Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Exp Biol Med (Maywood) ; 248(13): 1112-1123, 2023 07.
Article in English | MEDLINE | ID: mdl-37646078

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is a progressive form of non-alcoholic fatty liver disease (NAFLD) that causes cirrhosis and hepatocellular carcinoma. Iron is an essential trace element in the body; however, excess iron can cause tissue damage and dysfunction. Iron overload is often observed in patients with NASH, and the amount of iron accumulated in the liver positively correlates with the histological severity of NASH. Ferroptosis, a novel form of iron-dependent cell death, is caused by the accumulation of lipid peroxidation and oxidative stress and is related to NASH. In addition, ferroptosis is closely related to autophagy, an intracellular self-degradation process. Although autophagy has many beneficial effects, it may also be harmful to the organism, for example, inducing ferroptosis. It is unclear whether iron overload aggravates NASH via autophagy. The aim of this research is to determine the mechanism by which iron overload induces ferroptosis via autophagy and aggravates NASH. Stroke-prone spontaneously hypertensive rats (SHRSP5/Dmcr) were divided into two groups and fed a high-fat and high-cholesterol (HFC) diet for eight weeks. Iron dextran was administered to the Fe group in addition to the HFC diet. Blood analysis, histological staining, calcineurin activity assay, quantitative reverse transcription polymerase chain reaction (RT-PCR), immunofluorescence staining, and electron microscopy were performed. The results showed that iron overload promoted autophagy via nuclear translocation of transcription factor EB (TFEB) and induced ferritinophagy, which is the autophagic degradation of ferritin. In addition, the HFC diet induced lipophagy, the autophagic degradation of lipid droplets. The Fe group also exhibited promoted ferroptosis and aggravated hepatic inflammation and fibrosis. In conclusion, iron overload accelerates ferritinophagy and lipophagy, aggravating NASH pathology via ferroptosis. These findings indicate the therapeutic potential of inhibiting autophagy and ferroptosis for treating NASH.


Subject(s)
Ferroptosis , Iron Overload , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Rats , Animals , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/pathology , Rats, Inbred SHR , Iron Overload/complications , Fibrosis , Iron , Autophagy , Liver Neoplasms/complications
2.
J Appl Biomed ; 21(2): 80-90, 2023 06.
Article in English | MEDLINE | ID: mdl-37376883

ABSTRACT

BACKGROUND: Xanthine oxidase (XO) generates reactive oxygen species during uric acid production. Therefore, XO inhibitors, which suppress oxidative stress, may effectively treat non-alcoholic steatohepatitis (NASH) and atherosclerosis via uric acid reduction. In this study, we examined the antioxidant effect of the XO inhibitor febuxostat on NASH and atherosclerosis in stroke-prone spontaneously hypertensive 5 (SHRSP5/Dmcr) rats. METHODS: SHRSP5/Dmcr rats were divided into three groups: SHRSP5/Dmcr + high-fat and high-cholesterol (HFC) diet [control group, n = 5], SHRSP5/Dmcr + HFC diet + 10% fructose (40 ml/day) [fructose group, n = 5], and SHRSP5/Dmcr + HFC diet + 10% fructose (40 ml/day) + febuxostat (1.0 mg/kg/day) [febuxostat group, n = 5]. Glucose and insulin resistance, blood biochemistry, histopathological staining, endothelial function, and oxidative stress markers were evaluated. RESULTS: Febuxostat reduced the plasma uric acid levels. Oxidative stress-related genes were downregulated, whereas antioxidant factor-related genes were upregulated in the febuxostat group compared with those in the fructose group. Febuxostat also ameliorated inflammation, fibrosis, and lipid accumulation in the liver. Mesenteric lipid deposition decreased in the arteries, and aortic endothelial function improved in the febuxostat group. CONCLUSIONS: Overall, the XO inhibitor febuxostat exerted protective effects against NASH and atherosclerosis in SHRSP5/Dmcr rats.


Subject(s)
Atherosclerosis , Non-alcoholic Fatty Liver Disease , Rats , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Antioxidants/pharmacology , Antioxidants/therapeutic use , Febuxostat/pharmacology , Febuxostat/therapeutic use , Xanthine Oxidase , Uric Acid , Rats, Inbred SHR , Diet, High-Fat , Enzyme Inhibitors , Atherosclerosis/drug therapy , Lipids
3.
Ann Anat ; 249: 152104, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37209870

