Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Int Arch Allergy Immunol ; : 1-12, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38797160

ABSTRACT

INTRODUCTION: Kamebakaurin is an active constituent of both Rabdosia japonica and Rabdosia excisa, which are utilized in Chinese traditional medicine for improving symptoms in patients with allergies. We investigated the molecular mechanisms of the anti-allergic effects of kamebakaurin using BMMCs. METHODS: The degranulation ratio, histamine release, and the interleukin (IL)-4, leukotriene B4 (LTB4), and cysteinyl leukotriene productions on antigen-triggered BMMC were investigated. Additionally, the effects of kamebakaurin on signal transduction proteins were examined by Western blot and binding to the Syk and Lyn kinase domain was calculated. The effects of kamebakaurin on antigen-induced hyperpermeability were investigated using mouse model. RESULTS: At 10 µm, kamebakaurin partially inhibited degranulation, histamine release, and IL-4 production. At 30 µm, kamebakaurin partially reduced LTB4 and cysteinyl leukotriene productions and suppressed degranulation, histamine release, and IL-4 production. Phosphorylation of both Syk Y519/520 and its downstream protein, Gab2, was reduced by kamebakaurin, and complete inhibition was observed with 30 µm kamebakaurin. In contrast, phosphorylation of Erk was only partially inhibited, even in the presence of 30 µm kamebakaurin. Syk Y519/520 is known to be auto-phosphorylated via intramolecular ATP present in its own ATP-binding site, and this auto-phosphorylation triggers degranulation, histamine release, and IL-4 production. Docking simulation study indicated kamebakaurin blocked ATP binding to the ATP-binding site in Syk. Therefore, inhibition of Syk auto-phosphorylation by kamebakaurin binding to the Syk ATP-binding site appeared to cause a reduction of histamine release and IL-4 production. Kamebakaurin inhibited antigen-induced vascular hyperpermeability in a dose-dependent fashion but did not reduce histamine-induced vascular hyperpermeability. CONCLUSION: Kamebakaurin ameliorates allergic symptoms via inhibition of Syk phosphorylation; thus, kamebakaurin could be a lead compound for the new anti-allergic drug.

2.
J Appl Toxicol ; 44(6): 846-852, 2024 06.
Article in English | MEDLINE | ID: mdl-38291012

ABSTRACT

Trovafloxacin is a quinolone antibiotic drug with broad-spectrum activity, which was withdrawn from a global market relatively soon after approval because of serious liver injury. The characteristics of trovafloxacin-induced liver injury are consistent with an idiosyncratic reaction; however, the details of the mechanism have not been elucidated. We examined whether trovafloxacin induces the release of damage-associated molecular patterns (DAMPs) that activate inflammasomes. We also tested ciprofloxacin, levofloxacin, gatifloxacin, and grepafloxacin for their ability to activate inflammasomes. Drug bioactivation was performed with human hepatocarcinoma functional liver cell-4 (FLC-4) cells, and THP-1 cells (human monocyte cell line) were used for the detection of inflammasome activation. The supernatant from the incubation of trovafloxacin with FLC-4 cells for 7 days increased caspase-1 activity and production of IL-1ß by THP-1 cells. In the supernatant of FLC-4 cells that had been incubated with trovafloxacin, heat shock protein (HSP) 40 was significantly increased. Addition of a cytochrome P450 inhibitor to the FLC-4 cells prevented the release of HSP40 from the FLC-4 cells and inflammasome activation in THP-1 cells by the FLC-4 supernatant. These results suggest that reactive metabolites of trovafloxacin can cause the release of DAMPs from hepatocytes that can activate inflammasomes. Inflammasome activation may be an important step in the activation of the immune system by trovafloxacin, which, in some patients, can cause immune-related liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury , Fluoroquinolones , Inflammasomes , Naphthyridines , Humans , Inflammasomes/metabolism , Inflammasomes/drug effects , Fluoroquinolones/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Naphthyridines/toxicity , Naphthyridines/pharmacology , THP-1 Cells , Anti-Bacterial Agents/toxicity , Cell Line, Tumor , Interleukin-1beta/metabolism
3.
Am J Physiol Gastrointest Liver Physiol ; 304(9): G773-80, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23449671

