Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Int J Clin Pharmacol Ther ; 56(11): 501-506, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30148451

ABSTRACT

With the increasing popularity of E-cigarettes, chronic exposure to nicotine (NIC) is emerging as a novel risk factor for the kidney. NIC increases oxidative stress in the kidneys, which impairs the viability and function of renal tubular and endothelial cells, alters renal hemodynamics, and compromises overall kidney function. Moreover, long-term NIC exposure increases the risk of development and progression of chronic kidney diseases and may escalate the impact of coexisting morbidities such as obesity-associated renal disease, hypertension, renal transplant status, or the toxicity of various anticancer agents. In this review, we summarize experimental findings describing increased renal risk of chronic NIC exposure and explore therapeutic interventions to alleviate adverse effects of NIC.
.


Subject(s)
Kidney Diseases/chemically induced , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Animals , Electronic Nicotine Delivery Systems , Humans , Kidney Diseases/epidemiology , Kidney Diseases/prevention & control , Risk , Smoking/adverse effects
2.
In Vivo ; 32(5): 1033-1037, 2018.
Article in English | MEDLINE | ID: mdl-30150424

ABSTRACT

BACKGROUND/AIM: We have previously reported that simvastatin exhibits antioxidant properties via extracellular signal-regulated kinase (ERK)/cAMP-response element binding (CREB) protein-dependent induction of heme oxygenase-1 (HO1) and chronic nicotine exposure inhibits ERK/CREB signaling in renal proximal tubule cells (through p66shc). Herein, whether nicotine dampens simvastatin-dependent HO1 induction was determined. MATERIALS AND METHODS: Renal proximal tubule (NRK52E) cells were pre-treated with 200 µM nicotine for 24 h followed by 10 µM simvastatin. Promoter activity of HO1 and manganese superoxide dismutase (MnSOD) and activation of CREB and ERK (via ELK1) were determined in luciferase reporter assays. CREB and p66shc were modulated via genetic means. RESULTS: Nicotine suppressed simvastatin-dependent activation of HO1 and MnSOD promoters and activity of CREB and ELK1 via p66shc. Overexpression of CREB or knockdown of p66shc restored simvastatin-dependent induction of HO1 and MnSOD in the presence of nicotine. CONCLUSION: Antioxidant efficiency of simvastatin might be significantly lessened in smokers/E-cigarette users.


Subject(s)
Antioxidants/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Nicotine/adverse effects , Simvastatin/pharmacology , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Nicotine/administration & dosage , Promoter Regions, Genetic , Rats , Smoking/adverse effects , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcriptional Activation
3.
Int J Clin Pharmacol Ther ; 56(10): 467-475, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29974857

ABSTRACT

Baclofen is a centrally-acting γ-amino butyric acid agonist used mainly in the symptomatic management of spasticity originating from the spinal cord. It is absorbed completely from the gastrointestinal tract, metabolized by the liver to a minor degree, and excreted unchanged by the kidneys. Baclofen is moderately lipophilic and can cross the blood-brain barrier easily. At the usual dosage, it acts mainly at the spinal level without central nervous system (CNS) side effects. During renal failure, however, the elimination of the drug will decrease with a prolonged half-life, resulting in a larger area-under-the-curve exposure and disproportionate CNS toxicity. Clinically, these patients with renal failure may present with a variety of toxic symptoms manifesting at therapeutic/sub-therapeutic doses of baclofen. In cases of unexplained mental status changes in a patient receiving baclofen therapy, a careful assessment of renal function and a high suspicion of baclofen-induced encephalopathy will be key to the diagnosis.
.


Subject(s)
Baclofen/adverse effects , Central Nervous System Diseases/chemically induced , Muscle Relaxants, Central/adverse effects , Renal Insufficiency/complications , Baclofen/administration & dosage , Humans , Muscle Relaxants, Central/administration & dosage
4.
Med Hypotheses ; 108: 128-132, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29055386

ABSTRACT

Despite decades of research, the clinical efficacy of peritoneal dialysis (PD) remains enigmatic. We may wonder why the modality fail in some patients but perhaps the more proper question would be, why it works in so many? We know that the contribution of residual renal function (RRF), more so than in hemodialysis, is critically important to the well-being of many of the patients. Unique features of the modality include the relatively low volume of dialysate fluid needed to provide effective uremic control and the disproportionate tendency for both hypokalemia and hypoalbuminemia, when compared to hemodialysis. It is currently believed that most uremic toxins are generated on the interface of human and bacterial structures in the gastrointestinal tract, the intestinal biota. PD offers disproportionate removal of these toxins upon "first-pass", i.e., via PD fluid exchanges before reaching the systemic circulation beyond the gastrointestinal compartment. Studies examining the net removal gradient of protein-bound uremic toxins during PD are scarce, whereas RRF receives considerably more attention without effective interventions being developed to preserve it. We propose an alternative view on PD, emphasizing the modality's compartmental nature, both for its benefits and the limitations.