ABSTRACT

BACKGROUND: Secondary sarcopenia develops as a result of a bedridden state and illnesses, such as cachexia, liver disease, and diabetes. However, there is a lack of animal models to investigate the underlying mechanisms and potential treatments for secondary sarcopenia. Recently, secondary sarcopenia has been associated with the prognosis of nonalcoholic steatohepatitis. This study aimed to investigate whether stroke-prone spontaneously hypertensive rat 5 (SHRSP5/Dmcr) which developed severe nonalcoholic steatohepatitis by a high-fat and high-cholesterol (HFC; containing 2% cholic acid) diet is a useful model of secondary sarcopenia. METHODS: SHRSP5/Dmcr rats were divided into 6 groups fed with a Stroke-Prone (SP: normal chow) or HFC diets for different periods (4, 12, and 20 weeks), and WKY/Izm rats were divided into 2 groups fed an SP or HFC diet. Body weight, food intake, and muscle force were measured weekly for all rats. After the end of the diet period, skeletal muscle strength evoked by electrical stimulation was recorded, blood was collected, and organ weight was measured. The sera were used for biochemical analysis and the organs were used for histopathological analysis. RESULTS: SHRSP5/Dmcr rats fed an HFC diet developed nonalcoholic steatohepatitis, and their skeletal muscles, especially fast muscles, showed atrophy, indicating that muscle atrophy is aggravated by the progression of nonalcoholic steatohepatitis. In contrast, WKY/Izm rats fed an HFC diet did not exhibit sarcopenia. CONCLUSIONS: This study suggests that SHRSP5/Dmcr rats could be a useful novel model for investigate the mechanism of secondary sarcopenia disorder associated with nonalcoholic steatohepatitis.


Subject(s)
Non-alcoholic Fatty Liver Disease , Sarcopenia , Rats , Animals , Non-alcoholic Fatty Liver Disease/etiology , Sarcopenia/complications , Sarcopenia/pathology , Rats, Inbred WKY , Diet, High-Fat/adverse effects , Rats, Inbred SHR , Cholesterol , Liver/pathology , Disease Models, Animal
4.
Acta Med Okayama ; 77(1): 29-36, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36849143

ABSTRACT

The SHRSP5/Dmcr is a useful animal model for the development of nonalcoholic steatohepatitis (NASH) pathology when fed a high-fat, high-cholesterol diet, and further drug interventions can lead to concomitant cardiovascular disease. While SHRSP5/Dmcr rats have been used for basic research related to NASH, details of their bile acid metabolism in this condition are unknown. In this study, we aimed to clarify the changes in the serum bile acid (BA) fractions associated with NASH and found that glycine-conjugated and unconjugated bile acid increased with worsening NASH and cardiovascular disease while taurine-conjugated BA relatively decreased.


Subject(s)
Cardiovascular Diseases , Non-alcoholic Fatty Liver Disease , Animals , Rats , Cardiovascular Diseases/etiology , Bile Acids and Salts , Glycine , Taurine
5.
Pharmacol Rep ; 74(4): 669-683, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35819592

ABSTRACT

BACKGROUND: Non-alcoholic steatohepatitis (NASH) is a progressive subtype of non-alcoholic fatty liver disease (NAFLD) that is closely related to cardiovascular disease (CVD). Nitric oxide (NO) plays a critical role in the control of various biological processes. Dysfunction of the NO signaling pathway is associated with various diseases such as atherosclerosis, vascular inflammatory disease, and diabetes. Recently, it has been reported that NO is related to lipid and cholesterol metabolism. Chronic NO synthase (NOS) inhibition accelerates NAFLD by increasing hepatic lipid deposition. However, the detailed relationship between NO and abnormal lipid and cholesterol metabolism in NAFLD/NASH has not been completely explained. We aimed to determine the effects of NOS inhibition by N omega-nitro-L-arginine methyl ester hydrochloride (L-NAME), a NOS inhibitor, on NASH and CVD via lipid and cholesterol metabolism. METHODS: Stroke-prone spontaneously hypertensive rats were fed a high-fat and high-cholesterol diet for 8 weeks and administered L-NAME for the last 2 weeks. Following blood and tissue sampling, biochemical analysis, histopathological staining, quantitative RT-PCR analysis, and western blotting were performed. RESULTS: L-NAME markedly increased hepatic triglyceride (TG) and cholesterol levels by promoting TG synthesis and cholesterol absorption from the diet. L-NAME increased the mRNA levels of inflammatory markers and fibrotic areas in the liver. Cholesterol secretion from the liver was promoted in rats administered L-NAME, which increased serum cholesterol. L-NAME significantly increased the level of oxidative stress marker and lipid deposition in the arteries. CONCLUSIONS: NOS inhibition simultaneously aggravates NASH and atherosclerosis via hepatic lipid and cholesterol metabolism.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Non-alcoholic Fatty Liver Disease , Acceleration , Animals , Atherosclerosis/metabolism , Biomarkers , Cardiovascular Diseases/complications , Cholesterol , Diet, High-Fat , Lipid Metabolism , Liver , NG-Nitroarginine Methyl Ester/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Non-alcoholic Fatty Liver Disease/complications , Rats , Rats, Inbred SHR
6.
Int J Mol Sci ; 23(6)2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35328352