ABSTRACT

In antral mucous cells, acetylcholine (ACh, 1 µM) activates Ca(2+)-regulated exocytosis, consisting of an initial peak that declines rapidly (initial transient phase) followed by a second slower decline (late phase) lasting during ACh stimulation. The addition of 8-bromo-cGMP (8-BrcGMP) enhanced the initial phase, which was inhibited by the protein kinase G (PKG) inhibitor guanosine 3',5'-cyclic monophosphorothoiate, ß-phenyl-1,N(2)-etheno-8-bromo, Rp-isomer, sodium salt (Rp-8-BrPETcGMPS, 100 nM). However, Rp-8-BrPETcGMPS produced a delayed, but transient, increase in the exocytotic frequency during the late phase that was abolished by a protein kinase A (PKA) inhibitor (PKI-amide), suggesting that Rp-8-BrPETcGMPS accumulates cAMP. The cGMP-dependent phosphodiesterase 2 (PDE2), which degrades cAMP, may exist in antral mucous cells. The PDE2 inhibitor BAY-60-7550 (250 nM) mimicked the effect of Rp-8-BrPETcGMPS on ACh-stimulated exocytosis. Measurement of the cGMP and cAMP contents in antral mucosae revealed that ACh stimulates the accumulation of cGMP and that BAY-60-7550 accumulates cAMP similarly to Rp-8-BrPETcGMPS during ACh stimulation. Analyses of Western blot and immunohistochemistry demonstrated that PDE2A exists in antral mucous cells. In conclusion, Rp-8-BrPETcGMPS accumulates cAMP by inhibiting PDE2 in ACh-stimulated antral mucous cells, leading to the delayed, but transient, increase in the frequency of Ca(2+)-regulated exocytosis. PDE2 may prevent antral mucous cells from excessive mucin secretion caused by the cAMP accumulation.


Subject(s)
Calcium/physiology , Cyclic AMP/metabolism , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP/analogs & derivatives , Exocytosis/drug effects , Pyloric Antrum/physiology , Acetylcholine/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Dinoprostone/metabolism , Gastric Mucosa/drug effects , Guinea Pigs , Male , Protein Kinase Inhibitors/pharmacology , Pyloric Antrum/drug effects
4.
Heart Vessels ; 28(5): 646-57, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23277455

ABSTRACT

We aimed to investigate whether atrial natriuretic peptide (ANP) attenuates angiotensin II (Ang II)-induced myocardial remodeling and to clarify the possible molecular mechanisms involved. Thirty-five 8-week-old male Wistar-Kyoto rats were divided into control, Ang II, Ang II + ANP, and ANP groups. The Ang II and Ang II + ANP rats received 1 µg/kg/min Ang II for 14 days. The Ang II + ANP and ANP rats also received 0.1 µg/kg/min ANP intravenously. The Ang II and Ang II + ANP rats showed comparable blood pressure. Left ventricular fractional shortening and ejection fraction were lower in the Ang II rats than in controls; these indices were higher (P < 0.001) in the Ang II + ANP rats than in the Ang II rats. In the Ang II rats, the peak velocity of mitral early inflow and its ratio to atrial contraction-related peak flow velocity were lower, and the deceleration time of mitral early inflow was significantly prolonged; these changes were decreased by ANP. Percent fibrosis was higher (P < 0.001) and average myocyte diameters greater (P < 0.01) in the Ang II rats than in controls. ANP decreased both myocardial fibrosis (P < 0.01) and myocyte hypertrophy (P < 0.01). Macrophage infiltration, expression of mRNA levels of collagen types I and III, monocyte chemotactic protein-1, and a profibrotic/proinflammatory molecule, tenascin-C (TN-C) were increased in the Ang II rats; ANP significantly decreased these changes. In vitro, Ang II increased expression of TN-C and endothelin-1 (ET-1) in cardiac fibroblasts, which were reduced by ANP. ET-1 upregulated TN-C expression via endothelin type A receptor. These results suggest that ANP may protect the heart from Ang II-induced remodeling by attenuating inflammation, at least partly through endothelin 1/endothelin receptor A cascade.


Subject(s)
Angiotensin II , Anti-Inflammatory Agents/pharmacology , Atrial Natriuretic Factor/pharmacology , Endothelin-1/metabolism , Heart Diseases/prevention & control , Inflammation/prevention & control , Myocardium/metabolism , Receptor, Endothelin A/metabolism , Signal Transduction/drug effects , Ventricular Remodeling/drug effects , Animals , Anti-Inflammatory Agents/administration & dosage , Atrial Natriuretic Factor/administration & dosage , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Cells, Cultured , Disease Models, Animal , Fibrillar Collagens/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Inflammation Mediators/metabolism , Infusions, Intravenous , Macrophages/drug effects , Macrophages/metabolism , Male , Mitral Valve/drug effects , Mitral Valve/physiopathology , Myocardial Contraction/drug effects , Myocardium/pathology , Rats , Rats, Inbred WKY , Stroke Volume/drug effects , Time Factors , Ventricular Function, Left/drug effects
5.
Toxicol In Vitro ; 90: 105606, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37146920