Subject(s)
Albumins/metabolism , Kidney/physiopathology , Peritoneal Dialysis , Renal Insufficiency/therapy , Renal Replacement Therapy , Anti-Bacterial Agents/pharmacology , Calcium/metabolism , Communicable Diseases/complications , Gastrointestinal Microbiome , Gastrointestinal Tract/physiopathology , Humans , Hypokalemia/physiopathology , Models, Biological , Obesity/complications , Renal Dialysis , Renal Insufficiency/complications , Uremia/physiopathology
5.
Biologics ; 11: 97-106, 2017.
Article in English | MEDLINE | ID: mdl-28761330

ABSTRACT

AIM: Nephrilin peptide, a designed inhibitor of Rictor complex (mTORC2), exerts pleiotropic protective effects in metabolic, xenobiotic and traumatic stress models. Stress can generate enduring epigenetic changes in gene function. In this work we examine the possibility that nephrilin treatment protects against acute and enduring global changes in oxidative metabolism, with a focus on the Rictor-complex-mediated activation of Rac1, a subunit of NADPH oxidase (Nox) via PKCs, Prex1 and p66shc. METHODS: Given the wide range of animal models in which nephrilin peptide has previously demonstrated effectiveness in vivo, we chose three different experimental systems for this investigation: dermal fibroblasts, renal proximal tubule epithelial cells (PTECs), and kidney tissue and urine from an animal model of burn trauma in which nephrilin was previously shown to prevent loss of kidney function. RESULTS: (1) Nephrilin protects dermal fibroblasts from loss of viability and collagen synthesis after ultraviolet A (UV-A) or H2O2 insult. (2) Nephrilin reduces reactive oxygen species (ROS) formation by H2O2-treated (PTECs) with or without nicotine pretreatment. Using RNA arrays and pathway analysis we demonstrate that nicotine and H2O2-treated PTECs specifically induced Rac1 gene networks in these cells. (3) Using kidney tissue and urine from the burn trauma model we demonstrate significant elevations of [a] 8-aminoprostane in urine; [b] kidney tissue histone modification and DNA methylation; and [c] post-transcriptional phosphorylation events consistent with Rac1 activation in kidney tissue. CONCLUSION: Nephrilin protects against oxidative stress, possibly by modulating the activation of Rac1.

6.
Pediatr Res ; 82(5): 822-828, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28665927

ABSTRACT

BackgroundMany adolescents are exposed to nicotine via smoking, e-cigarette use, or second-hand smoke. Nicotine-induced renal oxidative stress and its long-term consequences may be higher in adolescents than in adults because of intrinsic factors in the adolescent kidney.MethodsAdolescent and adult male C57Bl/6J mice were subjected to 2 or 200 µg/ml nicotine, which closely emulates passive or active smoking, respectively, for 4 weeks. Extent of nicotine exposure (cotinine content), oxidative stress (HNE), renal function (creatinine), tubular injury (KIM-1), and pretreatment renal levels of select pro-oxidant (p66shc) and antioxidant (Nrf2/MnSOD) genes were determined. Impact of p66shc overexpression or Nrf2/MnSOD knockdown on low-/high-dose nicotine-induced oxidative stress was determined in cultured renal proximal tubule cells.ResultsDespite similar plasma/renal cotinine levels, renal HNE and KIM-1 contents were higher in adolescents compared with those in adults, whereas renal function was unaltered after passive or active smoking-equivalent nicotine exposure. Pretreatment levels of p66shc were higher, whereas Nrf2/MnSOD levels were lower in the adolescent kidney. In agreement with this, overexpression of p66shc or knockdown of Nrf2/MnSOD augmented nicotine-induced ROS production in renal proximal tubule cells.ConclusionChronic nicotine exposure incites higher oxidative stress in the adolescent than in adult kidney because of a pre-existent pro-oxidant milieu.