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a chronic liver disease that leads to liver cirrhosis and hepatocellular carcinoma. Endothelial dysfunction caused by hepatic lipotoxicity is an underlying NASH pathology observed in the liver and the cardiovascular system. Here, we evaluated the effect of dietary nitrite on a rat NASH model. Stroke-prone, spontaneously hypertensive 5/Dmcr rats were fed a high-fat/high-cholesterol diet to develop the NASH model, with nitrite or captopril (100 mg/L, each) supplementation in drinking water for 8 weeks. The effects of nitrite and captopril were evaluated using immunohistochemical analyses of the liver and heart tissues. Dietary nitrite suppressed liver fibrosis in the rats by reducing oxidative stress, as measured using the protein levels of nicotinamide adenine dinucleotide phosphate oxidase components and inflammatory cell accumulation in the liver. Nitrite lowered the blood pressure in hypertensive NASH rats and suppressed left ventricular chamber enlargement. Similar therapeutic effects were observed in a captopril-treated rat NASH model, suggesting the possibility of a common signaling pathway through which nitrite and captopril improve NASH pathology. In conclusion, dietary nitrite attenuates the development of NASH with cardiovascular involvement in rats and provides an alternative NASH therapeutic strategy.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Captopril/pharmacology , Cholesterol , Diet, High-Fat/adverse effects , Disease Models, Animal , Liver/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/etiology , Nitrites , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Rats , Rats, Inbred SHR
7.
Environ Pollut ; 298: 118799, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35007670

ABSTRACT

Increased levels of dialkylphosphates (DAP) in maternal urine are associated with a variety of adverse developmental outcomes in children. Although urinary DAP levels are usually considered to be a marker of exposure to organophosphate (OP) pesticides, excretion of DAP may also increase by ingesting preformed DAP. To date, no study has quantitatively assessed the possible contribution of the dietary intake of preformed DAP and OP pesticides to urinary levels of DAP. Therefore, we aimed to estimate the levels of 6 DAPs and 84 OP pesticides in duplicate diet samples and urine samples collected from 73 women living in urban areas of Japan in 2018. DAP and OP pesticides were detected in 94% and 45% of diet samples, while DAP was detected in 100% of urinary samples, respectively. The average daily intake of preformed DAP was significantly higher than that of parent OP pesticides in our participants. Dimethylphosphate and diethylphosphate were predominant in the preformed DAP, and the estimated average daily intake of total amount of DAP was 78.3 nmol. Fruits and vegetables were the major dietary sources of DAP. Dietary intake of DAP was positively associated with urinary DAP levels, suggesting that a considerable amount of urinary DAP was derived from ingesting preformed DAP. Our results show that attributing urinary DAP levels exclusively to OP pesticide exposure would result in a substantial overestimation of the exposure level. Therefore, the urinary levels of DAP may not be suitable for evaluating OP pesticide exposure in the general urban population.


Subject(s)
Insecticides , Pesticides , Child , Diet , Environmental Exposure/analysis , Female , Humans , Insecticides/analysis , Japan , Organophosphates , Organophosphorus Compounds
8.
ACS Omega ; 6(12): 8588-8597, 2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33817519

ABSTRACT

We developed a highly sensitive method for quantifying 21 bile acids (BAs) in the rat liver by capillary liquid chromatography tandem mass spectrometry (cLC/MS/MS) with one-pot extraction. High recovery rates were obtained for the one-pot methods with either methanol (MeOH) extraction or MeOH/acetonitrile (ACN) (1:1, v/v) mixture extraction; the results obtained for the MeOH/ACN mixture solution were better than the results obtained for MeOH. Thus, we determined that the one-pot method with MeOH/ACN was the most suitable method for the efficient extraction of BAs in the liver. Targeted BAs were well separated by cLC with gradient elution using ammonium acetate (NH4OAc)-MeOH mobile phases. Method validation proved that the intra-day and inter-day accuracies and precisions were primarily less than ±20 and 20% relative standard deviation, respectively. Also, the limit of detection (LOD) and the limit of quantitation (LOQ) were 0.9-10 and 2.3-27 ng/g liver, which proves the high sensitivity of the method. Finally, we quantitated 21 BA concentrations in the liver samples of normal and nonalcoholic steatohepatitis (NASH) rats, both of which were derived from stroke-prone spontaneously hypertensive five (SHRSP5) /Dmcr rat. The hepatic BA profiles were found to be substantially different between the normal and NASH groups; the two groups were clearly separated along the first component axis in the score plots of the principal component analysis. In particular, 10 BAs (ß-muricholic acid (MCA), glyco (G-) cholic acid (CA), G-chenodeoxycholic acid (CDCA), tauro (T-) CA, T-CDCA, T-ursodeoxycholic acid (UDCA), T-lithocholic acid (LCA), T-hiodeoxycholic acid (HDCA), T-α-MCA, and T-ß-MCA) were significantly different between the two groups using Welch's t-test with the false discovery rate correction method, demonstrating BA disruption in the NASH model rat. In conclusion, this method was able to quantify 21 BAs in the rat liver and will evaluate the hepatic BA pathophysiology of rat disease models.

9.
PLoS One ; 15(12): e0243846, 2020.
Article in English | MEDLINE | ID: mdl-33315911

ABSTRACT

Hypertension is an important risk factor for nonalcoholic steatohepatitis. We have previously demonstrated that hypertensive rats fed a high fat and cholesterol (HFC) diet incurred a more severe hepatic inflammatory response and fibrosis. Here we investigated the role of hypertension in NASH by comparing HFC-induced hepatic fibrogenesis between spontaneously hypertensive rats (SHRs) and their normotensive Wistar Kyoto counterpart. Compared to the counterpart, the HFC diet led to stronger aggregation of CD68-positive macrophages in SHRs. HFC feeding also resulted in significantly higher upregulation of the fibrosis-related gene alpha-smooth muscle actin in SHR. The HFC diet induced higher overexpression of serum tissue inhibitor of metalloproteinase-1 (TIMP1) and greater suppression of matrix metalloproteinase-2 (MMP2):TIMP1, MMP8:TIMP1, and MMP9:TIMP1 ratios, as a proxy of the activities of these MMPs in SHR. Administration of the antihypertensive agent hydralazine to SHRs significantly ameliorated HFC-induced liver fibrosis; it suppressed the aggregation of CD68-positive macrophages and the upregulation of platelet-derived growth factor receptor beta, and collagen, type 1, alpha-1 chain. In conclusion, a hypertensive environment exacerbated the hepatic fibrogenetic effects of the HFC diet; while the effects were partially reversed by the antihypertensive agent hydralazine. Our data suggest that antihypertensive drugs hold promise for treating NASH exacerbated by hypertension.


Subject(s)
Antihypertensive Agents/therapeutic use , Disease Progression , Extracellular Matrix/metabolism , Hydralazine/therapeutic use , Hypertension/complications , Hypertension/drug therapy , Liver Cirrhosis/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Body Weight/drug effects , Cholesterol, Dietary , Cytokines/blood , Diet, High-Fat , Gene Expression Regulation/drug effects , Hydralazine/administration & dosage , Hydralazine/pharmacology , Hypertension/physiopathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Matrix Metalloproteinases/blood , Models, Biological , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/pathology , Organ Size/drug effects , Rats, Inbred SHR , Rats, Inbred WKY , Tissue Inhibitor of Metalloproteinase-1/blood
10.
Exp Mol Pathol ; 114: 104437, 2020 06.
Article in English | MEDLINE | ID: mdl-32246926

ABSTRACT

BACKGROUND AND AIMS: Nonalcoholic steatohepatitis (NASH) is linked to an increased risk of cardiovascular disease, regardless of the risk factors in metabolic syndrome. However, the intermediary factors between NASH and cardiovascular disease are still unknown. A previous study revealed that serum and hepatic bile acid (BA) levels are increased in some NASH patients. We aimed to examine whether NASH and cardiovascular disease were aggravated by BA using an animal model. METHOD AND RESULTS: From 10 to 18 weeks of age, SHRSP5/Dmcr rats divided into 3 groups were fed 3 types of high-fat and high-cholesterol (HFC) diets which were changed in the cholic acid (CA) concentration (0%, 2%, or 4%). The nitro oxide synthase inhibition (L-NAME) was administered intraperitoneally from 16 to 18 weeks of age. The 4% CA groups showed the worst LV dysfunction and myocardial fibrosis, and demonstrated severe hepatic fibrosis and lipid depositions. In addition, a large amount of lipid accumulation was observed in the aortas of the 4% CA group, and NFκB and VCAM-1 gene expression levels were increased. These findings were not seen in the 0% CA group. CONCLUSION: In the SHRSP5/Dmcr rat model, NASH and cardiovascular disease were aggravated with increasing BAs concentrations in an HFC diet.


Subject(s)
Bile Acids and Salts/pharmacology , Cardiovascular Diseases/metabolism , Cholic Acid/pharmacology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Bile Acids and Salts/adverse effects , Bile Acids and Salts/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Cholic Acid/adverse effects , Cholic Acid/metabolism , Diet, High-Fat/adverse effects , Gene Expression Regulation/drug effects , Humans , Lipid Metabolism/genetics , NF-kappa B/genetics , NG-Nitroarginine Methyl Ester/pharmacology , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Rats , Vascular Cell Adhesion Molecule-1/genetics
11.
Food Chem Toxicol ; 135: 110927, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31678484

ABSTRACT

The present study was conducted to examine the influence of dietary canola oil (CAN) and partially-hydrogenated soybean oil (HSO) compared to soybean oil (SOY, control) on the morphology and function of testes using miniature pigs as the test subject. Male miniature pigs were fed a diet containing 10%SOY, 9%CAN+1%SOY, or 9%HSO+1%SOY for 18 months. The scheduled autopsies revealed no abnormalities in histopathological examination of the major organs, except the testes. Atrophy of the seminiferous tubules and hyperplasia in the Leydig cells were found in the SOY and CAN groups. DNA microarray analysis indicated downregulation in the CAN and the HSO groups of genes encoding for gonadotropins in the pituitary gland and of enzymes and proteins involved in steroid hormone metabolism in the testes, compared to the SOY group. Plasma levels of sex hormones in the CAN and HSO groups tended to be higher and testosterone and dihydrotestosteorne in the HSO group were significantly higher than in the SOY group. These results demonstrate that testes are morphologically and functionally affected by the dietary oils, while the plasma steroid hormone levels do not necessarily reflect the gene expression, probably owing to feedback regulation via the gonadal hormones in the hypothalamus-pituitary-gonadal axis.


Subject(s)
Rapeseed Oil/toxicity , Soybean Oil/toxicity , Testis/drug effects , Testosterone Congeners/metabolism , Animals , Body Weight/drug effects , Diet , Down-Regulation/genetics , Gene Expression/drug effects , Male , Organ Size/drug effects , Swine , Swine, Miniature , Testis/metabolism
12.
Sci Rep ; 9(1): 12514, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31467308

ABSTRACT

Precise molecular pathways involved in the progression of non-alcoholic steatohepatitis (NASH) remain to be elucidated. As Mallory-Denk bodies were occasionally observed in the enlarged hepatocytes in NASH model rat (SHRSP5/Dmcr) fed high-fat and high-cholesterol (HFC) diet, we aimed to clarify the roles of autophagy and endoplasmic reticulum (ER) stress in NASH progression. Male SHRSP5/Dmcr were randomly divided into 4 groups. Two groups were fed a control diet; the other two groups were fed a HFC diet for 2 and 8 weeks, respectively. The HFC diet increased the autophagy-related proteins levels and microtubule-associated protein 1 light chain 3-II/I ratio after 2 and 8 weeks, respectively. However, regarding ER stress-related proteins, the HFC diet decreased the levels of phosphorylated (p-) inositol-requiring kinase-1 (p-IRE-1) and p-protein kinase RNA-like ER kinase after 2 weeks. Additionally, the HFC diet increased anti-ubiquitin-positive cells and the level of the autophagy substrate p62, suggesting that the HFC diet induced dysfunction in ubiquitin-dependent protein degradation pathways. In conclusion, the HFC diet arrested the autophagy process in the liver; this was particularly associated with decreases in p-IRE-1 expression.


Subject(s)
Autophagy , Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Endoribonucleases/metabolism , Liver/physiopathology , Multienzyme Complexes/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cholesterol, Dietary/metabolism , Disease Models, Animal , Endoplasmic Reticulum Stress , Endoribonucleases/genetics , Humans , Liver/enzymology , Liver/metabolism , Male , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Multienzyme Complexes/genetics , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/physiopathology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Rats
13.
Nutrients ; 11(8)2019 Aug 20.
Article in English | MEDLINE | ID: mdl-31434232

ABSTRACT

We examined the nutritional status and its association with behavioral psychiatric symptoms of dementia (BPSD) among 741 memory clinic patients (normal cognition (NC), 152; mild cognitive impairment (MCI), 271; early-stage Alzheimer disease (AD), 318). Nutritional status and BPSD were assessed using the Mini Nutritional Assessment Short-Form (MNA-SF) and the Dementia Behavior Disturbance Scale (DBD), respectively. Compared to subjects with NC, more subjects with MCI and early-stage AD were at risk of malnutrition (MNA-SF, 8-11: NC, 34.2%; MCI, 47.5%; early-stage AD, 53.8%) and were malnourished (MNA-SF, 0-7: NC, 4.6%; MCI, 5.9%; early-stage AD, 8.2%). Among patients with MCI or early-stage AD, those at risk of/with malnutrition showed higher DBD scores than those well-nourished (12.7 ± 9.0 vs. 9.5 ± 7.3; p < 0.001). Moreover, analysis of covariance adjusting for confounders showed that nutritional status was significantly associated with specific BPSD, including "verbal aggressiveness/emotional disinhibition" (F = 5.87, p = 0.016) and "apathy/memory impairment" (F = 15.38, p < 0.001), which were revealed by factor analysis of DBD. Our results suggest that malnutrition is common among older adults with mild cognitive decline, and possibility that nutritional problems are associated with individual BPSD.


Subject(s)
Alzheimer Disease/psychology , Behavior , Cognitive Dysfunction/psychology , Dementia/psychology , Malnutrition/psychology , Aged , Aged, 80 and over , Female , Humans , Malnutrition/complications , Malnutrition/epidemiology , Nutrition Assessment , Nutritional Status , Psychiatric Status Rating Scales
14.
PLoS One ; 13(2): e0192863, 2018.
Article in English | MEDLINE | ID: mdl-29438418

ABSTRACT

During middle age, women are less susceptible to nonalcoholic steatohepatitis (NASH) than men. Thus, we investigated the underlying molecular mechanisms behind these sexual differences using an established rat model of NASH. Mature female and male stroke-prone spontaneously hypertensive 5/Dmcr rats were fed control or high-fat-cholesterol (HFC) diets for 2, 8, and 14 weeks. Although HFC-induced hepatic fibrosis was markedly less severe in females than in males, only minor gender differences were observed in expression levels of cytochrome P450 enzymes (CYP)7A1, CYP8B1 CYP27A1, and CYP7B1, and multidrug resistance-associated protein 3, and bile salt export pump, which are involved in fibrosis-related bile acid (BA) kinetics. However, the BA detoxification-related enzymes UDP-glucuronosyltransferase (UGT) and sulfotransferase (SULT) 2A1, and the nuclear receptors constitutive androstane receptor (CAR) and pregnane X receptor (PXR), were strongly suppressed in HFC-fed males, and were only slightly changed in HFC-diet fed females. Expression levels of the farnesoid X receptor and its small heterodimer partner were similarly regulated in a gender-dependent fashion following HFC feeding. Hence, the pronounced female resistance to HFC-induced liver damage likely reflects sustained expression of the nuclear receptors CAR and PXR and the BA detoxification enzymes UGT and SULT.


Subject(s)
Bile Acids and Salts/metabolism , Diet, High-Fat/adverse effects , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Animals , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/adverse effects , Constitutive Androstane Receptor , Disease Models, Animal , Disease Susceptibility , Female , Gene Expression , Glucuronosyltransferase/metabolism , Liver Cirrhosis/pathology , Male , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Pregnane X Receptor , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Sex Characteristics , Sulfotransferases/metabolism
15.
J Atheroscler Thromb ; 25(5): 439-453, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29162773

ABSTRACT

AIM: Non-alcoholic steatohepatitis (NASH) increases cardiovascular risk regardless of risk factors in metabolic syndrome. However, the intermediary factors between NASH and vascular disease are still unknown because a suitable animal model has never been established. The stroke-prone (SP) spontaneously hypertensive rat, SHRSP5/Dmcr, simultaneously develops hypertension, acute arterial lipid deposits in mesenteric arteries, and NASH when feed with a high-fat and high-cholesterol (HFC) diet. We investigated whether SHRSP5/Dmcr affected with NASH aggravates the cardiac or vascular dysfunction. METHOD: Wister Kyoto and SHRSP5/Dmcr rats were divided into 4 groups of 5 rats each, and fed with a SP or HFC diet. After 8 weeks of HFC or SP diet feeding, glucose and insulin resistance, echocardiography, blood biochemistry, histopathological staining, and endothelial function in aorta were evaluated. RESULTS: We demonstrate that SHRSP5/Dmcr rats fed with a HFC diet presented with cardiac and vascular dysfunction caused by cardiac fibrosis, endothelial dysfunction, and left ventricular diastolic dysfunction, in association with NASH and hypertension. These cardiac and vascular dysfunctions were aggravated and not associated with the presence of hypertension, glucose metabolism disorder, and/or obesity. CONCLUSIONS: SHRSP5/Dmcr rats may be a suitable animal model for elucidating the organ interaction between NASH and cardiac or vascular dysfunction.


Subject(s)
Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Endothelium, Vascular/pathology , Fibrosis/etiology , Myocardial Infarction/etiology , Non-alcoholic Fatty Liver Disease/physiopathology , Ventricular Dysfunction, Left/etiology , Animals , Diastole , Fibrosis/pathology , Male , Myocardial Infarction/pathology , Non-alcoholic Fatty Liver Disease/complications , Rats , Rats, Inbred SHR , Ventricular Dysfunction, Left/pathology
16.
Int J Exp Pathol ; 99(6): 282-294, 2018 12.
Article in English | MEDLINE | ID: mdl-30680827

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is linked to increased cardiovascular risk, independent of the broad spectrum of metabolic syndrome risk factors. Stroke-prone (SP) spontaneously hypertensive rats (SHRSP5/Dmcr) fed a high-fat and high-cholesterol (HFC) diet developed hepatic lesions similar to those in human NASH pathology. These rats simultaneously developed lipid deposits in the mesenteric arteries, cardiac fibrosis, endothelial dysfunction and left ventricle (LV) diastolic dysfunction. However, the intermediary factors between NASH and cardiovascular disease are still unknown. We investigated whether NASH aggravates nitric oxide (NO) synthase inhibition-induced arteriosclerosis in SHRSP5/Dmcr rats. Wistar Kyoto and SHRSP5/Dmcr rats were divided into 4 groups of 5 and fed the stroke-prone (SP) or HFC diets for 8 weeks. To induce NO synthase inhibition, Nω -nitro-L-arginine methyl ester hydrochloride (L-NAME) mixed with drinking water was administered in the final 2 weeks. The NASH+L-NAME group demonstrated the following characteristics related to arteriosclerosis and myocardial ischaemia: (a) LV systolic dysfunction with asynergy, (b) replacement fibrosis caused by the shedding of cardiomyocytes and (c) arterial lipid deposition and coronary occlusion secondary to endothelial dysfunction. These characteristics were not observed in the NASH or non-NASH+L-NAME groups. The SHRSP5/Dmcr rat model demonstrates that NASH significantly aggravates cardiovascular risk.


Subject(s)
Arteriosclerosis/etiology , Nitric Oxide Synthase/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease/complications , Animals , Arteriosclerosis/pathology , Arteriosclerosis/physiopathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Endothelium, Vascular/physiopathology , Heart Ventricles/pathology , Liver/pathology , Male , Myocardial Ischemia/etiology , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , NG-Nitroarginine Methyl Ester/pharmacology , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/physiopathology , Organ Size , Rats, Inbred SHR , Rats, Inbred WKY , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
17.
Nutrients ; 9(9)2017 Sep 14.
Article in English | MEDLINE | ID: mdl-28906458

ABSTRACT

Populations with essential hypertension have a high risk of nonalcoholic steatohepatitis (NASH). In this study, we investigated the mechanism that underlies the progression of hypertension-associated NASH by comparing differences in the development of high fat and cholesterol (HFC) diet-induced NASH among three strains of rats, i.e., two hypertensive strains comprising spontaneously hypertensive rats and the stroke-prone spontaneously hypertensive 5/Dmcr, and the original Wistar Kyoto rats as the normotensive control. We investigated histopathological changes and molecular signals related to inflammation in the liver after feeding with the HFC diet for 8 weeks. The diet induced severe lobular inflammation and fibrosis in the livers of the hypertensive rats, whereas it only caused mild steatohepatitis in the normotensive rats. An increased activation of proinflammatory signaling (transforming growth factor-ß1/mitogen-activated protein kinases pathway) was observed in the hypertensive strains fed with the HFC diet. In addition, the HFC diet suppressed the nuclear factor erythroid 2-related factor 2 pathway in the hypertensive rats and led to lower increases in the hepatic expression of heme oxygenase-1, which has anti-oxidative and anti-inflammatory activities. In conclusion, these signaling pathways might play crucial roles in the development of hypertension-associated NASH.


Subject(s)
Cholesterol, Dietary/adverse effects , Hypertension/pathology , Inflammation/pathology , Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cholesterol/blood , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Cholesterol, Dietary/administration & dosage , Diet, High-Fat/adverse effects , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Disease Models, Animal , Female , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Hypertension/genetics , Inflammation/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Male , Mitogen-Activated Protein Kinases/genetics , NF-E2-Related Factor 2/genetics , NF-kappa B/genetics , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction , Triglycerides/blood , gamma-Glutamyltransferase/blood
18.
Environ Health Prev Med ; 21(5): 368-381, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27209494

ABSTRACT

OBJECTIVES: High-fat and -cholesterol diet (HFC) induced fibrotic steatohepatitis in stroke-prone spontaneously hypertensive rat (SHRSP) 5/Dmcr, the fifth substrain from SHRSP, by dysregulating bile acid (BA) kinetics. This study aimed to clarify the histopathological and BA kinetic differences in HFC-induced fibrosis between SHRSP5/Dmcr and SHRSP. METHODS: Ten-week-old male SHRSP5/Dmcr and SHRSP were randomly allocated to groups and fed with either control diet or HFC for 2 and 8 weeks. The liver histopathology, biochemical features, and molecular signaling involved in BA kinetics were measured. RESULTS: HFC caused more severe hepatocyte ballooning, macrovesicular steatosis and fibrosis in SHRSP5/Dmcr than in SHRSP. It was noted that fibrosis was disproportionately formed in retroperitoneal side of both strains. As for BA kinetics, HFC greatly increased the level of Cyp7a1 and Cyp7b1 to the same degree in both strains at 8 weeks, while multidrug resistance-associated protein 3 was greater in SHRSP5/Dmcr than SHRSP. The diet decreased the level of bile salt export pump by the same degree in both strains, while constitutive androstane receptor, pregnane X receptor, and UDP-glucuronosyltransferase activity more prominent in SHRSP5/Dmcr than SHRSP at 8 weeks. In the fibrosis-related genes, only expression of collagen, type I, alpha 1 mRNA was greater in SHRSP5/Dmcr than SHRSP. CONCLUSIONS: The greater progression of fibrosis in SHRSP5/Dmcr induced by HFC may be due to greater suppression of UDP-glucuronosyltransferase activity detoxifying toxicants, such as hydrophobic BAs.


Subject(s)
Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Fatty Liver/genetics , Fatty Liver/pathology , Animals , Disease Progression , Fatty Liver/enzymology , Fibrosis , Inactivation, Metabolic , Liver/metabolism , Liver/pathology , Male , Random Allocation , Rats , Rats, Inbred SHR
19.
PLoS One ; 11(1): e0145939, 2016.
Article in English | MEDLINE | ID: mdl-26727365

ABSTRACT

Nonalcoholic steatohepatitis is related to lifestyle, particularly to dietary habits. We developed diet-induced fibrotic steatohepatitis model stroke-prone spontaneously hypertensive 5/Dmcr (SHRSP5/Dmcr) rats showing steatosis, hepatic inflammation, and severe fibrosis induced by high-fat and -cholesterol (HFC) diet feeding. We aimed to clarify the efficacy of dietary intervention on the disease before and after the appearance of fibrosis. Male SHRSP5/Dmcr rats were divided into 9 groups; of these, 6 groups were fed control or HFC diet for several weeks and the remaining 3 groups represented the dietary intervention groups, which were fed the control diet after HFC diet feeding for 2 (before the appearance of fibrosis) or 8 (after the appearance of fibrosis) weeks. Dietary intervention before the appearance of fibrosis significantly improved the steatosis and reset the increased serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), and serum total cholesterol (TC) levels. However, dietary intervention after the appearance of fibrosis was unable to reset the levels of hepatic TC, serum ALT, and fibrogenesis-related markers and had only a minor influence on hepatic fibrosis, although it reset the increased expression of transforming growth factor (TGF)-ß1 and α-smooth muscle actin (SMA). It was noted that dietary intervention improved the increased AST levels; however, aggregated CD68-positive cells were still observed around the fibrosis area, which may be related to the findings of inflammatory cytokine mRNAs. Taken together, dietary intervention for fibrotic steatohepatitis improved steatosis, although it could not completely improve fibrosis.


Subject(s)
Cholesterol, Dietary/adverse effects , Dietary Fats/administration & dosage , Non-alcoholic Fatty Liver Disease/therapy , Alanine Transaminase/biosynthesis , Animals , Antigens, CD/blood , Antigens, Differentiation, Myelomonocytic/blood , Aspartate Aminotransferases/blood , Biomarkers/blood , Body Weight , Cytokines/genetics , Dietary Fats/adverse effects , Inflammation Mediators/blood , Lipids/blood , Male , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/physiopathology , Organ Size , RNA, Messenger/genetics , Rats
20.
J Physiol Sci ; 65(5): 417-25, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25972297

ABSTRACT

Repeated cold stress (RCS) is known to transiently induce functional disorders associated with hypotension and hyperalgesia. In this study, we investigated the effects of RCS (24 and 4 °C alternately at 30-min intervals during the day and 4 °C at night for 2 days, followed by 4 °C on the next 2 consecutive nights) on the thresholds for cutaneous mechanical pain responses and on peripheral expression of "pain-related genes" in SHRSP5/Dmcr rats, which are derived from stroke-prone spontaneously hypertensive rats. To define genes peripherally regulated by RCS, we detected changes in the expression of pain-related genes in dorsal root ganglion cells by PCR-based cDNA subtraction analysis or DNA microarray analysis, and confirmed the changes by RT-PCR. We found significantly changed expression in eight pain-related genes (upregulated: Fyn, St8sia1, and Tac 1; downregulated: Ctsb, Fstl1, Itpr1, Npy, S100a10). At least some of these genes may play key roles in hyperalgesia induced by RCS.


Subject(s)
Cold Temperature , Gene Expression Profiling , Hyperalgesia/genetics , Pain Threshold , Stress, Physiological , Animals , Blood Pressure/genetics , Gene Expression Profiling/methods , Gene Expression Regulation , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Male , Oligonucleotide Array Sequence Analysis , Rats, Inbred SHR , Rats, Sprague-Dawley , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...