Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Biol Pharm Bull ; 47(7): 1350-1359, 2024.
Article in English | MEDLINE | ID: mdl-39085074

ABSTRACT

Indigo naturalis (IN), derived from the leaves of the indigo plant, is a traditional Chinese medicine that has historically been used for its anti-inflammatory properties in the treatment of various diseases, including ulcerative colitis (UC). However, long-term use of IN in UC patients is incontrovertibly associated with the onset of pulmonary arterial hypertension (PAH). To investigate the mechanisms by which IN induces PAH, we focused on the raw material of IN, indigo leaves (IL). Only the condition of long-term chronic (6 months) and high-dose (containing 5% IL in the control diet) administration of IL induced medial thickening in the pulmonary arteries without right ventricular hypertrophy in our rat model. IL administration for a month did not induce pulmonary arterial remodeling but increased endothelin-1 (ET-1) expression levels within endothelial cell (EC) layers in the lungs. Gene Expression Omnibus analysis showed that ET-1 is a key regulator of PAH and that the IL component indican and its metabolite IS induced ET-1 mRNA expression via reactive oxygen species-dependent mechanism. We identified the roles of indican and IS in ET-1 expression in ECs, which were linked to pulmonary arterial remodeling in an animal model.


Subject(s)
Endothelin-1 , Hypertrophy, Right Ventricular , Plant Leaves , Pulmonary Artery , Rats, Sprague-Dawley , Vascular Remodeling , Animals , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Male , Endothelin-1/metabolism , Vascular Remodeling/drug effects , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Rats , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Lung/drug effects , Lung/pathology , Lung/metabolism
2.
Life Sci ; 321: 121590, 2023 May 15.
Article in English | MEDLINE | ID: mdl-36940907

ABSTRACT

AIMS: Peroxisome proliferator-activated receptor-alpha (PPARα) levels are markedly lower in the kidneys of chronic kidney disease (CKD) patients. Fibrates (PPARα agonists) are therapeutic agents against hypertriglyceridemia and potentially against CKD. However, conventional fibrates are eliminated by renal excretion, limiting their use in patients with impaired renal function. Here, we aimed to evaluate the renal risks associated with conventional fibrates via clinical database analysis and investigate the renoprotective effects of pemafibrate, a novel selective PPARα modulator mainly excreted into the bile. MAIN METHODS: The risks associated with conventional fibrates (fenofibrate, bezafibrate) to the kidneys were evaluated using the Food and Drug Administration Adverse Event Reporting System. Pemafibrate (1 or 0.3 mg/kg/day) was administered daily using an oral sonde. Its renoprotective effects were examined in unilateral ureteral obstruction (UUO)-induced renal fibrosis model mice (UUO mice) and adenine-induced CKD model mice (CKD mice). KEY FINDINGS: The ratios of glomerular filtration rate decreased and blood creatinine increased were markedly higher after conventional fibrate use. Pemafibrate administration suppressed increased gene expressions of collagen-I, fibronectin, and interleukin 1 beta (IL-1ß) in the kidneys of UUO mice. In CKD mice, it suppressed increased plasma creatinine and blood urea nitrogen levels and decreased red blood cell count, hemoglobin, and hematocrit levels, along with renal fibrosis. Moreover, it inhibited the upregulation of monocyte chemoattractant protein-1, IL-1ß, tumor necrosis factor-alpha, and IL-6 in the kidneys of CKD mice. SIGNIFICANCE: These results demonstrated the renoprotective effects of pemafibrate in CKD mice, confirming its potential as a therapeutic agent for renal disorders.


Subject(s)
Fenofibrate , Renal Insufficiency, Chronic , Ureteral Obstruction , Mice , Animals , PPAR alpha/metabolism , Creatinine/metabolism , Kidney/metabolism , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/complications , Fenofibrate/pharmacology , Fibrosis , Ureteral Obstruction/pathology
3.
J Trace Elem Med Biol ; 67: 126798, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34087581

ABSTRACT

BACKGROUND: Cisplatin is widely used as an antitumor drug for the treatment of solid tumors. However, its use has been limited owing to nephrotoxicity, a major side effect. The mechanism of cisplatin-induced nephrotoxicity (CIN) has long been investigated in order to develop preventive/therapeutic drugs. Ferroptosis is a newly identified form of non-apoptotic regulated cell death induced by iron-mediated lipid peroxidation and is involved in the pathophysiology of various diseases. In this study, we examined the role of ferroptosis in CIN. METHODS: We evaluated the role of ferroptosis in CIN by in vivo experiments in a mouse model. RESULTS: Cisplatin increased the protein expressions of transferrin receptor-1 and ferritin, and iron content in the kidney of mice. In addition, treatment with cisplatin augmented renal ferrous iron and hydroxyl radical levels with co-localization. Mice administered cisplatin demonstrated kidney injury, with renal dysfunction and increased inflammatory cytokine expression; these changes were ameliorated by Ferrostatin-1 (Fer-1), an inhibitor of ferroptosis. The expression of the ferroptosis markers, COX2 and 4-hydroxynonenal (4-HNE), increased with cisplatin administration, and decreased with the administration of Fer-1. By contrast, cisplatin-induced apoptosis and necroptosis were inhibited by treatment with Fer-1. Moreover, deferoxamine, an iron chelator, also inhibited CIN, with a decrease in the expression of COX-2 and 4-HNE. CONCLUSION: Ferroptosis is involved in the pathogenesis of CIN and might be used as a new preventive target for CIN.


Subject(s)
Cisplatin/toxicity , Ferroptosis , Animals , Ferritins , Iron/metabolism , Lipid Peroxidation , Mice
4.
Eur J Pharmacol ; 902: 174099, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-33910036

ABSTRACT

Despite the availability of more than 20 clinical antiepileptic drugs, approximately 30% of patients with epilepsy do not respond to antiepileptic drug treatment. Therefore, it is important to develop antiepileptic products that function via novel mechanisms. In the present study, we evaluated data from one of the largest global databases to identify drugs with antiepileptic effects, and subsequently attempted to understand the effect of the combination of antiepileptic drugs and valacyclovir in epileptic seizures using a kindling model. To induce kindling in mice, pentylenetetrazol at a dose of 40 mg/kg was administered once every 48 h. Valacyclovir was orally administered 30 min before antiepileptic drug injection in kindled mice, and behavioral seizures were monitored for 20 min following pentylenetetrazol administration. Additionally, c-Fos expression in the hippocampal dentate gyrus was measured in kindled mice. Valacyclovir showed inhibitory effects on pentylenetetrazol-induced kindled seizures. In addition, simultaneous use of levetiracetam and valacyclovir caused more potent inhibition of seizure activity, and neither valproic acid nor diazepam augmented the anti-seizure effect in kindled mice. Furthermore, kindled mice showed increased c-Fos levels in the dentate gyrus. The increase in c-Fos expression was significantly inhibited by the simultaneous use of levetiracetam and valacyclovir. The findings of the present study indicate that a combination of levetiracetam and valacyclovir had possible anticonvulsive effects on pentylenetetrazol-induced kindled epileptic seizures. These results suggest that valacyclovir may have an antiseizure effect in patients with epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Kindling, Neurologic/drug effects , Seizures/drug therapy , Valacyclovir/pharmacology , Animals , Anticonvulsants/therapeutic use , Cefepime/adverse effects , Databases, Factual , Disease Models, Animal , Drug Repositioning , Drug Therapy, Combination , Hippocampus/drug effects , Humans , Levetiracetam/pharmacology , Levetiracetam/therapeutic use , Male , Mice , Pentylenetetrazole/toxicity , Proto-Oncogene Proteins c-fos/metabolism , Seizures/chemically induced , Valacyclovir/therapeutic use
5.
Biol Pharm Bull ; 44(4): 478-484, 2021.
Article in English | MEDLINE | ID: mdl-33790099

ABSTRACT

Patients who undergo multiple-day chemotherapy sessions experience hard-to-treat nausea and vomiting. Currently, there is no effective standard treatment for this condition. This study compared the preventive effect of first-generation 5-hydroxytryptamine 3 receptor antagonists (5-HT3 RAs) and second-generation 5-HT3 RAs palonosetron in multiple-day chemotherapy-induced nausea and vomiting. The design of this study was a retrospective case-control study of patients who received a five-day cisplatin-based chemotherapy and were treated with aprepitant, dexamethasone, granisetron, and ramosetron or palonosetron. The patients were divided into two groups: patients given granisetron and ramosetron (the first-generation group), and those given palonosetron (palonosetron group). The percentage of patients with a complete response or total control was assessed. They were divided into three phases: 0-216 h (overall phase), 0-120 h (remedial phase), and 120-216 h (after phase). The remedial phase was further divided into 0-24 h (early phase) and 24-120 h (later phase). Moreover, the nutritional status of each patient was assessed by noting the patients' total calorie-intake per day and total parenteral nutrition. First-generation 5-HT3 RAs and palonosetron were used for treatment in 18 and 28 patients, respectively. The complete response rate and caloric oral intake of the later phase were higher in the palonosetron group than in the first-generation group. We conclude that palonosetron treatment was more effective than first-generation 5-HT3 RAs in controlling multiple-day chemotherapy-induced nausea and vomiting.


Subject(s)
Antiemetics/administration & dosage , Benzimidazoles/administration & dosage , Granisetron/administration & dosage , Nausea/drug therapy , Palonosetron/administration & dosage , Serotonin 5-HT3 Receptor Antagonists/administration & dosage , Vomiting/drug therapy , Adult , Antineoplastic Agents/adverse effects , Bleomycin/adverse effects , Drug Therapy, Combination , Etoposide/adverse effects , Female , Humans , Male , Middle Aged , Nausea/chemically induced , Neoplasms, Germ Cell and Embryonal/drug therapy , Ovarian Neoplasms/drug therapy , Platinum Compounds/adverse effects , Retrospective Studies , Testicular Neoplasms/drug therapy , Vomiting/chemically induced
6.
Kidney Int ; 99(4): 885-899, 2021 04.
Article in English | MEDLINE | ID: mdl-33307103

ABSTRACT

Cisplatin is widely used as an anti-tumor drug for the treatment of solid tumors. Unfortunately, it causes kidney toxicity as a critical side effect, limiting its use, given that no preventive drug against cisplatin-induced kidney toxicity is currently available. Here, based on a repositioning analysis of the Food and Drug Administration Adverse Events Reporting System, we found that a previously developed drug, diphenhydramine, may provide a novel treatment for cisplatin-induced kidney toxicity. To confirm this, the actual efficacy of diphenhydramine was evaluated in in vitro and in vivo experiments. Diphenhydramine inhibited cisplatin-induced cell death in kidney proximal tubular cells. Mice administered cisplatin developed kidney injury with significant dysfunction (mean plasma creatinine: 0.43 vs 0.15 mg/dl) and showed augmented oxidative stress, increased apoptosis, elevated inflammatory cytokines, and MAPKs activation. However, most of these symptoms were suppressed by treatment with diphenhydramine. Furthermore, the concentration of cisplatin in the kidney was significantly attenuated in diphenhydramine-treated mice (mean platinum content: 70.0 vs 53.4 µg/g dry kidney weight). Importantly, diphenhydramine did not influence or interfere with the anti-tumor effect of cisplatin in any of the in vitro or in vivo experiments. In a selected cohort of 98 1:1 matched patients from a retrospective database of 1467 patients showed that patients with malignant cancer who had used diphenhydramine before cisplatin treatment exhibited significantly less acute kidney injury compared to ones who did not (6.1 % vs 22.4 %, respectively). Thus, diphenhydramine demonstrated efficacy as a novel preventive medicine against cisplatin-induced kidney toxicity.


Subject(s)
Acute Kidney Injury , Antineoplastic Agents , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Animals , Antineoplastic Agents/toxicity , Apoptosis , Cisplatin/toxicity , Diphenhydramine/metabolism , Diphenhydramine/pharmacology , Diphenhydramine/therapeutic use , Humans , Kidney/metabolism , Mice , Oxidative Stress , Retrospective Studies
7.
Cancer Med ; 10(1): 164-172, 2021 01.
Article in English | MEDLINE | ID: mdl-33231381

ABSTRACT

BACKGROUND: In patients treated with bevacizumab, hypertension may be a biomarker of therapeutic efficacy. However, it is not clear whether drugs that control blood pressure influence bevacizumab's efficacy. In this study, we investigated drugs that may affect hypertension in bevacizumab-treated patients and examined the impact on the therapeutic effect. PATIENTS AND METHODS: We analyzed 3,724,555 reports from the third quarter of 2010 to the second quarter of 2015. All data were obtained from the Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) analysis. In this retrospective cohort study, we investigated a total of 58 patients diagnosed with colorectal cancer and treated for the first time with bevacizumab containing XELOX or mFOLFOX6 at The University of Tokushima Hospital between January 2010 and December 2015. The effect of the treatment was evaluated according to Response Evaluation Criteria in Solid Tumors version 1.0. Thereafter, the effect was confirmed using Gene Expression Omnibus (GEO) and cultured cells. RESULTS: There are few reports in FAERS of hypertension in patients treated with omeprazole on bevacizumab. Based on the chart review, patients who used proton pump inhibitors (PPI) had a lower response to treatment than those who did not (response rate: 25% vs 50%). Furthermore, experiments on GEO and cell lines suggested that induction of vascular endothelial growth factor (VEGF) gene expression by PPIs is the cause of the reduced therapeutic effect. CONCLUSION: PPIs prevent hypertension in bevacizumab-treated patients but may reduce bevacizumab's anti-tumoral effects by inducing VEGF expression.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Antihypertensive Agents/therapeutic use , Antineoplastic Agents, Immunological/adverse effects , Bevacizumab/adverse effects , Blood Pressure/drug effects , Colorectal Neoplasms/drug therapy , Hypertension/prevention & control , Proton Pump Inhibitors/therapeutic use , Adult , Adverse Drug Reaction Reporting Systems , Aged , Aged, 80 and over , Antihypertensive Agents/adverse effects , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Databases, Factual , Drug Interactions , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypertension/chemically induced , Hypertension/diagnosis , Hypertension/physiopathology , Japan , Male , Middle Aged , Proton Pump Inhibitors/adverse effects , Retrospective Studies , Treatment Outcome , United States , United States Food and Drug Administration , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
8.
Diabetologia ; 63(8): 1588-1602, 2020 08.
Article in English | MEDLINE | ID: mdl-32430665

ABSTRACT

AIMS/HYPOTHESIS: Iron accumulation affects obesity and diabetes, both of which are ameliorated by iron reduction. Ferritin, an iron-storage protein, plays a crucial role in iron metabolism. H-ferritin exerts its cytoprotective action by reducing toxicity via its ferroxidase activity. We investigated the role of macrophage H-ferritin in obesity and diabetes. METHODS: Conditional macrophage-specific H-ferritin (Fth, also known as Fth1) knockout (LysM-Cre Fth KO) mice were used and divided into four groups: wild-type (WT) and LysM-Cre Fth KO mice with normal diet (ND), and WT and LysM-Cre Fth KO mice with high-fat diet (HFD). These mice were analysed for characteristics of obesity and diabetes, tissue iron content, inflammation, oxidative stress, insulin sensitivity and metabolic measurements. RAW264.7 macrophage cells were used for in vitro experiments. RESULTS: Iron concentration reduced, and mRNA expression of ferroportin increased, in macrophages from LysM-Cre Fth KO mice. HFD-induced obesity was lower in LysM-Cre Fth KO mice than in WT mice at 12 weeks (body weight: KO 34.6 ± 5.6 g vs WT 40.1 ± 5.2 g). mRNA expression of inflammatory cytokines and infiltrated macrophages and oxidative stress increased in the adipose tissue of HFD-fed WT mice, but was not elevated in HFD-fed LysM-Cre Fth KO mice. However, WT mice fed an HFD had elevated iron concentration in adipose tissue and spleen, which was not observed in LysM-Cre Fth KO mice fed an HFD (adipose tissue [µmol Fe/g protein]: KO 1496 ± 479 vs WT 2316 ± 866; spleen [µmol Fe/g protein]: KO 218 ± 54 vs WT 334 ± 83). Moreover, HFD administration impaired both glucose tolerance and insulin sensitivity in WT mice, which was ameliorated in LysM-Cre Fth KO mice. In addition, energy expenditure, mRNA expression of thermogenic genes, and body temperature were higher in KO mice with HFD than WT mice with HFD. In vitro experiments showed that iron content was reduced, and lipopolysaccharide-induced Tnf-α (also known as Tnf) mRNA upregulation was inhibited in a macrophage cell line transfected with Fth siRNA. CONCLUSIONS/INTERPRETATION: Deletion of macrophage H-ferritin suppresses the inflammatory response by reducing intracellular iron levels, resulting in the prevention of HFD-induced obesity and diabetes. The findings from this study highlight macrophage iron levels as a potential therapeutic target for obesity and diabetes.


Subject(s)
Apoferritins/metabolism , Diabetes Mellitus/metabolism , Diabetes Mellitus/therapy , Diet, High-Fat/adverse effects , Macrophages/metabolism , Obesity/metabolism , Obesity/therapy , Animals , Apoferritins/genetics , Diabetes Mellitus/etiology , Male , Mice , Mice, Knockout , Obesity/etiology , Random Allocation
9.
Naunyn Schmiedebergs Arch Pharmacol ; 393(7): 1239-1250, 2020 07.
Article in English | MEDLINE | ID: mdl-32307577

ABSTRACT

The roles of cancer-associated fibroblasts (CAFs) have been studied in the tumor progression, and CAFs are expected to become the new targets for cancer pharmacotherapies. CAFs contribute to tumor cell survival and proliferation, tumor angiogenesis, immune suppression, tumor inflammation, tumor cell invasion and metastasis, and extracellular matrix remodeling. However, detailed mechanisms of how CAFs function in the living system remain unclear. CAFs include α-smooth muscle actin, expressing activated fibroblasts similar to myofibroblasts, and are highly capable of producing collagen. Several reports have demonstrated the contributions of extracellular-signal-regulated kinase 5 (ERK5) in fibroblasts to the fibrotic processes; however, the roles of CAF-derived ERK5 remain unclear. To investigate the roles of CAF-derived ERK5 in the tumor progression, we created mice lacking the ERK5 gene specifically in fibroblasts. Colon-26 mouse colon cancer cells were implanted into the mice subcutaneously, and the histological analyses of the tumor tissue were performed after 2 weeks. Immunofluorescence analyses showed that recipient-derived fibroblasts existed within the tumor tissue. The present study demonstrated that fibroblast-specific ERK5 deficiency exacerbated tumor progression and it was accompanied with thicker tumor vessel formation and the increase in the number of activated fibroblasts. We combined the results of The Cancer Genome Atlas (TCGA) database analysis with our animal studies, and indicated that regulating ERK5 activity in CAFs or CAF invasion into the tumor tissue can be important strategies for the development of new targets in cancer pharmacotherapies.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Colonic Neoplasms/pathology , Mitogen-Activated Protein Kinase 7/genetics , Animals , Cell Line, Tumor , Colonic Neoplasms/blood supply , Colonic Neoplasms/genetics , Disease Progression , Female , Male , Mice , Mice, Knockout
10.
J Pharmacol Sci ; 142(3): 109-115, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31882204

ABSTRACT

Arterial calcification, a risk factor of cardiovascular events, develops with differentiation of vascular smooth muscle cells (VSMCs) into osteoblast-like cells. Cyclophilin A (CypA) is a peptidyl-prolyl isomerase involved in cardiovascular diseases such as atherosclerosis and aortic aneurysms, and rho-associated protein kinase (ROCK) is involved in the pathogenesis of vascular calcification. CypA is secreted in a ROCK activity-dependent manner and works as a mitogen via autocrine or paracrine mechanisms in VSMCs. We examined the involvement of the ROCK-CypA axis in VSMC calcification induced by inorganic phosphate (Pi), a potent cell mineralization initiator. We found that Pi stimulated ROCK activity, CypA secretion, extracellular signal-regulated protein kinase (ERK) 1/2 phosphorylation, and runt-related transcription factor 2 expression, resulting in calcium accumulation in rat aortic smooth muscle cells (RASMCs). The ROCK inhibitor Y-27632 significantly suppressed Pi-induced CypA secretion, ERK1/2 phosphorylation, and calcium accumulation. Recombinant CypA was found to be associated with increased calcium accumulation in RASMCs. Based on these results, we suggest that autocrine CypA is mediated by ROCK activity and is involved in Pi-induced ERK1/2 phosphorylation following calcification signaling in RASMCs.


Subject(s)
Calcinosis/genetics , Cyclophilins , Muscle, Smooth, Vascular/pathology , Phosphates/pharmacology , Signal Transduction , rho-Associated Kinases , Animals , Cells, Cultured , Male , Rats, Sprague-Dawley
11.
Toxicol Lett ; 318: 86-91, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31669099

ABSTRACT

Proton pump inhibitors (PPIs) have been used worldwide to treat gastrointestinal disorders. A recent study showed that long-term use of PPIs caused iron deficiency; however, it is unclear whether PPIs affect iron metabolism directly. We investigated the effect of PPIs on the peptide hepcidin, an important iron regulatory hormone. First, we used the FDA Adverse Event Reporting System database and analyzed the influence of PPIs. We found that PPIs, as well as H2 blockers, increased the odds ratio of iron-deficient anemia. Next, HepG2 cells were used to examine the action of PPIs and H2 blockers on hepcidin. PPIs augmented hepcidin expression, while H2 blockers did not. In fact, the PPI omeprazole increased hepcidin secretion, and omeprazole-induced hepcidin upregulation was inhibited by gene silencing or the pharmacological inhibition of the aryl hydrocarbon receptor. In mouse experiments, omeprazole also increased hepatic hepcidin mRNA expression and blood hepcidin levels. In mice treated with omeprazole, protein levels of duodenal and splenic ferroportin decreased. Taken together, PPIs directly affect iron metabolism by suppressing iron absorption through the inhibition of duodenal ferroportin via hepcidin upregulation. These findings provide a new insight into the molecular mechanism of PPI-induced iron deficiency.


Subject(s)
Anemia, Iron-Deficiency/chemically induced , Basic Helix-Loop-Helix Transcription Factors/metabolism , Duodenum/drug effects , Hepatocytes/drug effects , Hepcidins/metabolism , Intestinal Absorption/drug effects , Iron/blood , Proton Pump Inhibitors/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/physiopathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cation Transport Proteins/metabolism , Duodenum/metabolism , Duodenum/physiopathology , Hep G2 Cells , Hepatocytes/metabolism , Histamine H2 Antagonists/toxicity , Humans , Iron Deficiencies , Male , Mice, Inbred C57BL , Receptors, Aryl Hydrocarbon/genetics
12.
Nihon Yakurigaku Zasshi ; 154(6): 316-321, 2019.
Article in Japanese | MEDLINE | ID: mdl-31787683

ABSTRACT

Iron is an essential trace metal element for maintaining vital functions, and it is involved in hemoglobin synthesis, redox reaction, enzyme activity, cell proliferation and apoptosis in various cells. Iron deficient-related diseases represented anemia are well-known, on the other hand, iron overload disease has attracted little attention. Excessive iron produces hydroxyl radicals via Fenton/Haber-Weiss reaction, causing organ damage in hereditary iron overload diseases. Additionally, it has been clarified that iron accumulation is involved in the pathological conditions even in metabolic diseases thought to be unrelated to iron so far. Therefore, the role of iron in the living body has been raised attention again. Recent studies have reported that body iron content is associated with both obesity and diabetes, and iron might be an aggravating factor of obesity and diabetes. We have revealed that iron chelating agent reduced oxidative stress and inflammation, suppressing the development of adipose hypertrophy in KKAy mice. Dietary iron restriction also diminishes oxidative stress, leading to the inhibition of increased albuminuria excretion and glomerular lesions in db/db mice. In this review, we give an outline of the role of iron on obese and diabetes, and diabetic kidney disease, and present the possibility of application to treatment with iron regulation in those disorders.


Subject(s)
Diabetes Mellitus/physiopathology , Iron/physiology , Obesity/physiopathology , Animals , Diabetic Nephropathies/physiopathology , Humans , Iron, Dietary , Mice , Oxidative Stress
13.
FASEB J ; 33(8): 9551-9564, 2019 08.
Article in English | MEDLINE | ID: mdl-31145863

ABSTRACT

Skeletal muscle atrophy is caused by disruption in the homeostatic balance of muscle degeneration and regeneration under various pathophysiological conditions. We have previously reported that iron accumulation induces skeletal muscle atrophy via a ubiquitin ligase-dependent pathway. However, the potential effect of iron accumulation on muscle regeneration remains unclear. To examine the effect of iron accumulation on myogenesis, we used a mouse model with cardiotoxin (CTX)-induced muscle regeneration in vivo and C2C12 mouse myoblast cells in vitro. In mice with iron overload, the skeletal muscles exhibited increased oxidative stress and decreased expression of satellite cell markers. Following CTX-induced muscle injury, these mice also displayed delayed muscle regeneration with a decrease in the size of regenerating myofibers, reduced expression of myoblast differentiation markers, and decreased phosphorylation of MAPK signaling pathways. In vitro, iron overload also suppressed the differentiation of C2C12 myoblast cells but the suppression could be reversed by superoxide scavenging using tempol. Excess iron inhibits myogenesis via oxidative stress, leading to an imbalance in skeletal muscle homeostasis.-Ikeda, Y., Satoh, A., Horinouchi, Y., Hamano, H., Watanabe, H., Imao, M., Imanishi, M., Zamami, Y., Takechi, K., Izawa-Ishizawa, Y., Miyamoto, L., Hirayama, T., Nagasawa, H., Ishizawa, K., Aihara, K.-I., Tsuchiya, K., Tamaki, T. Iron accumulation causes impaired myogenesis correlated with MAPK signaling pathway inhibition by oxidative stress.


Subject(s)
Iron/metabolism , Muscle, Skeletal/metabolism , Oxidative Stress/physiology , RNA, Messenger/metabolism , Animals , Blotting, Western , Cell Line , Cell Survival/physiology , Hydroxyl Radical/metabolism , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Muscle Development/physiology , Thiobarbituric Acid Reactive Substances/metabolism
14.
Am J Hypertens ; 32(3): 249-256, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30351343

ABSTRACT

BACKGROUND: Several reports from basic researches and clinical studies have suggested that xanthine oxidase (XO) inhibitors have suppressive effects on cardiovascular diseases. However, the roles of a XO inhibitor, febuxostat (FEB), in the pathogenesis of vascular remodeling and hypertension independent of the serum uric acid level remain unclear. METHODS: To induce vascular remodeling in mice, angiotensin II (Ang II) was infused for 2 weeks with a subcutaneously implanted osmotic minipump. FEB was administered every day during Ang II infusion. Aortic fibrosis was assessed by elastica van Gieson staining. Mouse macrophage RAW264.7 cells (RAW) and mouse embryonic fibroblasts were used for in vitro studies. RESULTS: FEB suppressed Ang II-induced blood pressure elevation and aortic fibrosis. Immunostaining showed that Ang II-induced macrophage infiltration in the aorta tended to be suppressed by FEB, and XO was mainly colocalized in macrophages, not in fibroblasts. Transforming growth factor-ß1 (TGF-ß1) mRNA expression was induced in the aorta in the Ang II alone group, but not in the Ang II + FEB group. Ang II induced α-smooth muscle actin-positive fibroblasts in the aortic wall, but FEB suppressed them. XO expression and activity were induced by Ang II stimulation alone but not by Ang II + FEB in RAW. FEB suppressed Ang II-induced TGF-ß1 mRNA expression in RAW. CONCLUSIONS: Our results suggested that FEB ameliorates Ang II-induced aortic fibrosis via suppressing macrophage-derived TGF-ß1 expression.


Subject(s)
Aortic Diseases/drug therapy , Febuxostat/therapeutic use , Gout Suppressants/therapeutic use , Hypertension/complications , Macrophages/drug effects , Vascular Remodeling/drug effects , Actins/metabolism , Adventitia/cytology , Adventitia/metabolism , Angiotensin II , Animals , Aorta/drug effects , Aorta/pathology , Aortic Diseases/etiology , Disease Models, Animal , Febuxostat/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Gout Suppressants/pharmacology , Hypertension/chemically induced , Macrophages/metabolism , Male , Mice, Inbred C57BL , Transforming Growth Factor beta1/metabolism , Xanthine Oxidase/antagonists & inhibitors
15.
J Hypertens ; 37(1): 73-83, 2019 01.
Article in English | MEDLINE | ID: mdl-30303488

ABSTRACT

OBJECTIVE: Aortic dissection is a life-threatening disease. At present, the only therapeutic strategies available are surgery and antihypertensive drugs. Moreover, the molecular mechanisms underlying the onset of aortic dissection are still unclear. We established a novel aortic dissection model in mice using pharmacologically induced endothelial dysfunction. We then used the Japanese Adverse Drug Event Report database to investigate the role of pitavastatin in preventing the onset of aortic dissection. METHODS AND RESULTS: To induce endothelial dysfunction, Nω-nitro-L-arginine methyl ester, a nitric oxide synthase inhibitor, was administered to C57BL/6 mice. Three weeks later, angiotensin II (Ang II) and ß-aminopropionitrile (BAPN), a lysyl oxidase inhibitor, were administered with osmotic mini-pumps. False lumen formation was used as the pathological determinant of aortic dissection. The incidences of aortic dissection and death from aneurysmal rupture were significantly higher in the Nω-nitro-L-arginine methyl ester, Ang II, and BAPN (LAB) group than they were in the Ang II and BAPN (AB) group.Pitavastatin was administered orally to LAB mice. It significantly lowered the incidences of dissection and rupture. It also decreased inflammation and medial degradation, both of which were exacerbated in the LAB group. The Japanese Adverse Drug Event Report database analysis indicated that there were 113 cases of aortic dissection out of 95 090 patients (0.12%) not receiving statins but only six cases out of 16 668 patients receiving statins (0.04%) (odds ratio: 0.30; P = 0.0043). CONCLUSION: Our results suggest that endothelial dysfunction is associated with the onset of aortic dissection and pitavastatin can help prevent this condition.


Subject(s)
Aortic Dissection , Disease Models, Animal , Aortic Dissection/drug therapy , Aortic Dissection/physiopathology , Aortic Dissection/prevention & control , Animals , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Mice , Mice, Inbred C57BL , Quinolines/therapeutic use
16.
Support Care Cancer ; 27(3): 849-856, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30062585

ABSTRACT

PURPOSE: SN-38, an active metabolite of irinotecan, is reabsorbed by the intestinal tract during excretion, causing diarrhoea and neutropenia. In addition, the association between blood levels of SN-38 and neutropenia has been reported previously, and the rapid excretion of SN-38 from the intestinal tract is considered to prevent neutropenia. Oral alkalization drugs are used as prophylactic agents for suppressing SN-38 reabsorption. The relationship between oral alkalization drugs and neutropenia, however, has not been well studied. The aim of this study was to investigate the relationship between oral alkalization drugs and neutropenia in irinotecan-treated patients. METHODS AND RESULTS: Patients with cervical or ovarian cancer were administered irinotecan and investigated by medical chart reviews to determine whether oral alkalization drugs were effective at ameliorating irinotecan-induced neutropenia. The drug combination in the oral alkalization drugs-ursodeoxycholic acid, magnesium oxide, and sodium hydrogen carbonate-significantly improved neutrophil counts and reduced dose intensity compared with those of non-users. In the large-scale Japanese Adverse Drug Event Report database, the reporting odds ratio of irinotecan-induced neutropenia was significantly lower when irinotecan had been given in combination with oral alkalization drugs. CONCLUSIONS: These data indicate that oral alkalization drugs may reduce the frequency of neutropenia caused by irinotecan administration, making it possible to increase the dose safely.


Subject(s)
Irinotecan/adverse effects , Neutropenia/chemically induced , Topoisomerase I Inhibitors/adverse effects , Adult , Aged , Antacids/therapeutic use , Antineoplastic Agents, Phytogenic/adverse effects , Buffers , Camptothecin/analogs & derivatives , Cholagogues and Choleretics/therapeutic use , Diarrhea/prevention & control , Female , Humans , Intestines/drug effects , Magnesium Oxide/therapeutic use , Male , Middle Aged , Retrospective Studies , Sodium Bicarbonate/therapeutic use , Ursodeoxycholic Acid/therapeutic use , Uterine Cervical Neoplasms/drug therapy , Uterine Neoplasms/drug therapy
17.
Pharmacology ; 102(5-6): 287-299, 2018.
Article in English | MEDLINE | ID: mdl-30253416

ABSTRACT

BACKGROUND/AIMS: We have reported that nitrosonifedipine (NO-NIF), a photodegradation product of nifedipine, has strong antioxidant and endothelial protective effects, and can suppress several cardiovascular diseases in animal models. The objective of the present study was to investigate the effects of NO-NIF on aortic aneurysm formation. METHODS: The mice were infused with ß-aminopropionitrile for 2 weeks and angiotensin II for 6 weeks to induce aortic aneurysm formation. The oxidative stress was measured by dihydroethidium staining and nitrotyrosine staining. The expressions of inflammation-related genes were assessed by quantitative real-time PCR and immunohistochemical staining. To clarify the mechanisms of how NO-NIF suppresses vascular cell adhesion molecule (VCAM)-1, endothelial cells were used in in vitro system. RESULTS: NO-NIF suppressed pharmacologically induced the aortic aneurysm formation and aortic expansion without blood pressure changes. NO-NIF suppressed elastin degradation and matrix metalloproteinase-2 mRNA expression. NO-NIF suppressed the reactive oxygen species-cyclophilin A positive feedback loop. Upregulated mRNA expressions of inflammation-related genes and endothelial VCAM-1 were suppressed by NO-NIF co-treatment in aortae. CONCLUSION: NO-NIF has the potential to be a new, nifedipine-derived therapeutic drug for suppressing aortic aneurysm formation by directly improving aortic structure with its strong ability to reduce oxidative stress and inflammation.


Subject(s)
Aortic Aneurysm/drug therapy , Nifedipine/analogs & derivatives , Nitroso Compounds/pharmacology , Aminopropionitrile/administration & dosage , Angiotensin II/administration & dosage , Animals , Antigens, Differentiation/metabolism , Antioxidants/pharmacology , Aortic Aneurysm/chemically induced , Aortic Aneurysm/metabolism , Chemokine CCL2/metabolism , Cyclophilins/metabolism , Disease Models, Animal , Elastin/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Male , Matrix Metalloproteinase 2/metabolism , Mice , Nifedipine/pharmacology , Oxidative Stress/drug effects , Photolysis , Reactive Oxygen Species/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
18.
Sci Rep ; 8(1): 10858, 2018 Jul 18.
Article in English | MEDLINE | ID: mdl-30022146

ABSTRACT

Renal tubulointerstitial injury, an inflammation-associated condition, is a major cause of chronic kidney disease (CKD). Levels of activated factor X (FXa), a blood coagulation factor, are increased in various inflammatory diseases. Therefore, we investigated the protective effects of an FXa inhibitor against renal tubulointerstitial injury using unilateral ureteral obstruction (UUO) mice (a renal tubulointerstitial fibrosis model) and the Food and Drug Administration Adverse Events Reporting System (FAERS) database. The renal expression levels of FX and the FXa receptors protease-activated receptor (PAR)-1 and PAR-2 were significantly higher in UUO mice than in sham-operated mice. UUO-induced tubulointerstitial fibrosis and extracellular matrix expression were suppressed in UUO mice treated with the FXa inhibitor edoxaban. Additionally, edoxaban attenuated UUO-induced macrophage infiltration and inflammatory molecule upregulation. In an analysis of the FAERS database, there were significantly fewer reports of tubulointerstitial nephritis for patients treated with FXa inhibitors than for patients not treated with inhibitors. These results suggest that FXa inhibitors exert protective effects against CKD by inhibiting tubulointerstitial fibrosis.


Subject(s)
Databases, Factual , Factor Xa Inhibitors/pharmacology , Kidney Diseases/prevention & control , Macrophages/drug effects , Nephritis, Interstitial/drug therapy , Pyridines/pharmacology , Thiazoles/pharmacology , Ureteral Obstruction/drug therapy , Animals , Cells, Cultured , Humans , Inflammation/pathology , Inflammation/prevention & control , Kidney Diseases/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Nephritis, Interstitial/pathology , Ureteral Obstruction/pathology
19.
Biol Pharm Bull ; 41(4): 555-563, 2018.
Article in English | MEDLINE | ID: mdl-29607928

ABSTRACT

Artemisinin was discovered in 1971 as a constituent of the wormwood genus plant (Artemisia annua). This plant has been used as an herbal medicine to treat malaria since ancient times. The compound artemisinin has a sesquiterpene lactone bearing a peroxide group that offers its biological activity. In addition to anti-malarial activity, artemisinin derivatives have been reported to exert antitumor activity in cancer cells, and have attracted attention as potential anti-cancer drugs. Mechanisms that might explain the antitumor activities of artemisinin derivatives reportedly induction of apoptosis, angiogenesis inhibitory effects, inhibition of hypoxia-inducible factor-1α (HIF-1α) activation, and direct DNA injury. Reactive oxygen species (ROS) generation is involved in many cases. However, little is known about the mechanism of ROS formation from artemisinin derivatives and what types of ROS are produced. Therefore, we investigated the iron-induced ROS formation mechanism by using artesunate, a water-soluble artemisinin derivative, which is thought to be the underlying mechanism involved in artesunate-mediated cell death. The ROS generated by the coexistence of iron(II), artesunate, and molecular oxygen was a hydroxyl radical or hydroxyl radical-like ROS. Artesunate can reduce iron(III) to iron(II), which enables generation of ROS irrespective of the iron valence. We found that reduction from iron(III) to iron(II) was activated in the acidic rather than the neutral region and was proportional to the hydrogen ion concentration.


Subject(s)
Antineoplastic Agents/pharmacology , Artemisinins/pharmacology , Iron/pharmacology , Oxygen/pharmacology , Reactive Oxygen Species/metabolism , Antimalarials/pharmacology , Antipyrine/analogs & derivatives , Antipyrine/pharmacology , Artesunate , Cell Survival/drug effects , Edaravone , Free Radical Scavengers/pharmacology , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Oxidation-Reduction
20.
Nephrol Dial Transplant ; 33(4): 586-597, 2018 04 01.
Article in English | MEDLINE | ID: mdl-28992067

ABSTRACT

Background: Hepcidin secreted by hepatocytes is a key regulator of iron metabolism throughout the body. Hepcidin concentrations are increased in chronic kidney disease (CKD), contributing to abnormalities in iron metabolism. Levels of indoxyl sulfate (IS), a uremic toxin, are also elevated in CKD. However, the effect of IS accumulation on iron metabolism remains unclear. Methods: We used HepG2 cells to determine the mechanism by which IS regulates hepcidin concentrations. We also used a mouse model of adenine-induced CKD. The CKD mice were divided into two groups: one was treated using AST-120 and the other received no treatment. We examined control mice, CKD mice, CKD mice treated using AST-120 and mice treated with IS via drinking water. Results: In the in vitro experiments using HepG2 cells, IS increased hepcidin expression in a dose-dependent manner. Silencing of the aryl hydrocarbon receptor (AhR) inhibited IS-induced hepcidin expression. Furthermore, IS induced oxidative stress and antioxidant drugs diminished IS-induced hepcidin expression. Adenine-induced CKD mice demonstrated an increase in hepcidin concentrations; this increase was reduced by AST-120, an oral adsorbent of the uremic toxin. CKD mice showed renal anemia, decreased plasma iron concentration, increased plasma ferritin and increased iron content in the spleen. Ferroportin was decreased in the duodenum and increased in the spleen. These changes were ameliorated by AST-120 treatment. Mice treated by direct IS administration showed hepatic hepcidin upregulation. Conclusions: IS affects iron metabolism in CKD by participating in hepcidin regulation via pathways that depend on AhR and oxidative stress.


Subject(s)
Gene Expression Regulation/drug effects , Hepcidins/metabolism , Indican/pharmacology , Iron/metabolism , Renal Insufficiency, Chronic/metabolism , Adenine/toxicity , Animals , Disease Models, Animal , Hep G2 Cells , Hepcidins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/pathology
SELECTION OF CITATIONS
SEARCH DETAIL