Subject(s)
Kidney Diseases/etiology , Kidney Tubules, Proximal/drug effects , Nicotine/toxicity , Nicotinic Agonists/pharmacology , Oxidative Stress/drug effects , Smoking/adverse effects , Tobacco Smoke Pollution/adverse effects , Age Factors , Aldehydes/metabolism , Animals , Cells, Cultured , Cotinine/metabolism , Cotinine/toxicity , Creatinine/metabolism , Hepatitis A Virus Cellular Receptor 1/metabolism , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Nicotine/metabolism , Nicotinic Agonists/metabolism , Risk Factors , Smoking/metabolism , Smoking/pathology , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Superoxide Dismutase/metabolism , Time Factors
7.
Anticancer Res ; 37(8): 4075-4079, 2017 08.
Article in English | MEDLINE | ID: mdl-28739690

ABSTRACT

BACKGROUND/AIM: We have shown that either chronic nicotine (NIC) exposure or 5-aza-cytidine (AZA) augments oxidative stress-dependent injury through stimulating p66shc in renal cells. Hence, NIC could exacerbate adverse effects of AZA while antioxidants such as resveratrol (RES) could prevent it. MATERIALS AND METHODS: Renal proximal tubule cells (NRK52E) were treated with 20 µM RES prior to 200 µM NIC plus 100 nM AZA and cell injury (LDH release) was determined. Reporter luciferase assays determined p66shc activation and RES-induced antioxidant responses. Genetic manipulations identified the mechanism of RES action. RESULTS: NIC exacerbated AZA-dependent injury via augmenting p66shc transcription. While RES suppressed NIC+AZA-mediated injury, -surprisingly-it further enhanced activity of the p66shc promoter. RES protected cells via the cytoplasmic p66shc/Nrf2/heme oxygenase-1 (HO-1) axis. CONCLUSION: RES can protect the kidney from adverse effects of NIC in patients undergoing anticancer therapy.


Subject(s)
Kidney Tubules, Proximal/drug effects , Neoplasms/complications , Src Homology 2 Domain-Containing, Transforming Protein 1/biosynthesis , Stilbenes/administration & dosage , Antioxidants/administration & dosage , Azacitidine/adverse effects , Cell Line , Heme Oxygenase-1/genetics , Humans , Kidney Tubules, Proximal/injuries , Kidney Tubules, Proximal/pathology , NF-E2-Related Factor 2/genetics , Neoplasms/drug therapy , Neoplasms/pathology , Nicotine/adverse effects , Oxidative Stress/drug effects , Promoter Regions, Genetic , Resveratrol , Signal Transduction/drug effects , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics
8.
In Vivo ; 31(4): 551-555, 2017.
Article in English | MEDLINE | ID: mdl-28652419

ABSTRACT

BACKGROUND/AIM: Nicotine (NIC) exposure - via smoking and the increasingly popular E-cigarettes- increases oxidative stress and hence, renal risk in smokers. Resveratrol (RES) may help ameliorate this risk by mounting anti-oxidant responses in the kidney. MATERIALS AND METHODS: Renal proximal tubule cells (NRK52E) were treated with vehicle or 20 µM RES prior to treatment with 200 µM NIC and generation of reactive oxygen species (ROS) as well as cell viability was determined. RES-induced antioxidant responses were determined in reporter luciferase assays. Gene silencing was used to determine mechanism of RES action. RESULTS: RES protected NRK52E cells from NIC-induced oxidative injury. RES activated the promoter of the anti-oxidant manganese superoxide dismutase (MnSOD) gene via activation of the forkhead box O (FoxO3a) transcription factor. Silencing of MnSOD abolished the protective effects of RES on NIC-associated oxidative injury. CONCLUSION: RES may provide protection to the kidney from the adverse effects of NIC in smokers.


Subject(s)
Acute Kidney Injury/drug therapy , Kidney Tubules, Proximal/drug effects , Stilbenes/administration & dosage , Superoxide Dismutase/genetics , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/physiopathology , Antioxidants/administration & dosage , Humans , Kidney Tubules, Proximal/injuries , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/physiopathology , Nicotine/toxicity , Oxidation-Reduction , Oxidative Stress/drug effects , Promoter Regions, Genetic , Reactive Oxygen Species , Resveratrol , Smoking/adverse effects
9.
PLoS One ; 12(6): e0179777, 2017.
Article in English | MEDLINE | ID: mdl-28640852

ABSTRACT

Plant flavonoids are well known as antioxidants against oxidative stress induced by exposure to external pollutants. Nicotine (NIC) is one of those agents which increases renal oxidative stress, an important factor in the pathogenesis of renal epithelial injury in smokers. Although several studies had been conducted on flavonoids and oxidative stress, the mechanism of the protective pathways are not fully understood. Here, we present studies on antioxidant properties of two mono-hydroxyflavone isomers, 3-hydroxyflanove (3HF)- and 7-hydroxyflavone (7HF), against nicotine-associated oxidative stress and injury in cultured renal proximal tubule cells and correlate their antioxidant properties with their chemical structure. Our data clearly demonstrates, for the first time, that while both 3HF and 7HF protect renal cells from NIC-associated cytotoxicity, the mechanism of their action is different: 3HF elicits protective activity via the PKA/CREB/MnSOD pathway while 7HF does so via the ERK/Nrf2/HO-1 pathway. Molecular docking and dynamics simulations with two major signaling pathway proteins showed significant differences in the binding energies of 3HF (-5.67 and -7.39 kcal.mol-1) compared to 7HF (-5.41 and -8.55 kcal.mol-1) in the matrices of CREB and Keap1-Nrf2 proteins respectively, which corroborate with the observed differences in their protective properties in the renal cells. The implications of this novel explorative study is likely to promote the understanding of the mechanisms of the antioxidative functions of different flavones.


Subject(s)
Flavonoids/pharmacology , Kidney Tubules, Proximal/cytology , Nicotine/adverse effects , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Cell Line , Cyclic AMP Response Element-Binding Protein/chemistry , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation/drug effects , Flavonoids/metabolism , Heme Oxygenase-1/metabolism , Kidney Tubules, Proximal/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Promoter Regions, Genetic/genetics , Protein Conformation , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
10.
J. physiol. biochem ; 73(2): 267-273, mayo 2017.
Article in English | IBECS | ID: ibc-168483

ABSTRACT

α-Tocopherol (TOC) is a widely used supplement known for its role as an antioxidant. Previously, we have shown that TOC elicits adaptive responses by upregulating the ERK/CREB/HO-1 pathway, which depends on its concentration in cultured renal proximal tubule cells (RPTCs). This suggests that high-dose TOC (hTOC) may elicit adverse effects via inflicting oxidative stress. Since the pro-oxidant p66shc is a major mediator of oxidant injury in various models of renal toxicants, we tested the hypothesis that hTOC elicits renal toxicity through activation of p66shc and consequent oxidative stress. RPTCs (NRK52E) were treated with high-dose TOC (hTOC; 400 nM) in cells where expression or mitochondrial cytochrome c-binding of p66shc was manipulated by genetic means. Intracellular production of reactive oxygen species (ROS), mitochondrial depolarization, and cell viability was also determined. Additionally, activation of the pro-survival ERK/CREB/HO-1 signaling and the p66shc promoter was determined via reporter luciferase assays. hTOC decreased cell viability via increasing ROS-dependent mitochondrial depolarization and suppressing the pro-survival ERK/CREB/HO-1 pathway via transcriptional activation of p66shc. Conversely, either knockdown of p66shc, mutation of its mitochondrial cytochrome c-binding site, or overexpression of ERK or HO-1 ameliorated adverse effects of hTOC and restored the pro-survival signaling. The pro-oxidant p66shc plays dual role in toxicity of high-dose TOC: it provokes oxidative stress and suppresses adaptive responses (AU)


No disponible


Subject(s)
Animals , Rats , Antioxidants/adverse effects , Kidney Tubules, Proximal/metabolism , Gene Expression Regulation , Oxidative Stress , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , alpha-Tocopherol/adverse effects , Amino Acid Substitution , Dietary Supplements/adverse effects , Membrane Potential, Mitochondrial , MAP Kinase Signaling System , Recombinant Fusion Proteins , Reactive Oxygen Species , Gene Knockdown Techniques , Cytochromes c , Binding Sites
11.
J Physiol Biochem ; 73(2): 267-273, 2017 May.
Article in English | MEDLINE | ID: mdl-28155123

ABSTRACT

α-Tocopherol (TOC) is a widely used supplement known for its role as an antioxidant. Previously, we have shown that TOC elicits adaptive responses by upregulating the ERK/CREB/HO-1 pathway, which depends on its concentration in cultured renal proximal tubule cells (RPTCs). This suggests that high-dose TOC (hTOC) may elicit adverse effects via inflicting oxidative stress. Since the pro-oxidant p66shc is a major mediator of oxidant injury in various models of renal toxicants, we tested the hypothesis that hTOC elicits renal toxicity through activation of p66shc and consequent oxidative stress. RPTCs (NRK52E) were treated with high-dose TOC (hTOC; 400 nM) in cells where expression or mitochondrial cytochrome c-binding of p66shc was manipulated by genetic means. Intracellular production of reactive oxygen species (ROS), mitochondrial depolarization, and cell viability was also determined. Additionally, activation of the pro-survival ERK/CREB/HO-1 signaling and the p66shc promoter was determined via reporter luciferase assays. hTOC decreased cell viability via increasing ROS-dependent mitochondrial depolarization and suppressing the pro-survival ERK/CREB/HO-1 pathway via transcriptional activation of p66shc. Conversely, either knockdown of p66shc, mutation of its mitochondrial cytochrome c-binding site, or overexpression of ERK or HO-1 ameliorated adverse effects of hTOC and restored the pro-survival signaling. The pro-oxidant p66shc plays dual role in toxicity of high-dose TOC: it provokes oxidative stress and suppresses adaptive responses.


Subject(s)
Antioxidants/adverse effects , Gene Expression Regulation , Kidney Tubules, Proximal/metabolism , Oxidative Stress , Promoter Regions, Genetic , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , alpha-Tocopherol/adverse effects , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Cell Survival , Cytochromes c/chemistry , Cytochromes c/metabolism , Dietary Supplements/adverse effects , Gene Knockdown Techniques , Genes, Reporter , Kidney Tubules, Proximal/cytology , MAP Kinase Signaling System , Membrane Potential, Mitochondrial , Mutation , Rats , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/antagonists & inhibitors , Src Homology 2 Domain-Containing, Transforming Protein 1/chemistry , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics
12.
Apoptosis ; 22(2): 220-228, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27770269

ABSTRACT

Chronic nicotine exposure (via smoking, E-cigarettes) increases oxidative stress in the kidney that sensitizes it to additional injury in experimental models and in the renal patient. The pro-apoptotic p66shc protein-via serine36 phosphorylation that facilitates its mitochondrial translocation and therein cytochrome c binding-generates oxidative stress that leads to injury of renal proximal tubule cells during chronic nicotine exposure. Coenzyme Q10-a clinically safe antioxidant-has been used against nicotine/smoke extract-associated oxidative stress in various non-renal cells. This study explored the anti-oxidant/anti-apoptotic effect of Coenzyme Q10 on nicotine-induced oxidative stress and its impact on p66shc in cultured rat renal proximal tubule cells (NRK52E). We studied the anti-oxidant effect of 10 µM Coenzyme Q10 using various mutants of the p66shc gene and also determined the induction of selected anti-oxidant entities (antioxidant response element, promoter of the manganese superoxide dismutase gene) in reporter luciferase assay during oxidative stress induced by 200 µM nicotine. Our studies revealed that Coenzyme Q10 strongly inhibits nicotine-mediated production of reactive oxygen species and consequent apoptosis that requires serine36 phosphorylation but not mitochondrial translocation/cytochrome c binding of p66shc. While both nicotine and Coenzyme Q10 stimulates the p66shc promoter, only nicotine exposure results in mitochondrial translocation of p66shc. In contrast, the Coenzyme Q10-stimulated and non-mitochondrial p66shc activates the anti-oxidant manganese superoxide dismutase promoter via the antioxidant response elements and hence, rescues cells from nicotine-induced oxidative stress and consequent apoptosis.


Subject(s)
Apoptosis/genetics , Kidney Tubules, Proximal/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Ubiquinone/analogs & derivatives , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Cytochromes c/metabolism , Electronic Nicotine Delivery Systems , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Nicotine/toxicity , Oxidative Stress/drug effects , Phosphorylation , Rats , Reactive Oxygen Species , Smoking/adverse effects , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Ubiquinone/genetics
13.
Mol Med Rep ; 14(3): 2771-7, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27486058

ABSTRACT

Nicotine (NIC) exposure augments free fatty acid (FFA) deposition and oxidative stress, with a concomitant increase in the expression of the pro-oxidant p66shc. In addition, a decrease in the antioxidant manganese superoxide dismutase (MnSOD) has been observed in the kidneys of mice fed a high­fat diet. The present study aimed to determine whether the pro­oxidant p66shc mediates NIC­dependent increases in renal oxidative stress by augmenting the production of reactive oxygen species (ROS) and suppressing the FFA­induced antioxidant response in cultured NRK52E renal proximal tubule cells. Briefly, NRK52E renal proximal tubule cells were treated with 200 µM NIC, 100 µM oleic acid (OA), or a combination of NIC and OA. The expression levels of p66shc and MnSOD were modulated according to genetic methods. ROS production and cell injury, in the form of lactate dehydrogenase release, were subsequently detected. Promoter activity of p66shc and MnSOD, as well as forkhead box (FOXO)­dependent transcription, was investigated using reporter luciferase assays. The results demonstrated that NIC exacerbated OA­mediated intracellular ROS production and cell injury through the transcriptional activation of p66shc. NIC also suppressed OA­mediated induction of the antioxidant MnSOD promoter activity through p66shc­dependent inactivation of FOXO activity. Overexpression of p66shc and knockdown of MnSOD had the same effect as treatment with NIC on OA­mediated lipotoxicity. These data may be used to generate a therapeutic means to ameliorate renal lipotoxicity in obese smokers.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Nicotine/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Animals , Fatty Acids, Nonesterified/metabolism , Gene Expression , Mice , Promoter Regions, Genetic , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Transcriptional Activation
14.
In Vivo ; 30(4): 407-11, 2016.
Article in English | MEDLINE | ID: mdl-27381601

ABSTRACT

BACKGROUND/AIM: Studies have shown that simvastatin (SIM) inhibits epithelial-mesenchymal transition (EMT), a key step in fibrosis, and activates the anti-fibrotic heme oxygenase-1 (HO-1) gene in renal proximal tubule cells independent of its lipid-lowering. We tested the hypothesis that SIM inhibits EMT via HO-1-dependent suppression of reactive oxygen species (ROS) release. MATERIALS AND METHODS: Renal proximal tubule cells were treated with either 10 µM SIM or 10 ng/ml transforming growth factor-ß1 (TGFß1) or with their combination and promoter activity of the alpha-smooth muscle actin (α-SMA) gene, stress fiber formation (markers of EMT), as well as ROS production were determined. HO-1 was manipulated via genetic and pharmacologic means. RESULTS: SIM prevented TGFß1-dependent EMT and ROS production. Inhibition/knockdown of HO-1 reversed, while induction/overexpression of HO-1 emulated beneficial effects of SIM. CONCLUSION: SIM, via HO-1, suppresses TGFß1-dependent ROS production and, hence, EMT. Further evaluation of the anti-fibrotic nature of SIM in the kidney would be useful in the treatment of chronic kidney disease.


Subject(s)
Anticholesteremic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/metabolism , Kidney Tubules, Proximal/drug effects , Simvastatin/pharmacology , Animals , Cells, Cultured , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/enzymology , Reactive Oxygen Species/metabolism , Swine , Transforming Growth Factor beta1/pharmacology
15.
In Vivo ; 30(3): 219-24, 2016.
Article in English | MEDLINE | ID: mdl-27107078

ABSTRACT

BACKGROUND/AIM: Both maternal nicotine (NIC) exposure and placental insufficiency increase oxidative stress in the fetal kidney ensuing fetal programming of renal diseases in adult life. Their combined effects, however, are unknown. We tested the hypothesis that maternal NIC exposure exacerbates renal oxidative stress and injury in fetuses of pregnant rats with placental insufficiency. MATERIALS AND METHODS: Fourteen-day-pregnant rats were subjected to sham operation or reduced uterine perfusion pressure (RUPP) that received either nicotine (20 µg/ml in 1% saccharine) or vehicle (1% saccharine) in their drinking water. At gestational age of 21 days, male fetuses were collected by C-section and sacrificed: plasma and renal cotinine content, extent of renal oxidative stress (4-hydroxynonenal [HNE] and HO-1) and injury (KIM-1) were determined together with the weight of the fetal kidney and fetus. RESULTS: Prenatal NIC exposure resulted in cotinine accumulation in the plasma and kidney of the fetuses, augmented RUPP-associated increase in renal HNE content and HO-1 expression as well as KIM-1 expression. NIC also enhanced RUPP-induced reduction in fetal and fetal kidney weight. CONCLUSION: Prenatal NIC exposure augments the existing renal risk in the growth-restricted fetus, which may contribute to worsening in fetal programming of renal disease.


Subject(s)
Kidney/drug effects , Nicotine/toxicity , Nicotinic Agonists/toxicity , Oxidative Stress/drug effects , Uterus/drug effects , Animals , Blood Pressure/drug effects , Cotinine/analysis , Cotinine/blood , Female , Fetal Growth Retardation/blood , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/physiopathology , Kidney/embryology , Kidney/metabolism , Male , Maternal Exposure , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Perfusion , Placental Insufficiency/blood , Placental Insufficiency/chemically induced , Placental Insufficiency/physiopathology , Pregnancy , Rats, Sprague-Dawley , Uterus/blood supply , Uterus/physiopathology
16.
Nicotine Tob Res ; 18(7): 1628-34, 2016 07.
Article in English | MEDLINE | ID: mdl-26896163

ABSTRACT

INTRODUCTION: Life expectancy of an obese smoker is 13 years less than a normal weight smoker, which could be linked to the increased renal risk imposed by smoking. Both smoking-through nicotine (NIC)-and obesity-by free fatty acid overload-provoke oxidative stress in the kidney, which ultimately results in development of chronic kidney injury. Their combined renal risk, however, is virtually unknown. We tested the hypothesis that chronic NIC exposure worsens renal oxidative stress in mice on high-fat diet (HFD) by altering the balance between expression of pro-oxidant and antioxidant genes. METHODS: Nine-week-old male C57Bl/6J mice consumed normal diet (ND) or HFD and received either NIC (200 µg/ml) or vehicle (2% saccharine) in their drinking water. Body weight, plasma clinical parameters, renal lipid deposition, markers of renal oxidative stress and injury, as well as renal expression of the pro-oxidant p66shc and the antioxidant MnSOD were determined after 12 weeks. RESULTS: NIC significantly augmented levels of circulating free fatty acid, as well as lipid deposition, oxidative stress and sublethal injury in the kidneys of mice on HFD. In addition, NIC exposure suppressed HFD-mediated induction of MnSOD while increased expression of p66shc in the kidney. CONCLUSIONS: Tobacco smoking or the increasingly popular E-cigarettes-via NIC exposure-could worsen obesity-associated lipotoxicity in the kidney. Hence, our findings could help to develop strategies that mitigate adverse effects of NIC on the obese kidney. IMPLICATIONS: Life expectancy of an obese smoker is 13 years less than a normal weight smoker, which could be linked to the increased renal risk imposed by smoking. NIC-the main component of tobacco smoke, E-cigarettes and replacement therapies-links smoking to renal injury via oxidative stress, which could superimpose renal oxidative stress caused by obesity. Our results substantiate this scenario using a mouse model of diet induced obesity and NIC exposure and imply the augmented long-term renal risk in obese smokers. Also, our study may help to develop strategies that mitigate adverse effects of NIC on the obese kidney.


Subject(s)
Diet, High-Fat , Kidney/metabolism , Nicotine/pharmacology , Obesity , Oxidative Stress/drug effects , Smoking/adverse effects , Animals , Biomarkers/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
17.
Pediatr Res ; 79(2): 243-50, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26492285

ABSTRACT

BACKGROUND: Statins elicit antioxidant effects independently of their lipid-lowering properties. Heme oxygenase-1 (HO-1) induction may be a part of these pleiotropic effects, which are insufficiently described in the kidney. We hypothesize that simvastatin (SIM) transcriptionally activates HO-1 that protects renal proximal tubule cells from lipotoxic injury. METHODS: Impact of SIM on 100 µmol/l oleic acid (OA)-mediated reactive oxygen species (ROS) production and consequent oxidative stress (4-hydroxynonenal (HNE) content) as well as cell injury/apoptosis (lactate dehydrogenase (LDH) release, caspase-3 activation) were determined in cultured renal proximal tubule (NRK52E) cells. Effect of SIM on the HO-1 promoter and its enhancer elements (antioxidant response element (ARE), CCAAT, AP1, and cAMP response element (CRE)) was also determined in reporter luciferase assays. Dominant-negative (dnMEK, M1CREB) and pharmacologic (H89) approaches were used to inhibit activation of extracellular signal regulated kinase (ERK), CREB, and protein kinase A (PKA), respectively. RESULTS: SIM dose-dependently activated the HO-1 promoter that was essential for protection against OA-dependent ROS production/oxidative stress and LDH release/caspase-3 activation. We found that the HO-1 promoter was induced through ERK and PKA-dependent activation of the CRE by SIM. CONCLUSION: SIM may protect the kidney from adverse effects of circulating fatty acids by upregulating the antioxidant HO-1, aside from its well-described lipid-lowering effects.


Subject(s)
Antioxidants/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Heme Oxygenase (Decyclizing)/biosynthesis , Kidney Tubules, Proximal/drug effects , Oleic Acid/toxicity , Oxidative Stress/drug effects , Simvastatin/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoprotection , Dose-Response Relationship, Drug , Enzyme Induction , Extracellular Signal-Regulated MAP Kinases/metabolism , Heme Oxygenase (Decyclizing)/genetics , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/pathology , Promoter Regions, Genetic , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transfection
18.
J. physiol. biochem ; 71(1): 1-7, mar. 2015.
Article in English | IBECS | ID: ibc-133897

ABSTRACT

Smoking and obesity increases renal oxidative stress via nicotine (NIC) or free fatty acid such as oleic acid (OA) but decreases levels of the vitamin E-derivative alpha-tocopherol (TOC), which has shown to stimulate the antioxidant system such as heme oxygenase-1 (HO-1). Hence, we hypothesized that supplementation of TOC may protect renal proximal tubules from NIC- or OA-mediated oxidative stress by upregulating the HO-1 gene. NIC- or OA-dependent production of reactive oxygen species (ROS) was determined in the presence or absence of various pharmacologic or genetic inhibitors that modulate HO-1 activation and enhancer elements in the HO-1 promoter such as the antioxidant response element (ARE) and the cAMP-response element (CRE) in renal proximal tubule cells (NRK52E). Activity of the HO-1 promoter, the ARE and the CRE was determined in luciferase assays. We found that pre- or posttreatment with TOC attenuated NIC- or OA-dependent ROS production that required HO-1 activation. TOC activated the HO-1 promoter via the CRE but not the ARE enhancer through the extracellular signal-regulated kinase (ERK) and protein kinase A (PKA). Consequently, inhibitors of ERK, PKA, or CRE activation mitigated beneficial effects of TOC on NIC- or OA-mediated ROS production. Hence, vitamin E supplementation-via induction of the cytoprotective HO-1-may help to reduce renal oxidative stress imposed by smoking or obesity


Subject(s)
Humans , alpha-Tocopherol/pharmacokinetics , Obesity/physiopathology , Smoking/physiopathology , Kidney Tubules, Proximal , Protective Agents/pharmacokinetics , Reactive Oxygen Species/analysis , Vitamin E/pharmacokinetics , Oxidative Stress
19.
J Physiol Biochem ; 71(1): 1-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25471815

ABSTRACT

Smoking and obesity increases renal oxidative stress via nicotine (NIC) or free fatty acid such as oleic acid (OA) but decreases levels of the vitamin E-derivative α-tocopherol (TOC), which has shown to stimulate the antioxidant system such as heme oxygenase-1 (HO-1). Hence, we hypothesized that supplementation of TOC may protect renal proximal tubules from NIC- or OA-mediated oxidative stress by upregulating the HO-1 gene. NIC- or OA-dependent production of reactive oxygen species (ROS) was determined in the presence or absence of various pharmacologic or genetic inhibitors that modulate HO-1 activation and enhancer elements in the HO-1 promoter such as the antioxidant response element (ARE) and the cAMP-response element (CRE) in renal proximal tubule cells (NRK52E). Activity of the HO-1 promoter, the ARE and the CRE was determined in luciferase assays. We found that pre- or posttreatment with TOC attenuated NIC- or OA-dependent ROS production that required HO-1 activation. TOC activated the HO-1 promoter via the CRE but not the ARE enhancer through the extracellular signal-regulated kinase (ERK) and protein kinase A (PKA). Consequently, inhibitors of ERK, PKA, or CRE activation mitigated beneficial effects of TOC on NIC- or OA-mediated ROS production. Hence, vitamin E supplementation-via induction of the cytoprotective HO-1-may help to reduce renal oxidative stress imposed by smoking or obesity.


Subject(s)
Heme Oxygenase-1/biosynthesis , Kidney Tubules, Proximal/drug effects , Nicotine/pharmacology , Oleic Acid/pharmacology , Oxidative Stress/drug effects , alpha-Tocopherol/pharmacology , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Induction , Heme Oxygenase-1/genetics , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/metabolism , Promoter Regions, Genetic , Protein Kinases/metabolism , Rats , Reactive Oxygen Species/metabolism
20.
In Vivo ; 28(6): 1097-100, 2014.
Article in English | MEDLINE | ID: mdl-25398805

ABSTRACT

BACKGROUND/AIM: Gender-associated dimorphism in renal oxidative stress may be related to the protective effects of estrogens or the adverse effects of testosterone. Signal transducer and activator of transcription-3 (STAT3)-dependent transcription is vital in renal antioxidant responses, which may be differentially regulated by sex hormones. MATERIALS AND METHODS: Renal proximal tubule cells were treated with 400 µM H2O2 in the presence or absence of 100 nM dihydrotestosterone (DHT), 100 nM 17ß-estradiol (E2) or dominant-negative STAT3 (dnSTAT3). Production of reactive oxygen species (ROS), phosphorylation/transcriptional activation of STAT3 and promoter activity of the STAT3-regulated antioxidant gene (MnSOD) were determined. RESULTS: After treatment with H2O2, DHT decreased tyrosine phosphorylation/transcriptional activity of STAT3 and promoter activity of MnSOD while E2 increased them. Consequently, DHT augmented while E2 attenuated ROS production. Effects of dnSTAT3 were similar to DHT. CONCLUSION: Sex hormones may influence renal oxidative stress through differential regulation of STAT3-dependent antioxidant responses.


Subject(s)
Antioxidants/metabolism , Gonadal Steroid Hormones/metabolism , Kidney Tubules, Proximal/metabolism , Oxidative Stress , STAT3 Transcription Factor/metabolism , Animals , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Gonadal Steroid Hormones/pharmacology , Kidney Tubules, Proximal/drug effects , Oxidation-Reduction/drug effects , Phosphorylation/drug effects , Promoter Regions, Genetic , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...