ABSTRACT

Flutamide is a non-steroidal anti-androgen agent, which is mainly used for the treatment of prostate cancer. Flutamide is known to cause severe adverse events, which includes idiosyncratic liver injury. However, details of the mechanism of these adverse reactions have not been elucidated. We investigated whether flutamide induces the release of damage-associated molecular patterns (DAMPs) that activate inflammasomes. We also tested bicalutamide, enzalutamide, apalutamide, and darolutamide for their ability to activate inflammasomes in differentiated THP-1 cells. The supernatant from the incubation of flutamide and bicalutamide with human hepatocarcinoma functional liver cell-4 (FLC-4) cells increased caspase-1 activity and production of IL-1ß by differentiated THP-1 cells. In the supernatant of FLC-4 cells with flutamide and bicalutamide, the heat shock protein (HSP) 40 or 60 was significantly increased. Addition of a carboxylesterase or a CYP inhibitor to the FLC-4 cells prevented release of HSPs from the FLC-4 cells. These results suggested that the reactive metabolites of flutamide and bicalutamide can cause the release of DAMPs from hepatocytes and activate inflammasomes. Inflammasome activation may be an important step in the activation of the immune system by flutamide or bicalutamide, which in some patients, can cause immune-related adverse events.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Prostatic Neoplasms , Male , Humans , Flutamide/toxicity , Inflammasomes/metabolism , Androgen Antagonists/toxicity , Anilides/pharmacology , Nitriles/toxicity
6.
Cell Physiol Biochem ; 29(3-4): 511-22, 2012.
Article in English | MEDLINE | ID: mdl-22508058

ABSTRACT

The beating cilia play a key role in lung mucociliary transport. The ciliary beating frequency (CBF) and ciliary bend amplitude (CBA) of isolated mouse bronchiolar ciliary cells were measured using a light microscope equipped with a high-speed camera (500 Hz). Procaterol (aß(2)-agonist) increased CBA and CBF in a dose dependent manner via cAMP. The time course of CBA increase is distinct from that of CBF increase: procaterol at 10 nM first increased CBA and then CBF. Moreover, 10 pM procaterol increased CBA, not CBF, whereas 10 nM procaterol increased both CBA and CBF. Concentration-response studies of procaterol demonstrated that the CBA curve was shifted to a lower concentration than the CBF curve, which suggests that CBA regulation is different from CBF regulation. Measurements of microbead movements on the bronchiole of lung slices revealed that 10 pM procaterol increased the rate of ciliary transport by 37% and 10 nM procaterol increased it by 70%. In conclusion, we have shown that increased CBA is of particular importance for increasing the bronchiolar ciliary transport rate, although CBF also plays a role in increasing it.


Subject(s)
Bronchioles/drug effects , Cilia/drug effects , Mucociliary Clearance , Procaterol/pharmacology , Adrenergic beta-2 Receptor Agonists/pharmacology , Albuterol/pharmacology , Animals , Axoneme/metabolism , Axoneme/physiology , Bronchioles/metabolism , Bronchioles/physiology , Calcium/pharmacology , Cilia/metabolism , Cilia/physiology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Epithelium/drug effects , Epithelium/metabolism , Epithelium/physiology , Female , In Vitro Techniques , Mice , Mice, Inbred C57BL , Microspheres , Time Factors , Video Recording
7.
J Pharmacol Sci ; 113(4): 343-52, 2010.
Article in English | MEDLINE | ID: mdl-20644337

ABSTRACT

The activity of pyruvate dehydrogenase (PDH) is reduced in diabetic patients. Phosphorylation of the PDH E1alpha subunit by PDH kinase contributes to the suppression of PDH activity. PDH requires thiamine as a coenzyme. We investigated the exact mechanism of diabetes-induced PDH inhibition, and the effect of thiamine in both in vivo and in vitro experiments. Treatment of rats with thiamine significantly, although partially, recovered streptozotocin (STZ)-induced reductions in mitochondrial PDH activity. Nevertheless, we found that PDH E1alpha phosphorylation in the thiamine-treated STZ group was perfectly diminished to the same level as that in the control group. STZ treatment significantly caused enhancements of the expression of O-glycosylated protein in the rat hearts, which was decreased by thiamine repletion. Next, the rat cardiac fibroblasts (RCFs) were cultured in the presence of high glucose levels. Thiamine dramatically recovered high glucose-induced PDH inhibition. High glucose loads did not alter the phosphorylated PDH E1alpha. PDH inhibition in RCFs was not accompanied by an increase in the PDH E1alpha phosphorylation. The O-glycosylated protein was markedly increased in RCFs exposed to high glucose, which was inhibited by thiamine. These results suggest that thiamine ameliorates diabetes-induced PDH inhibition by suppressing the increased expression of the O-glycosylated protein. The O-glycosylation of PDH E1alpha may be involved in the regulation of the PDH activity.


Subject(s)
Glucose/administration & dosage , Heart/drug effects , Mitochondria, Heart/drug effects , Pyruvate Dehydrogenase Complex/antagonists & inhibitors , Thiamine/pharmacology , Animals , Base Sequence , Blotting, Western , Cells, Cultured , DNA Primers , Electrophoresis, Polyacrylamide Gel , Fibroblasts/drug effects , Male , Mitochondria, Heart/enzymology , Phosphorylation , Polymerase Chain Reaction , Pyruvate Dehydrogenase Complex/genetics , Pyruvate Dehydrogenase Complex/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar , Streptozocin
8.
J Pharmacol Sci ; 111(4): 446-50, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19966509

ABSTRACT

Diabetic cardiomyopathy may be accompanied by myocardial fibrosis. We have previously reported that cardiac fibrosis and protein O-glycosylation are elevated in diabetes. In this study, we examined if the hexosamine biosynthesis pathway (HBP) was involved with collagen expression in rat cardiac fibroblasts (RCFs). Long-term glucose load significantly increased type III collagen expression in RCFs, but did not affect the protein O-glycosylation. In addition, glucosamine treatment not only induced expressions of collagen types I and III, but also increased the O-glycosylated protein. These results suggest that O-glycosylation of protein induced by HBP activation modifies collagen expression and contributes to diabetic cardiomyopathy.


Subject(s)
Cardiomyopathies/metabolism , Collagen Type III/metabolism , Collagen Type I/metabolism , Diabetes Mellitus, Experimental/metabolism , Fibroblasts/metabolism , Glycosylation/drug effects , Hexosamines/biosynthesis , Animals , Cardiomyopathies/complications , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Fibroblasts/drug effects , Glucosamine/pharmacology , Glucose/pharmacology , Male , Rats , Rats, Wistar
9.
J Pharmacol Sci ; 111(4): 433-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19966510

ABSTRACT

Cisplatin causes chronic interstitial disease with fibrosis, but the development mechanism of interstitial fibrosis is not yet understood. We examined the effect of an antioxidant, N,N'-diphenyl-1,4-phenylenediamine (DPPD), on development of interstitial fibrosis induced by cisplatin. Cisplatin increased blood urea nitrogen (BUN), plasma creatinine, and elicited glucosuria and enzymuria at 3 days after administration, but these changes were restored to the normal level after 14 days. Type III collagen increased from 7 days after administration of cisplatin and the expansion of the interstitial fibrosis area became evident at 14 days. Sustained renal fibrosis worsened renal function again at 56 days. Administration of DPPD, which was started at 3 days after cisplatin treatment, significantly inhibited the increase in renal type III collagen contents and the expansion of the interstitial fibrosis area without affecting enzymuria and increased BUN. These results indicate that anti-fibrotic action of DPPD is not secondary due to the inhibition of acute renal injury but is rather a direct effect on renal fibrogenesis. DPPD did not prevent the infiltration of macrophages by cisplatin, suggesting that anti-fibrotic action of DPPD was not mediated by the inhibition of inflammatory cellular influx. It is suggested that reactive oxygen species are involved in cisplatin-induced renal interstitial fibrosis.


Subject(s)
Antioxidants/pharmacology , Cisplatin/antagonists & inhibitors , Fibrosis/prevention & control , Kidney/pathology , Phenylenediamines/pharmacology , Actins/metabolism , Aldehydes/metabolism , Animals , Cisplatin/toxicity , Collagen Type III/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney Function Tests , Male , Rats , Rats, Sprague-Dawley
10.
J Toxicol Sci ; 31(3): 207-17, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16960431

ABSTRACT

Zinc is an essential nutrient that can also be toxic. We have previously reported that zinc-related renal toxicity is due, in part, to free radical generation in the renal epithelial cell line, LLC-PK(1) cells. We have also shown that an MEK1/2 inhibitor, U0126, markedly inhibits zinc-induced renal cell injury. In this study, we investigated the role of an upstream MEK/ERK pathway, Raf-1 kinase pathway, and the transcription factor and ERK substrate Elk-1, in rat renal cortical slices exposed to zinc. Immediately after preparing slices from rat renal cortex, the slices were incubated in medium containing Raf-1 and MEK inhibitors. ERK1/2 and Elk-1 activation were determined by Western blot analysis for phosphorylated ERK (pERK) 1/2 and phosphorylated Elk-1 (pElk-1) in nuclear fractions prepared from slices exposed to zinc. Zinc caused not only increases in 4-hydroxynonenal (4-HNE) modified protein and lipid peroxidation, as an index of oxidant stress, and decreases in PAH accumulation, as that of renal cell injury in the slices. Zinc also induced a rapid increase in ERK/Elk-1 activity accompanied by increased expressions of pERK and pElk-1 in the nuclear fraction. A Raf-1 kinase inhibitor and an MEK1/2 inhibitor U0126 significantly attenuated zinc-induced decreases PAH accumulation in the slices. The Raf-1 kinase inhibitor and U0126 also suppressed ERK1/2 activation in nuclear fractions prepared from slices treated with zinc. The present results suggest that a Raf-1/MEK/ERK1/2 pathway and the ERK substrate Elk-1 are involved in free radical-induced injury in rat renal cortical slices exposed to zinc.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , Kidney Cortex/drug effects , MAP Kinase Kinase 1/physiology , MAP Kinase Kinase 2/physiology , Proto-Oncogene Proteins c-raf/physiology , Signal Transduction/physiology , Zinc/toxicity , Aldehydes/metabolism , Animals , Antioxidants/pharmacology , Kidney Cortex/metabolism , Lipid Peroxidation/drug effects , Male , Phenylenediamines/pharmacology , Phosphorylation , Rats , Rats, Sprague-Dawley
11.
Biochem Pharmacol ; 70(9): 1408-16, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16154539

ABSTRACT

Serum thymic factor (FTS), a thymic peptide hormone, has been reported to attenuate the bleomycin-induced pulmonary injury and also experimental pancreatitis and diabetes. In the present study, we investigated the effect of FTS on cis-diamminedichloroplatinum II (cisplatin)-induced nephrotoxicity. We have already demonstrated that cephaloridine, a nephrotoxic antibiotic, leads to extracellular signal-regulated protein kinase (ERK) activation in the rat kidney, which probably contributes to cephaloridine-induced renal dysfunction. The aim of this study was to examine the effect of cisplatin on ERK activation in the rat kidney and also the effect of FTS on cisplatin-induced nephrotoxicity in rats. In vitro treatment of LLC-PK1 cells with FTS significantly ameliorated cisplatin-induced cell injury. Treatment of rats with intravenous cisplatin for 3 days markedly induced renal dysfunction and increased platinum contents in the kidney cortex. An increase in pERK was detected in the nuclear fraction prepared from the rat kidney cortex from days 1 to 3 after injection of cisplatin. FTS suppressed cisplatin-induced renal dysfunction and ERK activation in the kidney. FTS did not influence any Pt contents in the kidney after cisplatin administration. FTS has been shown to enhance the in vivo expression of heat shock protein (HSP) 70 in the kidney cortex. The beneficial role of FTS against cisplatin nephrotoxicity may be mediated in part by HSP70, as suggested by its up-regulation in the kidney cortex treated with FTS alone. Our results suggest that FTS participates in protection from cisplatin-induced nephrotoxicity by suppressing ERK activation caused by cisplatin.


Subject(s)
Cisplatin/toxicity , Extracellular Signal-Regulated MAP Kinases/metabolism , Kidney/drug effects , Thymic Factor, Circulating/pharmacology , Animals , Cell Line , Cell Nucleus/metabolism , Enzyme Activation/drug effects , HSP70 Heat-Shock Proteins/biosynthesis , Kidney/pathology , Kidney/physiology , Phosphorylation , Rats , Swine
12.
J Toxicol Sci ; 30(3): 157-63, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16141650

ABSTRACT

We investigated the effects of a protein kinase C inhibitor and a tyrosine kinase inhibitor on the cellular injury induced by cephaloridine in an established renal epithelial cell line, LLC-PK(1). Cephaloridine increased the leakage of lactate dehydrogenase (LDH) from LLC-PK(1) cells into the medium and also caused an increase in the level of lipid peroxide (index of oxidative stress) in the cells. Treatment of the cells with a hydroxyl radical scavenger, dimethylthiourea (DMTU), inhibited the increases in LDH leakage and lipid peroxidation in LLC-PK(1) cells exposed to cephaloridine. A protein kinase C inhibitor, H-7, and tyrosine kinase inhibitors, genistein and lavendustinA, inhibited the increases in LDH leakage and lipid peroxidation in LLC-PK(1) cells exposed to cephaloridine. These results suggest that a signaling pathway which involves protein kinase C and tyrosine kinase plays a role in the generation of reactive oxygen species in LLC-PK(1) cells damaged by cephaloridine.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Cephaloridine/toxicity , Protein Kinase Inhibitors/pharmacology , Animals , Anti-Bacterial Agents/toxicity , Cell Survival/drug effects , Dose-Response Relationship, Drug , Free Radical Scavengers/pharmacology , Genistein/pharmacology , L-Lactate Dehydrogenase/metabolism , LLC-PK1 Cells , Lipid Peroxidation/drug effects , Phenols/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Swine , Thiobarbituric Acid Reactive Substances/metabolism , Thiourea/analogs & derivatives , Thiourea/pharmacology
13.
J Toxicol Sci ; 30(2): 135-44, 2005 May.
Article in English | MEDLINE | ID: mdl-15928461

ABSTRACT

Zinc is employed as a supplement; however, zinc-related nephropathy is not generally known. In this study, we investigated zinc-induced renal cell injury using a pig kidney-derived cultured renal epithelial cell line, LLC-PK(1), with proximal kidney tubule-like features, and examined the involvement of free radicals and extracellular signal-regulated kinase (ERK) in the cell injury. The LLC-PK(1) cells showed early uptake of zinc (30 microM), and the release of lactate dehydrogenase (LDH), an index of cell injury, was observed 24 hr after uptake. Three hours after zinc exposure, generation of reactive oxygen species (ROS) was increased. An antioxidant, N, N'-diphenyl-p-phenylenediamine (DPPD), inhibited a zinc-related increase in ROS generation and zinc-induced renal cell injury. An NADPH oxidase inhibitor, diphenyleneiodonium (DPI), inhibited a zinc-related increase in ROS generation and cell injury. We investigated translocation from the cytosol fraction of the p67(phox) subunit, which is involved in the activation of NADPH oxidase, to the membrane fraction, and translocation was induced 3 hr after zinc exposure. We examined the involvement of ERK1/2 in the deterioration of zinc-induced renal cell injury, and the association between ERK1/2 and an increase in ROS generation. Six hours after zinc exposure, the activation (phosphorylation) of ERK1/2 was observed. An antioxidant, DPPD, inhibited the zinc-related activation of ERK1/2. An MAPK/ERK kinase (MEK1/2) inhibitor, U0126, almost completely inhibited zinc-related cell injury (the release of LDH), but did not influence ROS generation. These results suggest that early intracellular uptake of zinc by LLC-PK(1) cells causes the activation of NADPH oxidase, and that ROS generation by the activation of the enzyme leads to the deterioration of renal cell injury via the activation of ERK1/2.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , Kidney/drug effects , NADPH Oxidases/physiology , Zinc/toxicity , Animals , Antioxidants/pharmacology , Enzyme Activation , Epithelial Cells/drug effects , Epithelial Cells/pathology , Kidney/pathology , Phenylenediamines/pharmacology , Reactive Oxygen Species , Swine , Zinc/metabolism
14.
Biochem Pharmacol ; 64(3): 543-9, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12147306

ABSTRACT

The development of nephrotoxicity induced by cephaloridine (CER) has been reported to be due to reactive oxygen species (ROS). Protein kinase C (PKC) has been suggested to modulate the generation of ROS. We investigated the possible participation of ROS generation assessed by chemiluminescence (CL) and PKC activity in rat kidney cortical mitochondria in the development of CER-induced nephrotoxicity. We first evaluated the magnitude of the nephrotoxic damage caused by CER in rats. The plasma parameters and ultrastructural morphology changes were increased markedly 24hr after the treatment of rats with CER. We demonstrated that the treatment of rats with CER clearly evoked not only enhancement of Cypridina luciferin analog (CLA)-dependent CL intensity, but also the activation of PKC in mitochondria isolated from the kidney cortex of rats 1.5 and 3.5 hr after injection of the drug. These changes were detected in advance of those observed in plasma and by electron microscopy. The increase in CLA-dependent CL intensity detected in the kidney cortical mitochondria 1.5 and 3.5 hr after injection of CER was inhibited completely by the addition of superoxide dismutase, suggesting the generation of superoxide anion in these mitochondria during the early stages of CER-induced nephrotoxicity. These results suggest that the activation of PKC and the enhancement of superoxide anion generation in kidney cortical mitochondria precede the increases in plasma parameters and the electron micrographic changes indicative of renal dysfunction in rats treated with CER. Additionally, they suggest a possible relationship between PKC activation in mitochondria and free radical-induced CER nephrotoxicity in rats.


Subject(s)
Cephaloridine/toxicity , Cephalosporins/toxicity , Kidney Cortex/drug effects , Mitochondria/drug effects , Protein Kinase C/metabolism , Renal Insufficiency/chemically induced , Animals , Kidney Cortex/cytology , Luminescent Measurements , Male , Mitochondria/enzymology , Mitochondria/metabolism , Pyrazines/chemistry , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Renal Insufficiency/enzymology , Renal Insufficiency/pathology
15.
Hypertens Res ; 25(5): 751-5, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12452329

ABSTRACT

Efonidipine, a calcium antagonist, has been reported to dilate not only afferent but also efferent arterioles, thereby reducing glomerular hydrostatic pressure. We investigated the effect of chronic treatment with efonidipine or lisinopril on the afferent and efferent arteriolar diameters by the vascular cast technique. Four-week-old spontaneously hypertensive rats (SHR) were divided into three groups: untreated, efonidipine (25 mg/kg/day)-treated, and lisinopril (3 mg/kg/day)-treated. At 22 weeks of age, the renal vasculatures were fixed at the maximally dilated condition. The morphometrical measurements showed that the treatments with efonidipine and lisinopril caused structural alteration of the vasculature, resulting in significantly greater efferent arteriolar diameters than in untreated SHR. In addition, lisinopril-treated rats had wider afferent lumina. The renoprotective effect of efonidipine and lisinopril might be partly due to the structurally larger efferent arteriolar lumen.


Subject(s)
Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Hypertension, Renal/drug therapy , Nitrophenols/pharmacology , Organophosphorus Compounds/pharmacology , Renal Circulation/drug effects , Animals , Arterioles/drug effects , Arterioles/ultrastructure , Blood Pressure/drug effects , Body Weight/drug effects , Corrosion Casting , Kidney Glomerulus/blood supply , Male , Rats , Rats, Inbred SHR
16.
Toxicol Lett ; 143(2): 185-94, 2003 Jul 20.
Article in English | MEDLINE | ID: mdl-12749822

ABSTRACT

We have previously reported that free radical-mediated injury induced by cephaloridine (CER) is enhanced by phorbol 12-myristate 13-acetate (PMA), a protein kinase C (PKC) activator, in rat renal cortical slices. We have also shown that PKC activation in mitochondria is involved in CER-induced nephrotoxicity in rats. We investigated the role of a downstream PKC pathway, a MEK/ERK pathway, in free radical-induced injury in rat renal cortical slices exposed to CER. Immediately after preparing slices from rat renal cortex, the slices were incubated in the medium containing MEK inhibitors. ERK1/2 activation was determined by Western blot analysis for phosphorylated ERK (pERK) 1/2 protein in nucleus fraction prepared from the slices exposed to CER. Prominently, CER caused not only increases in lipid peroxidation as an index of free radical generation and in LDH leakage as that of cell injury in the slices, but also marked activation of ERK1/2 in nucleus fraction. PD98059 and U0126, MEK1/2 inhibitors, significantly attenuated CER-induced increases in lipid peroxidation and LDH leakage in the slices. PD98059 also suppressed ERK1/2 activation in nucleus fraction prepared from the slices treated with CER. Inhibition of other MAP kinase pathways, p38 MAP kinase and c-Jun N-terminal kinase (JNK) had no effect on CER-induced increases in lipid peroxidation level and LDH leakage in the slices. The present results suggest that a MEK/ERK pathway down stream of a PKC pathway is probably involved in free radical-induced injury in rat renal cortical slices exposed to CER.


Subject(s)
Cephaloridine/pharmacology , Kidney Cortex/drug effects , Kidney Cortex/pathology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Animals , Antioxidants/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , In Vitro Techniques , Kidney Cortex/enzymology , Kidney Cortex/metabolism , Male , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phenylenediamines/pharmacology , Phosphorylation , Rats , Rats, Sprague-Dawley
17.
Biomed Res ; 32(5): 321-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22033301

ABSTRACT

Ciliary beat frequency (CBF) was measured in slice preparations of the Fallopian tube fimbria, using videomicroscopy with a high-speed (500 Hz) camera in guinea pigs that were treated with ß-oestradiol benzoate (ßE2B) and medroxy progesterone (mPRG). In non-ovulating guinea pigs at 4 weeks of age, the CBF of the fimbria was high (17.8 Hz). In sexually mature guinea pigs (12-16 weeks of age) with constant ovulation, the CBF varied from 12 Hz to 16 Hz. The in vivo administration of both ICI-182,780 (a blocker of ßE2 receptors) and mifepristone (a blocker of PRG receptors) induced high CBF (17.4 Hz). The administration of ßE2B at a low (3.2 mg/kg/day) or high (32 mg/kg/day) dose decreased the CBF to 14.5 Hz or 11 Hz, respectively. ICI-182,780 abolished the ßE2B-induced changes in CBF and decreased CBF to 12 Hz. The administration of mPRG (6.4 mg/kg/day) decreased CBF to 12.5 Hz. Mifepristone abolished this mPRG-induced decrease in CBF and maintained the CBF at 15 Hz. However, administering both ßE2B and mPRG increased CBF to 17.5 Hz, suggesting that ßE2B inhibits mPRG actions and vice versa. To confirm the interactions between ßE2B and mPRG, we administered both ßE2B and mPRG to guinea pigs that were pretreated for 1.5 days with either mPRG (6.4 mg/kg/day) or ßE2B (3.2 mg/kg/day). Prior treatment with ßE2B or mPRG prevented the increase in CBF that was otherwise by ßE2B plus mPRG, and maintained the CBF at 14.5 Hz or 13 Hz, respectively. The administration of ßE2B plus mPRG still induced the expression of PRG receptors, indicating that the highest CBF is not the result of no expression of the receptors. In the beating cilia of the fimbria, the signals that are activated by the ßE2 and PRG receptors are proposed to antagonize each other in regulating the frequency.


Subject(s)
Estradiol/analogs & derivatives , Fallopian Tubes/drug effects , Fallopian Tubes/physiology , Medroxyprogesterone/pharmacology , Ovary/physiology , Animals , Cilia/drug effects , Cilia/physiology , Drug Interactions , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Fornix, Brain/metabolism , Guinea Pigs , Hormone Antagonists/metabolism , Humans , In Vitro Techniques , Medroxyprogesterone/metabolism , Receptors, Progesterone/metabolism
18.
J Nutr Sci Vitaminol (Tokyo) ; 56(6): 335-46, 2010.
Article in English | MEDLINE | ID: mdl-21422702

ABSTRACT

We previously found that thiamine mitigates metabolic disorders in spontaneously hypertensive rats, harboring defects in glucose and fatty acid metabolism. Mutation of thiamine transporter gene SLC19A2 is linked to type 2 diabetes mellitus. The current study extends our hypothesis that thiamine intervention may impact metabolic abnormalities in Otsuka Long-Evans Tokushima Fatty (OLETF) rats, exhibiting obesity and metabolic disorders similar to human metabolic syndrome. Male OLETF rats (4 wk old) were given free access to water containing either 0.2% or 0% of thiamine for 21 and 51 wk. At the end of treatment, blood parameters and cardiac functions were analyzed. After sacrifice, organs weights, histological findings, and hepatic pyruvate dehydrogenase (PDH) activity in the liver were evaluated. Thiamine intervention averted obesity and prevented metabolic disorders in OLETF rats which accompanied mitigation of reduced lipid oxidation and increased hepatic PDH activity. Histological evaluation revealed that thiamine alleviated adipocyte hypertrophy, steatosis in the liver, heart, and skeletal muscle, sinusoidal fibrosis with formation of basement membranes (called pseudocapillarization) which accompanied significantly reduced expression of laminin ß1 and nidogen-1 mRNA, interstitial fibrosis in the heart and kidney, fatty degeneration in the pancreas, thickening of the basement membrane of the vasculature, and glomerulopathy and mononuclear cell infiltration in the kidney. Cardiac and renal functions were preserved in thiamine treatment. Thiamine has a potential to prevent obesity and metabolic disorders in OLETF rats.


Subject(s)
Adipocytes/drug effects , Lipid Peroxidation/drug effects , Liver/drug effects , Metabolic Diseases/prevention & control , Obesity/prevention & control , Thiamine/therapeutic use , Vitamin B Complex/therapeutic use , Adipocytes/pathology , Animals , Basement Membrane/drug effects , Blood Vessels/drug effects , Blood Vessels/pathology , Fibrosis/drug therapy , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Laminin/genetics , Laminin/metabolism , Leukocytes, Mononuclear/drug effects , Liver/metabolism , Liver/pathology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Myocardium/metabolism , Myocardium/pathology , Obesity/metabolism , Obesity/pathology , Oxidoreductases/metabolism , Pancreas/drug effects , Pancreas/pathology , RNA/metabolism , Rats , Rats, Inbred OLETF , Thiamine/pharmacology , Vitamin B Complex/pharmacology
19.
J Toxicol Sci ; 33(4): 459-72, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18827445

ABSTRACT

Diabetic cardiomyopathy can progress toward overt heart failure with increased mortality. The hexosamine biosynthesis pathway has been implicated in signaling for fibrosis by the kidney. Thiamine (vitamin B(1)) is an indispensable coenzyme and required at intracellular glucose metabolism. In this study, we assessed if decrease of flux through the hexosamine biosynthesis pathway induced by high-dose thiamine therapy counteracts diabetes-induced cardiac fibrosis. The diabetes model used was the streptozotocin-induced diabetic rat. Normal control and diabetic rats were studied for 2 weeks with and without thiamine, and followings were analyzed; plasma biochemicals (total cholesterol and triglycerides), morphological changes, mRNA abundance relevant to cardiac failure (brain natriuretic peptide) and fibrosis (transforming growth factor-beta1, thrombospondine, fibronectin, plasminogen activator-I and connective tissue growth factor) as well as and matrix metalloproteinase activity were investigated. Thiamine repletion prevented diabetes-induced cardiac fibrosis without changes in plasma glucose concentration. This was achieved by prevention of thiamine depletion, increased pro-fibrotic mRNA abundance and decreased metalloproteinase activity in the heart of diabetic rats. O-glycosylated protein was significantly higher in the left ventricular of diabetic rats compared to control rats, which was decreased by thiamine administration. Thiamine repletion prevented diabetes-induced cardiac fibrosis in experimental diabetes, probably by suppression of hexosamine biosynthesis pathway.


Subject(s)
Cardiomyopathies/etiology , Cardiomyopathies/prevention & control , Diabetes Mellitus, Experimental/complications , Thiamine/therapeutic use , Animals , Blood Glucose , Fibrosis , Glycosylation , Hexosamines/biosynthesis , Male , Myocardium/pathology , Rats , Rats, Wistar , Streptozocin , Thiamine/blood
20.
J Pharmacol Sci ; 100(1): 65-72, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16410676

ABSTRACT

We investigated the involvement of reactive oxygen species (ROS) and intracellular calcium in nephrotoxicity related to an antitumor agent, cisplatin. In this study, we employed cultured renal epithelial cells (LLC-PK1). Cisplatin at 500 microM significantly increased the production of ROS 5 h and caused cell injury. This agent significantly increased the intracellular calcium level ([Ca2+]i) in a dose-dependent manner 1 h or more after exposure. DPPD (N,N'-diphenyl-p-phenylenediamine), an antioxidant, inhibited a cisplatin-related increase in active oxygen production and cell injury but did not inhibit an early increase in the [Ca2+]i level. An intracellular calcium-chelating compound BAPTA-AM (1,2-bis(O-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetra(acetoxymethyl) ester) inhibited an increase in ROS production and cell injury induced by cisplatin. Furthermore, BAPTA-AM suppressed the rise of [Ca2+]i level in 1 h after exposure; however, an extracellular calcium chelator EGTA and a calcium antagonist nicardipine did not inhibit the rise in [Ca2+]i level in the early phase. An NADPH oxidase inhibitor inhibited a cisplatin-related increase in ROS production and cell disorder. These results suggest that cisplatin-related calcium release from the site of intracellular calcium storage in the early phase causes oxidative stress in renal tubular epithelial cells. Cisplatin may increase the intracellular production of ROS via NADPH oxidase.


Subject(s)
Calcium Signaling , Cisplatin/toxicity , Epithelial Cells/drug effects , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents/toxicity , Antioxidants/pharmacology , Cell Line , Cell Survival , Chelating Agents/pharmacology , Dose-Response Relationship, Drug , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/enzymology , Kidney Tubules/drug effects , Kidney Tubules/enzymology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Onium Compounds/pharmacology , Phenylenediamines/pharmacology , Swine , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL