Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
FASEB J ; 38(2): e23425, 2024 01 31.
Article in English | MEDLINE | ID: mdl-38226852

ABSTRACT

Postprandial hyperglycemia is an early indicator of impaired glucose tolerance that leads to type 2 diabetes mellitus (T2DM). Alterations in the fatty acid composition of phospholipids have been implicated in diseases such as T2DM and nonalcoholic fatty liver disease. Lysophospholipid acyltransferase 10 (LPLAT10, also called LPCAT4 and LPEAT2) plays a role in remodeling fatty acyl chains of phospholipids; however, its relationship with metabolic diseases has not been fully elucidated. LPLAT10 expression is low in the liver, the main organ that regulates metabolism, under normal conditions. Here, we investigated whether overexpression of LPLAT10 in the liver leads to improved glucose metabolism. For overexpression, we generated an LPLAT10-expressing adenovirus (Ad) vector (Ad-LPLAT10) using an improved Ad vector. Postprandial hyperglycemia was suppressed by the induction of glucose-stimulated insulin secretion in Ad-LPLAT10-treated mice compared with that in control Ad vector-treated mice. Hepatic and serum levels of phosphatidylcholine 40:7, containing C18:1 and C22:6, were increased in Ad-LPLAT10-treated mice. Serum from Ad-LPLAT10-treated mice showed increased glucose-stimulated insulin secretion in mouse insulinoma MIN6 cells. These results indicate that changes in hepatic phosphatidylcholine species due to liver-specific LPLAT10 overexpression affect the pancreas and increase glucose-stimulated insulin secretion. Our findings highlight LPLAT10 as a potential novel therapeutic target for T2DM.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase , Diabetes Mellitus, Type 2 , Glucose Intolerance , Animals , Mice , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Glucose/pharmacology , Insulin Secretion , Liver , Phosphatidylcholines , Phospholipids
2.
PLoS One ; 17(9): e0274297, 2022.
Article in English | MEDLINE | ID: mdl-36099304

ABSTRACT

The liver is the main organ that regulates lipid and glucose metabolism. Ectopic lipid accumulation in the liver impairs insulin sensitivity and glucose metabolism. Lipoprotein lipase (LPL), mainly expressed in the adipose tissue and muscle, is a key enzyme that regulates lipid metabolism via the hydrolysis of triglyceride in chylomicrons and very-low-density lipoproteins. Here, we aimed to investigate whether the suppression level of hepatic lipid accumulation via overexpression of LPL in mouse liver leads to improved metabolism. To overexpress LPL in the liver, we generated an LPL-expressing adenovirus (Ad) vector using an improved Ad vector that exhibited considerably lower hepatotoxicity (Ad-LPL). C57BL/6 mice were treated with Ad vectors and simultaneously fed a high-fat diet (HFD). Lipid droplet formation in the liver decreased in Ad-LPL-treated mice relative to that in control Ad vector-treated mice. Glucose tolerance and insulin resistance were remarkably improved in Ad-LPL-treated mice compared to those in control Ad vector-treated mice. The expression levels of fatty acid oxidation-related genes, such as peroxisome proliferator-activated receptor α, carnitine palmitoyltransferase 1, and acyl-CoA oxidase 1, were 1.7-2.0-fold higher in Ad-LPL-treated mouse livers than that in control Ad-vector-treated mouse livers. Furthermore, hepatic LPL overexpression partly maintained mitochondrial content in HFD-fed mice. These results indicate that LPL overexpression in the livers of HFD-fed mice attenuates the accumulation of lipid droplets in the liver and improves glucose metabolism. These findings may enable the development of new drugs to treat metabolic syndromes such as type 2 diabetes mellitus and non-alcoholic fatty liver disease.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Animals , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Glucose/metabolism , Insulin Resistance/physiology , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Triglycerides/metabolism
3.
Exp Clin Endocrinol Diabetes ; 130(4): 254-261, 2022 Apr.
Article in English | MEDLINE | ID: mdl-33782927

ABSTRACT

Genome-wide association studies have identified more than 300 loci associated with type 2 diabetes mellitus; however, the mechanisms underlying their role in type 2 diabetes mellitus susceptibility remain largely unknown. Zinc finger AN1-type domain 3 (ZFAND3), known as testis-expressed sequence 27, is a type 2 diabetes mellitus-susceptibility gene. Limited information is available regarding the physiological role of ZFAND3 in vivo. This study aimed to investigate the association between ZFAND3 and type 2 diabetes mellitus. ZFAND3 was significantly upregulated in the liver of diabetic mice compared to wild-type mice. To overexpress ZFAND3, we generated a ZFAND3-expressing adenovirus (Ad) vector using an improved Ad vector exhibiting significantly lower hepatotoxicity (Ad-ZFAND3). Glucose tolerance was significantly improved in Ad-ZFAND3-treated mice compared to the control Ad-treated mice. ZFAND3 overexpression in the mouse liver also improved insulin resistance. Furthermore, gluconeogenic gene expression was significantly lower in primary mouse hepatocytes transduced with Ad-ZFAND3 than those transduced with the control Ad vector. The present results suggest that ZFAND3 improves glucose tolerance by improving insulin resistance and suppressing gluconeogenesis, serving as a potential novel therapeutic target for type 2 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Insulin Resistance , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Genome-Wide Association Study , Glucose/metabolism , Insulin Resistance/genetics , Liver/metabolism , Male , Mice
4.
J Immunol ; 206(2): 410-421, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33277385

ABSTRACT

Adenovirus (Ad) vector-mediated transduction can cause hepatotoxicity during two phases, at ∼2 and 10 days after administration. Early hepatotoxicity is considered to involve inflammatory cytokines; however, the precise mechanism remains to be clarified. We examined the mechanism of early Ad vector-induced hepatotoxicity by using a conventional Ad vector, Ad-CAL2, and a modified Ad vector, Ad-E4-122aT-CAL2. Ad-E4-122aT-CAL2 harbors sequences complementary to the liver-specific miR-122a in the 3' untranslated region of E4, leading to significant suppression of leaky Ad gene expression in the liver via posttranscriptional gene silencing and a significant reduction in late-phase hepatotoxicity. We found that Ad-E4-122aT-CAL2 transduction significantly attenuated acute hepatotoxicity, although Ad-E4-122aT-CAL2 and Ad-CAL2 induced comparable cytokine expression levels in the liver and spleen. IL-6, a major inflammatory cytokine induced by Ad vectors, significantly enhanced leaky Ad gene expression and cytotoxicity in primary mouse hepatocytes following Ad-CAL2 but not Ad-E4-122aT-CAL2 transduction. Furthermore, leaky Ad gene expression and cytotoxicity in Ad-CAL2-treated hepatocytes in the presence of IL-6 were significantly suppressed upon inhibition of JAK and STAT3. Ad vector-mediated acute hepatotoxicities and leaky Ad expression were significantly reduced in IL-6 knockout mice compared with those in wild-type mice. Thus, Ad vector-induced IL-6 promotes leaky Ad gene expression, leading to acute hepatotoxicity.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/physiology , Genetic Vectors/genetics , Hepatocytes/physiology , Inflammation/immunology , Interleukin-6/metabolism , Liver Diseases/genetics , Animals , Cells, Cultured , Cytokines/metabolism , Gene Expression Regulation , Hepatocytes/virology , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Viral Proteins/genetics , Viral Proteins/metabolism
5.
Int J Food Sci Nutr ; 72(3): 335-347, 2021 May.
Article in English | MEDLINE | ID: mdl-32862731

ABSTRACT

Obesity is associated with an increased risk of metabolic abnormalities. The citrus fruit calamondin contains nobiletin and hesperidin, which are involved in lipid metabolism, and vitamin C, which is an antioxidant. We investigated the metabolic profiles of C57BL/6 mice fed a normal diet, high-fat diet (HFD), HFD + 1% (w/w) calamondin puree (HFD + CL1), or HFD + 5% (w/w) calamondin puree (HFD + CL5). Glucose tolerance was significantly higher in HFD + CL than in HFD-fed mice. Histological analysis revealed less lipid accumulation in the livers of HFD + CL-fed mice than in those of HFD-fed control mice. Hepatocyte ballooning and large lipid droplets - key non-alcoholic fatty liver disease characteristics - were observed in HFD-fed mice after 4 weeks; however, they were nearly absent in HFD + CL-fed mice. The serum expression level of inflammation-associated Ccl2 was lower in HFD + CL-fed mice than in HFD-fed mice. Thus, calamondin may ameliorate HFD-induced metabolic disturbances, including the progression of non-alcoholic fatty liver disease.


Subject(s)
Citrus , Diet, High-Fat/adverse effects , Diet , Dietary Supplements , Non-alcoholic Fatty Liver Disease/metabolism , Adipose Tissue/pathology , Alanine Transaminase/blood , Animals , Antioxidants/metabolism , Ascorbic Acid/metabolism , Body Weight , Chemokine CCL2/blood , Gene Expression , Inflammation , Insulin/blood , Lipid Metabolism , Liver/metabolism , Liver/pathology , Male , Metabolic Syndrome , Mice , Mice, Inbred C57BL , Obesity/metabolism
6.
Biol Pharm Bull ; 42(8): 1295-1302, 2019.
Article in English | MEDLINE | ID: mdl-31366865

ABSTRACT

Obesity is characterized by abnormal or excessive fat accumulation, which leads to the development of metabolic syndrome. Because oxidative stress is increased in obesity, antioxidants are regarded as suitable agents for preventing metabolic syndrome. Here, we examined the impact of cranberry, which contains various antioxidants, on metabolic profiles, including that during the progression of non-alcoholic fatty liver disease (NAFLD), in high-fat diet (HFD)-fed C57BL/6 mice. We observed that oxidative stress was diminished in mice that were fed HFD diets supplemented with 1 and 5% cranberry powder as compared with that in HFD-fed control mice. Notably, from 1 week after beginning the diets to the end of the study, the body weight of mice in the cranberry-treatment groups was significantly lower than that of mice in the HFD-fed control group; during the early treatment phase, cranberry suppressed the elevation of serum triglycerides; and adipocytes in the adipose tissues of cranberry-supplemented-HFD-fed mice were smaller than these cells in HFD-fed control mice. Lastly, we examined the effect of cranberry on NAFLD, which is one of the manifestations of metabolic syndrome in the liver. Histological analysis of the liver revealed that lipid-droplet formation and hepatocyte ballooning, which are key NAFLD characteristics, were both drastically decreased in cranberry-supplemented-HFD-fed mice relative to the levels in HFD-fed control mice. Our results suggest that cranberry ameliorates HFD-induced metabolic disturbances, particularly during the early treatment stage, and exhibits considerable potential for preventing the progression of NAFLD.


Subject(s)
Antioxidants/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Plant Preparations/therapeutic use , Vaccinium macrocarpon , Animals , Antioxidants/pharmacology , Blood Glucose/analysis , Diet, High-Fat , Gene Expression/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/drug effects , Plant Preparations/pharmacology , Powders , Sterol Regulatory Element Binding Protein 1/genetics , Triglycerides/blood
7.
Chem Biol Interact ; 302: 36-45, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30707979

ABSTRACT

Aldose reductase (AR), a member of aldo-keto reductase family, is the rate-limiting enzyme in the polyol pathway, and is known to play a key role in the pathogenesis of diabetic complications. AR also catalyzes the reduction of reactive aldehydes, thereby detoxifying endogenous as well as xenobiotic aldehydes in various tissues. The transcription of the AR gene was previously shown to be augmented by various stimuli including osmotic and oxidative stresses. A highly conserved region composed of an antioxidant response element (ARE), AP-1 site, and tonicity responsive enhancer (TonE) has been identified within the 5'-flanking region of the AR genes of humans, rats, and mice, which we designated as the multiple stress response region (MSRR). We previously showed that the transcription factor Nrf2 activated AR transcription via ARE within MSRR. In the present study, we examined the interactions among Nrf2, c-Jun, and the TonE-binding protein (TonEBP) in the regulation of AR gene transcription. In gene reporter assays using luciferase reporter constructs containing the MSRR of the mouse AR (AKR1B3) gene with HepG2 cells, the forced expression of Nrf2 or TonEBP significantly increased promoter activity. The synergistic augmentation of promoter activity was observed when Nrf2 and TonEBP were co-introduced. On the other hand, the co-expression of c-Jun repressed promoter activation by Nrf2 and TonEBP. The mutation of the AP-1 site within MSRR did not affect the repressive effects of c-Jun, while the introduction of truncated c-Jun proteins lacking the leucine zipper domain no longer suppressed Nrf2-or TonEBP-mediated transactivation, suggesting that c-Jun repressed promoter activity independently of the AP-1 site and that interactions with protein factors via the leucine zipper domain were necessary for its negative effects on Nrf2 and TonEBP. These results indicate that AR promoter activity is cooperatively regulated by multiple transcription factors via MSRR.


Subject(s)
Aldehyde Reductase/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/metabolism , NFATC Transcription Factors/metabolism , Aldehyde Reductase/genetics , Animals , Genes, Reporter , Hep G2 Cells , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Mutagenesis, Site-Directed , NF-E2-Related Factor 2/genetics , NFATC Transcription Factors/genetics , Phosphorylation , Plasmids/genetics , Plasmids/metabolism , Promoter Regions, Genetic , Transcription, Genetic
8.
J Pharmacol Sci ; 139(3): 137-142, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30665845

ABSTRACT

Ischemia/reperfusion injury is the most common cause of acute kidney injury. We previously revealed that pre-treatment with yohimbine or JP-1302 attenuated renal ischemia/reperfusion injury by inhibition of α2C-adrenoceptor antagonist. The aim of the present study is to investigate the effects of post-treatment with JP-1302 on renal ischemia/reperfusion injury in rats. Male Sprague Dawley rats were randomly divided into four groups: sham operation, ischemia/reperfusion, pre-treatment with JP-1302 (3.0 mg/kg) and post-treatment with JP-1302 groups. In ischemia/reperfusion injury, renal functional parameters, such as blood urea nitrogen, plasma creatinine and creatinine clearance, deteriorated after reperfusion. Renal venous norepinephrine concentrations, as well as inflammatory molecules in the kidney increased after reperfusion. Both pre- and post-treatment with JP-1302 improved renal dysfunction, tissue damage, renal venous norepinephrine concentrations and inflammatory molecules expression in the kidney. In conclusion, these results suggest that post-treatment with JP-1302 protects on ischemia/reperfusion-induced acute kidney injury by suppressing cytokine upregulation via α2C-adrenoceptors.


Subject(s)
Acridines/pharmacology , Acute Kidney Injury/prevention & control , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Piperazines/pharmacology , Reperfusion Injury/drug therapy , Acridines/administration & dosage , Adrenergic alpha-2 Receptor Antagonists/administration & dosage , Animals , Blood Urea Nitrogen , Creatinine/blood , Cytokines/metabolism , Drug Administration Schedule , Male , Piperazines/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Reperfusion Injury/complications , Up-Regulation/drug effects
9.
Yakugaku Zasshi ; 139(1): 47-51, 2019.
Article in Japanese | MEDLINE | ID: mdl-30606928

ABSTRACT

Although many treatments for type 2 diabetes mellitus (T2DM) have been developed, the quality of life for people with T2DM still tends to be lower than in those without the disease. Thus, the development of new T2DM treatments and prevention methods is required. Genetic predisposition and environmental factors are understood to be involved in the onset and pathology of T2DM. Therefore, we have attempted to explore genes and foods with potential for use in the treatment and prevention of T2DM. LipoQuality, which describes the functional features of diverse lipid species, has recently been a focus of study in the pathology of metabolic diseases. Phospholipids, the major components of biological membranes, are known to change in composition during the development of obesity and diabetes. Therefore, for our research, we focused on genes that regulate the composition of phospholipids. We examined the effects of such genes on T2DM using an improved adenovirus vector that demonstrates safer, higher, and longer-term transgene expression than that of the conventional adenovirus vector. We also found that certain foods inhibit the progression of non-alcoholic fatty liver disease, which is related to T2DM. In this review, we introduce our research results, demonstrating how genes and food independently contribute to the mechanisms of T2DM pathology.


Subject(s)
Adenoviridae , Diabetes Mellitus, Type 2/prevention & control , Diabetes Mellitus, Type 2/therapy , Functional Food , Genetic Vectors , Phospholipids , Vaccinium macrocarpon , 1-Acylglycerophosphocholine O-Acyltransferase/physiology , Animals , Diabetes Mellitus, Type 2/etiology , Gene-Environment Interaction , Humans , Liver/metabolism , Mice , Non-alcoholic Fatty Liver Disease/prevention & control , Oxidative Stress , Phospholipids/metabolism
10.
Eur J Pharmacol ; 838: 113-119, 2018 Nov 05.
Article in English | MEDLINE | ID: mdl-30201375

ABSTRACT

Nephrotoxicity is a major adverse reaction of the anticancer drug, cisplatin. We investigated the renoprotective effects of the α2-adrenoceptor antagonist, yohimbine and selective α2C-adrenoceptor antagonist, JP-1302, in cisplatin-treated Sprague Dawley rats. Rats were given a single intravenous dose of 7.5 mg/kg cisplatin and then yohimbine or JP-1302 was administered intraperitoneally at 0.1 or 3 mg/kg/day, respectively, for four days. Renal functional parameters, such as blood urea nitrogen, plasma creatinine, creatinine clearance and renal venous norepinephrine concentrations were measured. Kidney tissue damage and tumour necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) mRNA levels were assessed after the animals were euthanized. Cisplatin treatment aggravated the kidney functional parameters of blood urea nitrogen, plasma creatinine and creatinine clearance. Renal venous norepinephrine concentrations were also elevated after cisplatin administration. Treatment with yohimbine or JP-1302 clearly ameliorated kidney function and cell apoptosis. These treatments suppressed elevated renal plasma norepinephrine, TNF-α, MCP-1 and cleaved caspase 3 expressions which occurred after administration of cisplatin. These results suggest that yohimbine can prevent cisplatin-induced renal toxicity associated with acute kidney injury by suppressing cytokine expression through α2C-adrenoceptors.


Subject(s)
Acridines/pharmacology , Acute Kidney Injury/drug therapy , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Cisplatin/adverse effects , Piperazines/pharmacology , Yohimbine/pharmacology , Acridines/therapeutic use , Acute Kidney Injury/blood , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Animals , Apoptosis/drug effects , Chemokine CCL2/metabolism , Disease Models, Animal , Injections, Intraperitoneal , Kidney/drug effects , Kidney/pathology , Male , Norepinephrine/blood , Piperazines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Yohimbine/therapeutic use
11.
Eur J Pharmacol ; 818: 38-42, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29032106

ABSTRACT

Increases in renal sympathetic nerve activity during ischaemia and renal venous norepinephrine levels after reperfusion play important roles in the development of ischaemia/reperfusion-induced acute kidney injury. In the present study, we examined the effect of isatin, an endogenous monoamine oxidase inhibitor, on renal venous norepinephrine levels, superoxide production after reperfusion, and ischaemia/reperfusion-induced acute kidney injury. Ischaemia/reperfusion-induced acute kidney injury was accomplished by clamping the left renal artery and vein for 45min, followed by reperfusion, 2 weeks after contralateral nephrectomy. Renal superoxide production and norepinephrine overflow were elevated and significant renal tissue damage was observed following ischaemia/reperfusion injury. Intravenous injection of isatin (10mg/kg) at 5min before ischaemia increased the renal venous plasma norepinephrine level after reperfusion and aggravated ischaemia/reperfusion-induced renal dysfunction and histological damage. The excessive superoxide production after reperfusion was significantly suppressed by isatin administration, indicating that the inhibition of oxidative deamination effectively suppressed superoxide production. These data suggest that the exacerbation effect of isatin is associated, at least in part, with increased norepinephrine levels but not with superoxide production. To the best of our knowledge, this is the first report of isatin involvement in the pathogenesis and/or development of acute kidney injury.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Monoamine Oxidase Inhibitors/pharmacology , Reperfusion Injury/complications , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Monoamine Oxidase Inhibitors/therapeutic use , Norepinephrine/blood , Rats , Rats, Sprague-Dawley , Superoxides/metabolism
12.
Biochem Biophys Rep ; 10: 192-197, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28955747

ABSTRACT

Japanese patients with type 2 diabetes mellitus present a different responsiveness in terms of insulin secretion to glucose and body mass index (BMI) from other populations. The genetic background that predisposes Japanese individuals to type 2 diabetes mellitus is under study. Recent genetic studies demonstrated that the locus mapped in macrophage erythroblast attacher (MAEA) increases the susceptibility to type 2 diabetes mellitus in East Asians, including Japanese individuals. MAEA encodes a protein that plays a role in erythroblast enucleation and in the normal differentiation of erythroid cells and macrophages. However, the contribution of MAEA to type 2 diabetes mellitus remains unknown. In this study, to overexpress MAEA in the mouse liver and primary mouse hepatocytes, we generated a MAEA-expressing adenovirus (Ad) vector using a novel Ad vector exhibiting significantly lower hepatotoxicity (Ad-MAEA). Blood glucose and insulin levels in Ad-MAEA-treated mice were comparable to those in control Ad-treated mice. Primary mouse hepatocytes transduced with Ad-MAEA showed lower levels of expression of gluconeogenesis genes than those transduced with the control Ad vector. Hepatocyte nuclear factor-4α (HNF-4α) mRNA expression in primary mouse hepatocytes was also suppressed by MAEA overexpression. These results suggest that MAEA overexpression attenuates hepatic gluconeogenesis, which could potentially lead to improvement of type 2 diabetes mellitus.

13.
Biochem Pharmacol ; 138: 185-192, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28450226

ABSTRACT

Human carbonyl reductase 1 (CBR1), a member of the short-chain dehydrogenase/reductase (SDR) superfamily, reduces a variety of carbonyl compounds including endogenous isatin, prostaglandin E2 and 4-oxo-2-nonenal. It is also a major non-cytochrome P450 enzyme in the phase I metabolism of carbonyl-containing drugs, and is highly expressed in the intestine. In this study, we found that long-chain fatty acids and their CoA ester derivatives inhibit CBR1. Among saturated fatty acids, myristic, palmitic and stearic acids were inhibitory, and stearic acid was the most potent (IC50 9µM). Unsaturated fatty acids (oleic, elaidic, γ-linolenic and docosahexaenoic acids) and acyl-CoAs (palmitoyl-, stearoyl- and oleoyl-CoAs) were more potent inhibitors (IC50 1.0-2.5µM), and showed high inhibitory selectivity to CBR1 over its isozyme CBR3 and other SDR superfamily enzymes (DCXR and DHRS4) with CBR activity. The inhibition by these fatty acids and acyl-CoAs was competitive with respect to the substrate, showing the Ki values of 0.49-1.2µM. Site-directed mutagenesis of the substrate-binding residues of CBR1 suggested that the interactions between the fatty acyl chain and the enzyme's Met141 and Trp229 are important for the inhibitory selectivity. We also examined CBR1 inhibition by oleic acid in cellular levels: The fatty acid effectively inhibited CBR1-mediated 4-oxo-2-nonenal metabolism in colon cancer DLD1 cells and increased sensitivity to doxorubicin in the drug-resistant gastric cancer MKN45 cells that highly express CBR1. The results suggest a possible new food-drug interaction through inhibition of CBR1-mediated intestinal first-pass drug metabolism by dietary fatty acids.


Subject(s)
Acyl Coenzyme A/metabolism , Alcohol Oxidoreductases/antagonists & inhibitors , Fatty Acids, Nonesterified/metabolism , Intestinal Mucosa/enzymology , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Binding Sites , Binding, Competitive , Cell Line, Tumor , Drug Resistance, Neoplasm , Food-Drug Interactions , Humans , Mutation , Myristic Acid/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/genetics , Oxidoreductases/metabolism , Palmitic Acid/metabolism , Palmitoyl Coenzyme A/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Stearic Acids/metabolism , Sugar Alcohol Dehydrogenases/antagonists & inhibitors , Sugar Alcohol Dehydrogenases/genetics , Sugar Alcohol Dehydrogenases/metabolism
14.
Chem Biol Interact ; 276: 40-45, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28342890

ABSTRACT

The functional genetic polymorphism of aldehyde dehydrogenase 2 (ALDH2) influences the enzymatic activities of its wild type (Glu504 encoded by ALDH2*1) and mutant type (Lys504 encoded by ALDH2*2) proteins. The enzymatic activities of mutant-type ALDH2 are limited compared with those of the wild type. ALDH2 has been suggested as a critical factor for nitroglycerin-mediated vasodilation by some human studies and in vitro studies. Currently, there is no research on direct observations of the vasodilatory effect of nitroglycerin sublingual tablets, which is the generally used dosage form. In the present study, the contribution of ALDH2 to the vasodilatory effect of nitroglycerin sublingual tablets was investigated among three genotype groups (ALDH2*1/*1, ALDH2*1/*2, and ALDH2*2/*2) in Japanese. The results by direct assessments of in vivo nitroglycerin-mediated dilation showed no apparent difference in vasodilation among all genotypes of ALDH2. Furthermore, to analyze the effect of other factors (age and flow-mediated dilation), multiple regression analysis and Pearson's correlation coefficient analysis were carried out. These analyses also indicated that the genotypes of ALDH2 were not related to the degree of vasodilation. These results suggest the existence of other predominant pathway(s) for nitroglycerin biotransformation, at least with regard to clinical nitroglycerin (e.g., a sublingual tablet) in Japanese subjects.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/genetics , Asian People/genetics , Nitroglycerin/pharmacology , Vasodilator Agents/pharmacology , Adult , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Biotransformation , Brachial Artery/diagnostic imaging , Female , Genotype , Heterozygote , Homozygote , Humans , Japan , Lung/blood supply , Lung/drug effects , Male , Middle Aged , Nitroglycerin/chemistry , Phenotype , Polymorphism, Genetic , Ultrasonography , Vasodilation/drug effects , Vasodilator Agents/chemistry
15.
Drug Metab Lett ; 11(1): 48-52, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28332451

ABSTRACT

BACKGROUND AND OBJECTIVE: Doxorubicin, an anthracycline anti-cancer drug, is effective for breast cancer and childhood lymphoma. Chronic cardiotoxicity has been known as a critical adverse effect of doxorubicin and is attributed to its metabolite doxorubicinol produced by carbonyl reductase 1 (CBR1, SDR21C1). Some flavonoids have been reported to act as inhibitors for CBR1, therefore, commercially available juices containing flavonoids are likely to be applicable as a prophylactic treatment against doxorubicin-induced cardiotoxicity by suppression of doxorubicinol production. In the study, fruit juices containing flavonoids were investigated for inhibitory effects on CBR1. METHOD: Recombinant CBR1 protein was subjected to in vitro enzymatic assays with/without juices. An apple juice and a grapefruit juice were selected in the present study as candidates capable of inhib-iting CBR1. RESULTS: The enzymatic assays revealed that both juices potently inhibit the CBR1-mediated metabolic conversion of doxorubicin to doxorubicinol in concertation-dependent manner. The concentrations required for obtaining 50% inhibition (IC50) were 0.0012% (v/v) and 0.0014% (v/v) for apple and grapefruit juices, respectively. Additionally, it is worth noting that these juices showed inhibitory effects on doxorubicin metabolism by CBR1 even at very low concentrations (0.0001% (v/v)). CONCLUSION: An apple juice and a grape fruit juice showed strong inhibitory effects on doxorubicin metabolism by CBR1 in vitro. These results suggest that the intake of flavonoid-containing juices can be a promising measure for protection against doxorubicin-induced cardiac toxicity, enabling patients to keep higher adherence with routine use in light of safety, economic performance and stable supply to market.


Subject(s)
Alcohol Oxidoreductases/chemistry , Antineoplastic Agents/chemistry , Citrus paradisi/chemistry , Doxorubicin/chemistry , Fruit and Vegetable Juices , Malus/chemistry , Doxorubicin/analogs & derivatives , Flavonoids/chemistry , Humans , Recombinant Proteins/chemistry
16.
Chem Biol Interact ; 276: 160-166, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28219640

ABSTRACT

AKR1B10 is a human-type aldo-keto reductase. The up-regulation of AKR1B10 has been associated with various cancers including non-small cell lung carcinoma, viral and bacterial infections, and skin diseases. However, the mechanisms underlying AKR1B10 gene regulation are not fully understood. We previously indicated the involvement of the transcription factor Nrf2 in AKR1B10 gene regulation. There are at least five potential Nrf2-responsive consensus sequences, so-called antioxidant response elements (AREs), and several ARE-like sequences in the 5'-flanking region up to -3282 bp of the AKR1B10 gene. In the present study, we attempted to identify functional AREs by luciferase reporter analyses using various mutants for each ARE. And we found that only those between -530 and -520 bp (ARE-A), which is the closest location to the translation start site, were functional among the five ARE consensus sites examined. Furthermore, ARE-A functioned co-operatively with the neighboring AP-1 site. Since the AP-1 site resembles ARE, the tandem arrangement of these two elements may be essential for augmented responsiveness to Nrf2 and plays an important role in AKR1B10 gene regulation by various Nrf2-mediating stimuli.


Subject(s)
Aldehyde Reductase/genetics , Antioxidant Response Elements/genetics , A549 Cells , Aldehyde Reductase/chemistry , Aldehyde Reductase/metabolism , Aldo-Keto Reductases , Base Sequence , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Genes, Reporter , Humans , NF-E2-Related Factor 2/genetics , Promoter Regions, Genetic
17.
Eur J Pharmacol ; 781: 36-44, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-27041645

ABSTRACT

Excitation of renal sympathetic nervous activity and the resulting increased levels of renal venous norepinephrine play important roles in renal ischaemia/reperfusion injury in rats. This study examined the effects of yohimbine, a non-selective α2-adrenoceptor antagonist, on renal venous norepinephrine levels and kidney function in acute kidney injury. Acute ischaemia/reperfusion-induced kidney injury was induced in rats by clamping the left renal artery and vein for 45min, followed by reperfusion, 2 weeks after a contralateral nephrectomy. Intravenous injection of yohimbine (0.1mg/kg) 5min prior to ischaemia significantly attenuated kidney injury and decreased the renal venous norepinephrine levels, as compared with vehicle-treated rats. To investigate the involvement of α2-adrenoceptor subtypes, we pre-treated with JP-1302, a selective α2C-adrenoceptor antagonist (1mg/kg). This suppressed renal venous norepinephrine levels and tumour necrosis factor-α and monocyte chemoattractant protein-1 mRNA levels after reperfusion and improved kidney function. Pre-treatment with BRL44408, a selective α2A-adrenoceptor antagonist (1mg/kg), or imiloxan, a selective α2B-adrenoceptor antagonist (1mg/kg) had no effect on renal function or tissue injury. These results suggest that yohimbine prevented ischaemia/reperfusion-induced kidney injury by inhibiting α2C-adrenoceptors and suppressing pro-inflammatory cytokine expression.


Subject(s)
Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Cytoprotection/drug effects , Kidney/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Reperfusion Injury/complications , Yohimbine/pharmacology , Acridines/pharmacology , Acute Kidney Injury/blood , Acute Kidney Injury/etiology , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Chemokine CCL2/metabolism , Gene Expression Regulation/drug effects , Kidney/metabolism , Kidney/pathology , Male , Norepinephrine/metabolism , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
18.
Biol Pharm Bull ; 38(9): 1309-19, 2015.
Article in English | MEDLINE | ID: mdl-26328486

ABSTRACT

Doxorubicin (DOX) is widely used for the treatment of a wide range of cancers such as breast and lung cancers, and malignant lymphomas, but is generally less efficacious in gastrointestinal cancers. The most accepted explanation for the DOX refractoriness is its resistance development. Here, we established DOX-resistant phenotypes of human gastric MKN45 and colon LoVo cells by continuous exposure to incremental concentrations of the drug. While the parental MKN45 and LoVo cells expressed carbonyl reductase 1 (CBR1) highly and moderately, respectively, the gain of DOX resistance further elevated the CBR1 expression. Additionally, the DOX-elicited cytotoxicity was lowered by overexpression of CBR1 and inversely strengthened by knockdown of the enzyme using small interfering RNA or pretreating with the specific inhibitor quercetin, which also reduced the DOX refractoriness of the two resistant cells. These suggest that CBR1 is a key enzyme responsible for the DOX resistance of gastrointestinal cancer cells and that its inhibitor is useful in the adjuvant therapy. Although CBR1 is known to metabolize DOX to a less toxic anticancer metabolite doxorubicinol, its overexpression in the parental cells hardly show significant reductase activity toward low concentration of DOX. In contrast, the overexpression of CBR1 increased the reductase activity toward an oxidative stress-derived cytotoxic aldehyde 4-oxo-2-nonenal. The sensitivity of the DOX-resistant cells to 4-oxo-2-nonenal was lower than that of the parental cells, and the resistance-elicited hyposensitivity was almost completely ameliorated by addition of the CBR1 inhibitor. Thus, CBR1 may promote development of DOX resistance through detoxification of cytotoxic aldehydes, rather than the drug's metabolism.


Subject(s)
Alcohol Oxidoreductases/biosynthesis , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/physiology , Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Cell Line, Tumor , Gastrointestinal Neoplasms/metabolism , Humans , NF-E2-Related Factor 2/metabolism , Quercetin/pharmacology , Up-Regulation
19.
Org Biomol Chem ; 13(27): 7487-99, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26068795

ABSTRACT

Human carbonyl reductase 1 (CBR1), a member of the short-chain dehydrogenase/reductase superfamily, reduces anthracycline anticancer drugs to their less potent anticancer C-13 hydroxy metabolites, which are linked with pathogenesis of cardiotoxicity, a side effect of the drugs. CBR1 inhibitors are thought to be promising agents for adjuvant therapy with a twofold beneficial effect in prolonging the anticancer efficacy of the anthracyclines while decreasing cardiotoxicity. In order to search for new potential inhibitors of CBR1, we synthesized a series of des-methoxyphenyl derivatives of (Z)-2-(4-methoxyphenylimino)-7-hydroxy-N-(pyridin-2-yl)-2H-chromene-3-carboxamide (1) that was developed previously as a potent inhibitor of aldo-keto reductase (AKR) 1B10 and AKR1B1. Among the newly synthesized inhibitors, 8-hydroxy-2-imino-2H-chromene-3-carboxylic acid (2-chlorophenyl)amide (13h) was the most potent competitive inhibitor of CBR1, showing a Ki value of 15 nM. 13h also showed high selectivity to CBR1 over its isozyme CBR3 and other enzymes with CBR activity (AKR1B1, AKR1B10, AKR1C1, AKR1C2, AKR1C4, DXCR and DHRS4). Furthermore, 13h inhibited the cellular metabolism by CBR1 at its concentration of 4 µM. The structure-activity relationship of the derivatives, site-directed mutagenesis of putative binding residues (Met141 and Trp229) and molecular docking of 13h in CBR1 revealed that the interactions of 13h with the substrate-binding residues (Ser139, Met141, Tyr193 and Trp229) are important for the tight binding.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Benzopyrans/chemical synthesis , Benzopyrans/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Imines/chemical synthesis , Imines/pharmacology , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Aorta/cytology , Cattle , Cell Survival/drug effects , Coumarins/chemistry , Coumarins/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Inhibitors/chemistry , Humans , Molecular Dynamics Simulation , Mutation/genetics
20.
Cell Biol Int ; 39(7): 808-15, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25677373

ABSTRACT

CHCR3, a member of the short-chain dehydrogenase/reductase superfamily, is a carbonyl reductase 3 enzyme in Chinese hamsters. Carbonyl reductase 3 in humans has been believed to involve the metabolism and/or pharmacokinetics of anthracycline drugs, and the mechanism underlying the gene regulation has been investigated. In this study, the nucleotide sequence of the Chcr3 promoter was originally determined, and its promoter activity was characterised. The proximal promoter region is TATA-less and GC-rich, similar to the promoter region of human carbonyl reductase 3. Cobalt stimulated the transcriptional activity of the Chcr3 gene. The results of a luciferase gene reporter assay demonstrated that cobalt-induced stimulation required an antioxidant responsive element. Forced expression of Nrf2, the transcription factor that binds to antioxidant responsive elements, enhanced the transcriptional activity of the Chcr3 gene. These results suggest that cobalt induces the expression of the Chcr3 gene via the Nrf2-antioxidant responsive element pathway.


Subject(s)
Alcohol Oxidoreductases/genetics , Antioxidant Response Elements , Cricetulus/genetics , Promoter Regions, Genetic , Animals , Base Sequence , Cobalt/metabolism , Cricetinae , HEK293 Cells , Humans , Molecular Sequence Data , NF-E2-Related Factor 2/genetics , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...