantly over the last four decades due to advances in breast cancer early diagnosis and therapy. However, breast cancer survivors have an increased risk of cardiovascular complications following chemotherapy. While this increased risk of later occurring structural cardiac remodeling and/or dysfunction has largely been attributed to the cardiotoxic effects of breast cancer therapies, the effect of the breast tumor itself on the heart prior to cancer treatment has been largely overlooked. Thus, the objectives of this study were to assess the cardiac phenotype in breast cancer patients prior to cancer chemotherapy and to determine the effects of human breast cancer cells on cardiomyocytes. METHODS: We investigated left ventricular (LV) function and structure using cardiac magnetic resonance imaging in women with breast cancer prior to systemic therapy and a control cohort of women with comparable baseline factors. In addition, we explored how breast cancer cells communicate with the cardiomyocytes using cultured human cardiac and breast cancer cells. RESULTS: Our results indicate that even prior to full cancer treatment, breast cancer patients already exhibit relative LV hypertrophy (LVH). We further demonstrate that breast cancer cells likely contribute to cardiomyocyte hypertrophy through the secretion of soluble factors and that at least one of these factors is endothelin-1. CONCLUSION: Overall, the findings of this study suggest that breast cancer cells play a greater role in inducing structural cardiac remodeling than previously appreciated and that tumor-derived endothelin-1 may play a pivotal role in this process.


Subject(s)
Breast Neoplasms/complications , Cell Communication/physiology , Endothelin-1/metabolism , Hypertrophy, Left Ventricular/etiology , Myocytes, Cardiac/physiology , Breast Neoplasms/blood , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Case-Control Studies , Cell Line, Tumor , Cells, Cultured , Culture Media, Conditioned/metabolism , Endothelin-1/blood , Female , Humans , Hypertrophy/etiology , Magnetic Resonance Imaging , Middle Aged , Myocytes, Cardiac/pathology , Paracrine Communication , Retrospective Studies , Tumor Cells, Cultured , Ventricular Remodeling
10.
Inflamm Res ; 69(11): 1073-1076, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32860528

ABSTRACT

PURPOSE: Despite the fact that the risk versus benefit of smoking cannabis has not been extensively studied, many individuals with multiple sclerosis are smoking cannabis to reduce their pain intensity and spasticity. The lack of information about inhaled cannabis might be attributed to the fact that most trials focus on orally administered cannabis. Given the fact that the administration of cannabis via inhalation is known to rapidly deliver cannabinoids with a higher total bioavailability than what can be achieved through oral or buccal routes, it is important to understand the clinical trials conducted using smoked cannabis on patients with multiple sclerosis. METHODS: We sought to discuss the relevant literature about the safety and efficacy of smoked cannabis in multiple sclerosis patients in order to further understand the risks and benefits of this potential therapy for this patient population. RESULTS: The current knowledge about the potential effects of smoked cannabis on treating neuropathic pain associated with multiple sclerosis is reviewed. In addition, we discuss the possible adverse effects associated with smoking cannabis and we suggest safer as well as new effective inhaled cannabis formulations for the treatment of neuropathic pain associated with multiple sclerosis.


Subject(s)
Marijuana Smoking , Medical Marijuana/therapeutic use , Multiple Sclerosis/drug therapy , Neuralgia/drug therapy , Administration, Inhalation , Cannabis , Cognitive Dysfunction/etiology , Humans , Medical Marijuana/adverse effects
11.
Inflamm Res ; 69(6): 549-558, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32239248

ABSTRACT

PURPOSE: Cannabis has been used for thousands of years in many cultures for the treatment of several ailments including pain. The benefits of cannabis are mediated largely by cannabinoids, the most prominent of which are tetrahydrocannabinol (THC) and cannabidiol (CBD). As such, THC and/or CBD have been investigated in clinical studies for the treatment of many conditions including neuropathic pain and acute or chronic inflammation. While a plethora of studies have examined the biochemical effects of purified THC and/or CBD, only a few have focused on the effects of full-spectrum cannabis plant extract. Accordingly, studies using purified THC or CBD may not accurately reflect the potential health benefits of full-spectrum cannabis extracts. Indeed, the cannabis plant produces a wide range of cannabinoids, terpenes, flavonoids, and other bioactive molecules which are likely to contribute to the different biological effects. The presence of all these bioactive molecules in cannabis extracts has garnered much attention of late especially with regard to their potential role in the treatment of neuropathic pain associated with multiple sclerosis. METHODS: Literature review was performed to further understand the effect of clinically used full-spectrum cannabis extract in patients with multiple sclerosis. RESULTS: Herein, the current knowledge about the potential beneficial effects of existing products of full-spectrum cannabis extract in clinical studies involving patients with multiple sclerosis is extensively reviewed. In addition, the possible adverse effects associated with cannabis use is discussed along with how the method of extraction and the delivery mechanisms of different cannabis extracts contribute to the pharmacokinetic and biological effects of full-spectrum cannabis extracts.Herein, the current knowledge about the potential beneficial effects of existing products of full-spectrum cannabis extract in clinical studies involving patients with multiple sclerosis is extensively reviewed. In addition, the possible adverse effects associated with cannabis use is discussed along with how the method of extraction and the delivery mechanisms of different cannabis extracts contribute to the pharmacokinetic and biological effects of full-spectrum cannabis extracts.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Cannabinoids/therapeutic use , Cannabis , Multiple Sclerosis/drug therapy , Neuralgia/drug therapy , Plant Extracts/therapeutic use , Humans
12.
J Cardiovasc Pharmacol ; 74(3): 235-245, 2019 09.
Article in English | MEDLINE | ID: mdl-31306370

ABSTRACT

A plethora of studies have demonstrated that cardiomyopathy represents a serious source of morbidity and mortality in patients with diabetes. Yet, the underlying mechanisms of diabetic cardiomyopathy are still poorly understood. Of interest, cytochrome P450 2J (CYP2J) and soluble epoxide hydrolase (sEH) are known to control the maintenance of cardiovascular health through the regulation of cardioprotective epoxyeicosatrienoic acids (EETs) and its less active products, dihydroxyeicosatrienoic acids (DHETs). Therefore, we examined the role of the aforementioned pathway in the development of diabetic cardiomyopathy. Our diabetic model initiated cardiomyopathy as indexed by the increase in the expression of hypertrophic markers such as NPPA. Furthermore, diabetic cardiomyopathy was associated with a low level of cardiac EETs and an increase of the DHETs/EETs ratio both in vivo and in cardiac cells. The modulation in EETs and DHETs was attributed to the increase of sEH and the decrease of CYP2J. Interestingly, the reduction of sEH attenuates cardiotoxicity mediated by high glucose in cardiac cells. Mechanistically, the beneficial effect of sEH reduction might be due to the decrease of phosphorylated ERK1/2 and p38. Overall, the present work provides evidence that diabetes initiates cardiomyopathy through the increase in sEH, the reduction of CYP2J, and the decrease of cardioprotective EETs.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 2/enzymology , Diabetic Cardiomyopathies/enzymology , Eicosanoids/metabolism , Epoxide Hydrolases/metabolism , Myocytes, Cardiac/enzymology , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Blood Glucose/metabolism , Cell Line , Cytochrome P-450 Enzyme System/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/genetics , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Diet, High-Fat , Epoxide Hydrolases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Mice, Inbred C57BL , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Obesity/complications , Obesity/enzymology , Phosphorylation , Signal Transduction , Streptozocin , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Toxicol Mech Methods ; 29(2): 119-127, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30273082

ABSTRACT

Although a plethora of studies have examined tobacco smoke-cancer disease association, the involvement of cellular genetic toxicity remains unclear. Therefore, the present study provides molecular evidence for a pathway involved in the DNA damage induced by long-term cigarette and waterpipe smoke in human subjects. The study population consisted of 45 subjects who were divided into three groups; healthy nonsmokers group, cigarette smokers group, and waterpipe smokers group. A questionnaire and consent form was distributed and signed by all participants. Total RNA was extracted from the blood using PAXgene Blood RNA Kit and mRNA expression levels of target genes were quantified by RT-PCR. Our results showed that 80% of the participants smoke 20-39 cigarettes/day, whereas 12% smoke more than 40 cigarettes/day. With regard to waterpipe smoke, the majority (46%) smoke more than 5 times/week. Both cigarette and waterpipe smokers showed increased the plasma levels 8-hydroxy-2'-deoxyguanosine (8-OHdG), of DNA damage marker. In addition, the mRNA expression levels of DNA repair genes (OGG1 and XRCC1) were significantly inhibited in both cigarette and waterpipe smokers groups by 30% and 60%, respectively. This was associated with a marked decrease (50%) in the expression of detoxifying genes (NQO1 and GSTA1) with an increase in CYP1A1 mRNA expression, a cancer-activating gene. Both cigarette and waterpipe smokers increased in the plasma concentrations of several toxic heavy metals such as Cd (130%), Pb (47%), and Ni (30%). In conclusion: the present findings clearly explore the genotoxic effect of cigarette and waterpipe smoking on human DNA.


Subject(s)
Cigarette Smoking/adverse effects , DNA Damage , Inhalation Exposure/adverse effects , Oxidative Stress , Smoke/adverse effects , Smokers , Water Pipe Smoking/adverse effects , 8-Hydroxy-2'-Deoxyguanosine , Adult , Biomarkers/blood , Case-Control Studies , Cigarette Smoking/blood , Cigarette Smoking/genetics , Cytochrome P-450 CYP1A1/blood , Cytochrome P-450 CYP1A1/genetics , DNA Glycosylases/blood , DNA Glycosylases/genetics , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/blood , Female , Gene Expression Regulation, Enzymologic , Glutathione Transferase/blood , Glutathione Transferase/genetics , Healthy Volunteers , Humans , Male , Middle Aged , NAD(P)H Dehydrogenase (Quinone)/blood , NAD(P)H Dehydrogenase (Quinone)/genetics , Risk Assessment , Time Factors , Transcriptome , Water Pipe Smoking/blood , Water Pipe Smoking/genetics , X-ray Repair Cross Complementing Protein 1/blood , X-ray Repair Cross Complementing Protein 1/genetics , Young Adult
14.
J Mol Cell Cardiol ; 125: 162-173, 2018 12.
Article in English | MEDLINE | ID: mdl-30381233

ABSTRACT

Numerous epidemiological studies have demonstrated that approximately 40% of myocardial infarctions (MI) are associated with heart failure (HF). Resveratrol, a naturally occurring polyphenol, has been shown to be beneficial in the treatment of MI-induced HF in rodent models. However, the mechanism responsible for the effects of resveratrol are poorly understood. Interestingly, resveratrol is known to inhibit cytochrome P450 1B1 (CYP1B1) which is involved in the formation of cardiotoxic hydroxyeicosatetraenoic acid (HETE) metabolites. Therefore, we investigated whether resveratrol could improve MI-induced cardiac remodeling and HF in rats through the inhibition of CYP1B1 and its metabolites. To do this, rats were subjected to either sham surgery or a surgery to ligate the left anterior descending artery to induce a MI and subsequent HF. Three weeks post-surgery, rats with established HF were treated with control diet or administered a diet containing low dose of resveratrol. Our results showed that low dose resveratrol treatment significantly improves % ejection fraction in MI rats and reduces MI-induced left ventricular and atrial remodeling. Furthermore, non-cardiac symptoms of HF such as reduced physical activity improved with low dose resveratrol treatment. Mechanistically, low dose resveratrol treatment of rats with established HF restored levels of fatty acid oxidation and significantly improved cardiac energy metabolism as well as significantly inhibited CYP1B1 and cardiotoxic HETE metabolites induced in MI rats. Overall, the present work provides evidence that low dose resveratrol reduces the severity of MI-induced HF, at least in part, through the inhibition of CYP1B1 and cardiotoxic HETE metabolites.


Subject(s)
Heart Failure/drug therapy , Heart Failure/etiology , Heart Failure/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Myocardial Infarction/complications , Resveratrol/therapeutic use , Animals , Chromatography, Liquid , Male , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization
15.
J Pharm Pharm Sci ; 21(1s): 29683, 2018.
Article in English | MEDLINE | ID: mdl-29702047

ABSTRACT

PURPOSE: MyoNovin is a novel skeletal muscle-regenerating compound developed through synthesis of two nitro groups onto a guaifenesin backbone to deliver nitric oxide to skeletal muscle with a potential to treat muscle atrophy. The purpose of this study was to utilize in silico, in vitro, and in vivo approaches to characterize MyoNovin and examine its safety, biodistribution, and feasibility for drug delivery. METHODS: In silico software packages were used to predict the physicochemical and biopharmaceutical properties of MyoNovin. In vitro cardiotoxicity was assessed using human cardiomyocytes (RL-14) while effects on CYP3A4 metabolic enzyme and antioxidant activity were examined using commercial kits. A novel HPLC assay was developed to measure MyoNovin concentration in serum, and delineate initial pharmacokinetic and acute toxicity after intravenous administration (20 mg/kg) to male Sprague-Dawley rats. RESULTS: MyoNovin showed relatively high lipophilicity with a LogP value of 3.49, a 20-fold higher skin permeability (19.89 cm/s*107) compared to guaifenesin (0.66 cm/s*107), and ~10-fold higher effective jejunal permeability (2.24 cm/s*104) compared to guaifenesin (0.26 cm/s*104). In vitro, MyoNovinwas not cytotoxic to cardiomyocytes at concentrations below 8 µM and did not inhibit CYP3A4 or show antioxidant activity. In vivo, MyoNovin had a short half-life (t1/2) of 0.16 h, and a volume of distribution Vss of 0.62 L/kg. Biomarkers of MyoNovincardiac and renal toxicity did not differ significantly from baseline control levels. CONCLUSIONS: The predicted high lipophilicity and skin permeability of MyoNovin render it a potential candidate for transdermal administration while its favourable intestinal permeation suggests it may be suitable for oral administration. Pharmacokinetics following IV administration of MyoNovin were delineated for the first time in a rat model. Preliminary single 20 mg/kg dose assessment of MyoNovin suggest no influenceon cardiac troponin or ß-N-Acetylglucosaminidase. This article is open to POST-PUBLICATION REVIEW. Registered readers (see "For Readers") may comment by clicking on ABSTRACT on the issue's contents page.


Subject(s)
Guaifenesin/analogs & derivatives , Muscle, Skeletal/drug effects , Nitrates/pharmacology , Troponin I/blood , Animals , Guaifenesin/administration & dosage , Guaifenesin/pharmacology , Humans , Injections, Intravenous , Male , Muscle, Skeletal/metabolism , Nitrates/administration & dosage , Nitrates/blood , Rats , Rats, Sprague-Dawley
16.
Mol Cell Biochem ; 429(1-2): 151-165, 2017 May.
Article in English | MEDLINE | ID: mdl-28251434

ABSTRACT

Numerous experimental studies have demonstrated the role of cytochrome P450 1B1 (CYP1B1) and its associated mid-chain hydroxyeicosatetraenoic acids (mid-chain HETEs) metabolite in the pathogenesis of cardiac hypertrophy. However, the ability of isoproterenol (ISO) to induce cardiac hypertrophy through mid-chain HETEs has not been investigated yet. Therefore, we hypothesized that ISO induces cardiac hypertrophy through the induction of CYP1B1 and its associated mid-chain HETE metabolites. To test our hypothesis, Sprague-Dawley rats were treated with ISO (5 mg/kg i.p.) for 12 and 72 h whereas, human ventricular cardiomyocytes RL-14 cells were exposed to 100 µM ISO in the presence and absence of 0.5 µM tetramethoxystilbene (TMS) a selective CYP1B1 inhibitor, or 25 nM CYP1B1-siRNA. Moreover, RL-14 cells were transiently transfected with the CRISPR-CYP1B1 plasmid. Thereafter, real-time PCR, western blot analysis, and liquid chromatography-electrospray ionization mass spectroscopy were used to determine the level of gene expression, protein expression, and mid-chain HETEs, respectively. Our results showed that ISO induced CYP1B1 protein expression and the level of cardiac mid-chain HETEs in vivo at pre-hypertrophic and hypertrophic stage. In vitro, inhibition of CYP1B1 using TMS or CYP1B1-siRNA significantly attenuates ISO-induced hypertrophy. Furthermore, overexpression of CYP1B1 significantly induced cellular hypertrophy and mid-chain HETEs metabolite. Mechanistically, the protective effect of TMS against cardiac hypertrophy was mediated through the modulation of superoxide anion, mitogen-activated protein kinases (MAPKs), and nuclear factor-κB (NF-κB). In conclusion, our study provides the first evidence that CYP1B1 and its associated mid-chain HETE metabolites are directly involved in the ISO-induced cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Cytochrome P-450 CYP1B1/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Isoproterenol/adverse effects , Animals , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cell Line , Cytochrome P-450 CYP1B1/genetics , Gene Expression Regulation, Enzymologic/drug effects , Humans , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley
17.
J Cardiovasc Pharmacol ; 70(1): 16-24, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28437282

ABSTRACT

Recent data demonstrated the role of CYP1B1 in cardiovascular disease. It was, therefore, necessary to examine whether the inhibition of CYP1B1 and hence inhibiting the formation of its metabolites, using 2,4,3',5'-tetramethoxystilbene (TMS), would have a cardioprotective effect against angiotensin II (Ang II)-induced cardiac hypertrophy. For this purpose, male Sprague Dawley rats were treated with Ang II with or without TMS (300 µg/kg every third day i.p.). Thereafter, cardiac hypertrophy and the formation of mid-chain HETEs and arachidonic acid were assessed. In vitro, RL-14 cells were treated with Ang II (10 µM) in the presence and absence of TMS (0.5 µM). Then, reactive oxygen species, mitogen-activated protein kinase phosphorylation levels, and nuclear factor-kappa B-binding activity were determined. Our results demonstrated that TMS protects against Ang II-induced cardiac hypertrophy as indicated by the improvement in cardiac functions shown by the echocardiography as well as by reversing the increase in heart weight to tibial length ratio caused by Ang II. In addition, the cardioprotective effect of TMS was associated with a significant decrease in cardiac mid-chain HETEs levels. Mechanistically, TMS inhibited reactive oxygen species formation, the phosphorylation of ERK1/2, p38 mitogen-activated protein kinase, and the binding of p65 NF-κB.


Subject(s)
Angiotensin II/toxicity , Cardiomegaly/metabolism , Cardiomegaly/prevention & control , Cytochrome P-450 CYP1B1/antagonists & inhibitors , Cytochrome P-450 CYP1B1/metabolism , Hydroxyeicosatetraenoic Acids/antagonists & inhibitors , Hydroxyeicosatetraenoic Acids/metabolism , Animals , Cardiomegaly/chemically induced , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cells, Cultured , Humans , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Stilbenes/pharmacology , Stilbenes/therapeutic use , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Pharmacol Res ; 105: 28-43, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26772815

ABSTRACT

Doxorubicin (DOX) has been reported to be a very potent and effective anticancer agent. However, clinical treatment with DOX has been greatly limited due to its cardiotoxicity. Furthermore, several studies have suggested a role for cytochrome P450 1B1 (CYP1B1) and mid-chain hydroxyeicosatetraenoic acids (mid-chain HETEs) in DOX-induced cardiac toxicity. Therefore, we hypothesized that DOX induced cardiotoxicity is mediated through the induction of CYP1B1 and its associated mid-chain HETEs metabolite. To test our hypothesis, Sprague-Dawley rats and RL-14 cells were treated with DOX in the presence and absence of 2,3',4,5'-tetramethoxystilbene (TMS), a selective CYP1B1 inhibitor. Thereafter, cardiotoxicity parameters were determined using echocardiography, histopathology, and gene expression. Further, the level of mid-chain HETEs was quantified using liquid chromatography-electron spray ionization-mass spectrometry. Our results showed that DOX induced cardiotoxicity in vivo and in vitro as evidenced by deleterious changes in echocardiography, histopathology, and hypertrophic markers. Importantly, the TMS significantly reversed these changes. Moreover, the DOX-induced cardiotoxicity was associated with a proportional increase in the formation of cardiac mid-chain HETEs both in vivo and in our cell culture model. Interestingly, the inhibition of cardiotoxicity by TMS was associated with a dramatic decrease in the formation of cardiac mid-chain HETEs suggesting a mid-chain HETEs-dependent mechanism. Mechanistically, the protective effect of TMS against DOX-induced cardiotoxicity was mediated through the inhibition of mitogen activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB). In conclusion, our study provides the first evidence that the inhibition of CYP1B1 and mid-chain HETE formation attenuate DOX-induced cardiotoxicity.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Cardiotoxicity/drug therapy , Cytochrome P-450 CYP1B1/antagonists & inhibitors , Doxorubicin/toxicity , Enzyme Inhibitors/therapeutic use , Hydroxyeicosatetraenoic Acids/metabolism , Stilbenes/therapeutic use , Animals , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Cell Line , Cytochrome P-450 CYP1B1/metabolism , Humans , Male , Myocardium/metabolism , Myocardium/pathology , Rats, Sprague-Dawley
19.
Arch Toxicol ; 90(2): 359-73, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25600587

ABSTRACT

Recent studies have established the role of mid-chain hydroxyeicosatetraenoic acids (HETEs) in the development of cardiovascular disease. Mid-chain HETEs have been reported to have vasoconstrictive and pro-inflammatory effects. However, whether mid-chain HETEs can induce cardiac hypertrophy remains unclear. Therefore, the overall objective of the present study was to elucidate the potential hypertrophic effect of mid-chain HETEs in the human ventricular cardiomyocytes, RL-14 cells, and to explore the mechanisms involved. For this purpose, RL-14 cells were treated with increasing concentrations of mid-chain HETEs (2.5, 5, 10 and 20 µM). Thereafter, the cardiac hypertrophy markers and cell size were determined using real-time polymerase chain reaction and phase contrast imaging, respectively. Phosphorylated mitogen-activated protein kinase (MAPK) level and nuclear factor kappa B (NF-κB) binding activity were determined. Our results showed that mid-chain HETEs induced cellular hypertrophy in RL-14 cells as evidenced by the induction of cardiac hypertrophy markers, α- and ß-myocin heavy chain and atrial and brain natriuretic peptide as well as the increase in cell size. Mechanistically, all mid-chain HETEs were able to induce the binding activity of NF-κB to its responsive element in a HETE-dependent manner, and they significantly induced the phosphorylation of ERK 1/2. The induction of cellular hypertrophy was associated with proportional increase in the formation of dihydroxyeicosatrienoic acids parallel to the increase of soluble epoxide hydrolase enzyme activity. In conclusion, our study provides the first evidence that mid-chain HETEs induce cellular hypertrophy in RL-14 cells through MAPK- and NF-κB-dependent mechanism.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/adverse effects , Cardiomegaly/chemically induced , Hydroxyeicosatetraenoic Acids/adverse effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Arachidonic Acid/metabolism , Atrial Natriuretic Factor/genetics , Cardiomegaly/pathology , Cell Line , Cell Size/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Heart Ventricles/cytology , Humans , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Natriuretic Peptide, Brain/genetics
20.
Arch Toxicol ; 90(1): 119-36, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26525395

ABSTRACT

The incidence, prevalence, and hospitalization rates associated with cardiovascular diseases (CVDs) are projected to increase substantially in the world. Understanding of the biological and pathophysiological mechanisms of survival can help the researchers to develop new management modalities. Numerous experimental studies have demonstrated that mid-chain HETEs are strongly involved in the pathogenesis of the CVDs. Mid-chain HETEs are biologically active eicosanoids that result from the metabolism of arachidonic acid (AA) by both lipoxygenase and CYP1B1 (lipoxygenase-like reaction). Therefore, identifying the localizations and expressions of the lipoxygenase and CYP1B1 and their associated AA metabolites in the cardiovascular system is of major importance in understanding their pathological roles. Generally, the expression of these enzymes is shown to be induced during several CVDs, including hypertension and cardiac hypertrophy. The induction of these enzymes is associated with the generation of mid-chain HETEs and subsequently causation of cardiovascular events. Of interest, inhibiting the formation of mid-chain HETEs has been reported to confer a protection against different cardiac hypertrophy and hypertension models such as angiotensin II, Goldblatt, spontaneously hypertensive rat and deoxycorticosterone acetate (DOCA)-salt-induced models. Although the exact mechanisms of mid-chain HETEs-mediated cardiovascular dysfunction are not fully understood, the present review proposes several mechanisms which include activating G-protein-coupled receptor, protein kinase C, mitogen-activated protein kinases, and nuclear factor kappa B. This review provides a clear understanding of the role of mid-chain HETEs in the pathogenesis of cardiovascular diseases and their importance as novel targets in the treatment for hypertension and cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Cardiovascular System/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Hypertension/metabolism , Signal Transduction , Animals , Cardiomegaly/drug therapy , Cardiomegaly/epidemiology , Cardiomegaly/physiopathology , Cardiovascular Agents/therapeutic use , Cardiovascular System/drug effects , Cardiovascular System/physiopathology , Cytochrome P-450 CYP1B1/antagonists & inhibitors , Cytochrome P-450 CYP1B1/metabolism , Cytochrome P-450 Enzyme Inhibitors/therapeutic use , Drug Design , Humans , Hypertension/drug therapy , Hypertension/epidemiology , Hypertension/physiopathology , Lipoxygenase/metabolism , Lipoxygenase Inhibitors/therapeutic use , Molecular Targeted Therapy , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL