Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 148
1.
Exp Biol Med (Maywood) ; 246(24): 2671-2678, 2021 12.
Article En | MEDLINE | ID: mdl-34525859

Dietary cholesterol supplements cause hypercholesterolemia and atherosclerosis along with a reduction of copper concentrations in the atherosclerotic wall in animal models. This study was to determine if target-specific copper delivery to the copper-deficient atherosclerotic wall can block the pathogenesis of atherosclerosis. Male New Zealand white rabbits, 10-weeks-old and averaged 2.0 kg, were fed a diet containing 1% (w/w) cholesterol or the same diet without cholesterol as control. Twelve weeks after the feeding, the animals were injected with copper-albumin microbubbles and subjected to ultrasound sonication specifically directed at the atherosclerotic lesions (Cu-MB-US) for target-specific copper delivery, twice a week for four weeks. This regiment was repeated 3 times with a gap of two weeks in between. Two weeks after the last treatment, the animals were harvested for analyses of serum and aortic pathological changes. Compared to controls, rabbits fed cholesterol-rich diet developed atherosclerotic lesion with a reduction in copper concentrations in the lesion tissue. Cu-MB-US treatment significantly increased copper concentrations in the lesion, and reduced the size of the lesion. Furthermore, copper repletion reduced the number of apoptotic cells as well as the content of cholesterol and phospholipids in the atherosclerotic lesion without a disturbance of the stability of the lesion. The results thus demonstrate that target-specific copper supplementation suppresses the progression of atherosclerosis at least in part through preventing endothelial cell death, thus reducing lipid infiltration in the atherosclerotic lesion.


Atherosclerosis/pathology , Copper/administration & dosage , Microbubbles , Ultrasonics , Animals , Aorta, Abdominal/pathology , Cholesterol, Dietary/toxicity , Diet, High-Fat/adverse effects , Drug Delivery Systems , Male , Rabbits , Ultrasonics/methods
2.
Toxicol Appl Pharmacol ; 415: 115430, 2021 03 15.
Article En | MEDLINE | ID: mdl-33524446

Air pollutants may increase risk for cardiopulmonary disease, particularly in susceptible populations with metabolic stressors such as diabetes and unhealthy diet. We investigated effects of inhaled ozone exposure and high-cholesterol diet (HCD) in healthy Wistar and Wistar-derived Goto-Kakizaki (GK) rats, a non-obese model of type 2 diabetes. Male rats (4-week old) were fed normal diet (ND) or HCD for 12 weeks and then exposed to filtered air or 1.0 ppm ozone (6 h/day) for 1 or 2 days. We examined pulmonary, vascular, hematology, and inflammatory responses after each exposure plus an 18-h recovery period. In both strains, ozone induced acute bronchiolar epithelial necrosis and inflammation on histopathology and pulmonary protein leakage and neutrophilia; the protein leakage was more rapid and persistent in GK compared to Wistar rats. Ozone also decreased lymphocytes after day 1 in both strains consuming ND (~50%), while HCD increased circulating leukocytes. Ozone increased plasma thrombin/antithrombin complexes and platelet disaggregation in Wistar rats on HCD and exacerbated diet effects on serum IFN-γ, IL-6, KC-GRO, IL-13, and TNF-α, which were higher with HCD (Wistar>GK). Ex vivo aortic contractility to phenylephrine was lower in GK versus Wistar rats at baseline(~30%); ozone enhanced this effect in Wistar rats on ND. GK rats on HCD had higher aortic e-NOS and tPA expression compared to Wistar rats. Ozone increased e-NOS in GK rats on ND (~3-fold) and Wistar rats on HCD (~2-fold). These findings demonstrate ways in which underlying diabetes and HCD may exacerbate pulmonary, systemic, and vascular effects of inhaled pollutants.


Air Pollutants/toxicity , Aorta, Thoracic/drug effects , Cholesterol, Dietary/toxicity , Diabetes Mellitus, Type 2/complications , Diet, Atherogenic/adverse effects , Lung Injury/chemically induced , Lung/drug effects , Ozone/toxicity , Vascular Diseases/chemically induced , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Biomarkers/blood , Blood Platelets/drug effects , Blood Platelets/metabolism , Cholesterol, Dietary/metabolism , Cytokines/blood , Diabetes Mellitus, Type 2/blood , Disease Models, Animal , Inflammation Mediators/blood , Inhalation Exposure , Lung/metabolism , Lung/pathology , Lung Injury/blood , Lung Injury/pathology , Male , Necrosis , Pulmonary Edema/blood , Pulmonary Edema/chemically induced , Pulmonary Edema/pathology , Rats, Wistar , Vascular Diseases/blood , Vascular Diseases/physiopathology , Vasoconstriction/drug effects
3.
Toxicol Appl Pharmacol ; 415: 115427, 2021 03 15.
Article En | MEDLINE | ID: mdl-33524448

Epidemiological studies show that individuals with underlying diabetes and diet-associated ailments are more susceptible than healthy individuals to adverse health effects of air pollution. Exposure to air pollutants can induce metabolic stress and increase cardiometabolic disease risk. Using male Wistar and Wistar-derived Goto-Kakizaki (GK) rats, which exhibit a non-obese type-2 diabetes phenotype, we investigated whether two key metabolic stressors, type-2 diabetes and a high-cholesterol atherogenic diet, exacerbate ozone-induced metabolic effects. Rats were fed a normal control diet (ND) or high-cholesterol diet (HCD) for 12 weeks and then exposed to filtered air or 1.0-ppm ozone (6 h/day) for 1 or 2 days. Metabolic responses were analyzed at the end of each day and after an 18-h recovery period following the 2-day exposure. In GK rats, baseline hyperglycemia and glucose intolerance were exacerbated by HCD vs. ND and by ozone vs. air. HCD also resulted in higher insulin in ozone-exposed GK rats and circulating lipase, aspartate transaminase, and alanine transaminase in all groups (Wistar>GK). Histopathological effects induced by HCD in the liver, which included macrovesicular vacuolation and hepatocellular necrosis, were more severe in Wistar vs. GK rats. Liver gene expression in Wistar and GK rats fed ND showed numerous strain differences, including evidence of increased lipid metabolizing activity and ozone-induced alterations in glucose and lipid transporters, specifically in GK rats. Collectively, these findings indicate that peripheral metabolic alterations induced by diabetes and high-cholesterol diet can enhance susceptibility to the metabolic effects of inhaled pollutants.


Adipose Tissue, White/drug effects , Air Pollutants/toxicity , Cholesterol, Dietary/toxicity , Diabetes Mellitus, Type 2/complications , Diet, High-Fat/adverse effects , Energy Metabolism/drug effects , Liver/drug effects , Ozone/toxicity , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Composition/drug effects , Cholesterol, Dietary/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Gene Expression Regulation , Inhalation Exposure , Insulin/blood , Lipids/blood , Liver/metabolism , Liver/pathology , Male , Rats, Wistar , Species Specificity
4.
Pharmacol Res ; 160: 105201, 2020 10.
Article En | MEDLINE | ID: mdl-32942017

BACKGROUND AND PURPOSE: The pathogenesis of cardiomyopathy in metabolically unhealthy obesity (MUO) has been well studied. However, the pathogenesis of cardiomyopathy typically associated with high cholesterol levels in metabolically unhealthy nonobesity (MUNO) remains unclear. We investigated whether cholesterol-generated LysoPCs contribute to cardiomyopathy and the role of cytosolic phospholipase A2 (cPLA2) inhibitor in cholesterol-induced MUNO. EXPERIMENTAL APPROACH: Cholesterol diet was performed in Sprague-Dawley rats that were fed either regular chow (C), or high cholesterol chow (HC), or HC diet with 10 % fructose in drinking water (HCF) for 12 weeks. LysoPCs levels were subsequently measured in rats and in MUNO human patients. The effects of cholesterol-mediated LysoPCs on cardiac injury, and the action of cPLA2 inhibitor, AACOCF3, were further assessed in H9C2 cardiomyocytes. KEY RESULTS: HC and HCF rats fed cholesterol diets demonstrated a MUNO-phenotype and cholesterol-induced dilated cardiomyopathy (DCM). Upregulated levels of LysoPCs were found in rat myocardium and the plasma in MUNO human patients. Further testing in H9C2 cardiomyocytes revealed that cholesterol-induced atrophy and death of cardiomyocytes was due to mitochondrial dysfunction and conditions favoring DCM (i.e. reduced mRNA expression of ANF, BNP, DSP, and atrogin-1), and that AACOCF3 counteracted the cholesterol-induced DCM phenotype. CONCLUSION AND IMPLICATIONS: Cholesterol-induced MUNO-DCM phenotype was counteracted by cPLA2 inhibitor, which is potentially useful for the treatment of LysoPCs-associated DCM in MUNO.


Cardiomyopathy, Dilated/drug therapy , Cholesterol, Dietary/toxicity , Metabolic Diseases/drug therapy , Myocytes, Cardiac/drug effects , Phospholipase A2 Inhibitors/therapeutic use , Animals , Cell Line , Diet , Electrocardiography , Fructose/toxicity , Hemodynamics/drug effects , Humans , Lysophosphatidylcholines/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Rats , Rats, Sprague-Dawley
5.
Redox Rep ; 25(1): 51-58, 2020 Dec.
Article En | MEDLINE | ID: mdl-32396454

ABSTRACTObjectives: This study explores the protective role of losartan (LT) against oxidative and inflammatory damages in different physiological systems including heart, liver, and kidney tissue in hypercholesterolemic rats.Methods: After induction of hypercholesterolemia by high cholesterol diet for 6 weeks, LT was administered for 4 weeks. In serum, the levels of lipoproteins, aminotransferases, creatine kinases, urea, apoptosis, and inflammatory markers were measured. In cardiac, hepatic, and renal tissues, lipid peroxidation product and GSH as well as antioxidant enzymatic activities were assayed. Finally, histopathological assessment evaluated the structural damage in cardiac, hepatic, and renal tissues.Results: Serum markers of cardiac, hepatic, and renal toxicities including creatine kinases, aminotransferases, and urea were attenuated by LT in hypercholesterolemic animals. Moreover, LT markedly corrected the elevated levels of lipoproteins, apoptosis, and inflammatory biomarkers. Hypercholesterolemia-induced lipid peroxidation, low GSH levels, and diminished activities of antioxidant enzymes were prominently improved in LT treated animals. Histopathological alterations by hypercholesterolemia in heart, liver, and kidney tissues were ameliorated by LT.Conclusion: This study confirmed the pathological enrollment of renin-angiotensin system in hypercholesterolemia-associated metabolic alterations. LT had a significant cardiac, hepatic, and renal protective role against these impairments through down-regulation of oxidative damage, inflammation and necrosis.


Antihypertensive Agents/pharmacology , Cholesterol, Dietary/toxicity , Hypercholesterolemia/complications , Inflammation/drug therapy , Losartan/pharmacology , Oxidative Stress , Renin-Angiotensin System/drug effects , Animals , Hypercholesterolemia/chemically induced , Hypercholesterolemia/pathology , Inflammation/etiology , Inflammation/pathology , Male , Rats , Rats, Wistar
6.
Article En | MEDLINE | ID: mdl-31446160

Neuroinflammation and metabolic deficits contribute to the etiology of human affective disorders, such as anxiety and depression. The zebrafish (Danio rerio) has recently emerged as a powerful new model organism in CNS disease modeling. Here, we exposed zebrafish to 2% glucose and 10% cholesterol for 19 days to experimentally induce type 2 diabetes (DM) and to assess stress responses, microglia, inflammation and apoptosis. We analyzed zebrafish anxiety-like behavior in the novel tank and light-dark box (Days 15-16) tests, as well as examined their biochemical and genomic biomarkers (Day 19). Confirming DM-like state in zebrafish, we found higher whole-body glucose, triglyceride, total cholesterol, low-density lipoprotein levels and glucagon mRNA expression, and lower high-density lipoprotein levels. DM zebrafish also showed anxiety-like behavior, elevated whole-body cortisol and cytokines IFN-γ and IL-4, as well as higher brain mRNA expression of the glucocorticoid receptor, CD11b (a microglial biomarker), pro-inflammatory cytokines IL-6 and TNF-α (but not IL-1ß or anti-inflammatory cytokines IL-4 and IL-10), GFAP (an astrocytal biomarker), neurotrophin BDNF, its receptors p75 and TrkB, as well as apoptotic Bax and Caspase-3 (but not BCl-2) genes. Collectively, this supports the overlapping nature of DM-related affective pathogenesis and emphasizes the role of peripheral and central inflammation and apoptosis in DM-related affective and neuroendocrine deficits in zebrafish.


Apoptosis/physiology , Cholesterol, Dietary/toxicity , Diabetes Mellitus, Experimental/metabolism , Glucose/toxicity , Inflammation Mediators/metabolism , Microglia/metabolism , Animals , Apoptosis/drug effects , Cholesterol, Dietary/adverse effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/metabolism , Female , Glucose/administration & dosage , Male , Microglia/drug effects , Zebrafish
7.
Pathol Res Pract ; 215(11): 152599, 2019 Nov.
Article En | MEDLINE | ID: mdl-31564568

Understanding of the pathogenesis of nonalcoholic steatohepatitis (NASH)-associated fibrosis has been hampered by the lack of a comprehensive and physiological small animal model of NASH with fibrosis. Feeding a high-fat and high-cholesterol (HFC) diet supplemented with cholic acid to rats is known to replicate human NASH pathology, and it induces fibrosis earlier than with an HFC diet alone. In the present study, physiological and histopathological observations from 65 Sprague-Dawley (SD) rats fed an HFC diet with or without cholic acid for 9 or 18 weeks in our laboratory between January 2013 and February 2018 were retrospectively reviewed. The liver weight/body weight ratio at the end of the rearing period was higher in rats fed an HFC diet than in rats fed a normal diet in a cholesterol dose-, cholic acid dose-, or rearing period dependent manner. Dietary fat, cholesterol and/or cholic acid and rearing period affected the histopathologic severity of NASH. Overall, 56 (86.2%) of 65 SD rats fed an HFC diet for 9 or 18 weeks developed histopathologically proven NASH. It is noted that the SD rats fed an HFC diet supplemented with 2% (w/w) cholic acid for 18 weeks frequently developed advanced fibrosis, including cirrhosis. Thus, this diet-induced NASH rat model is likely to be a highly reproducible.


Cholesterol, Dietary/toxicity , Cholic Acid/toxicity , Dietary Fats/toxicity , Disease Models, Animal , Non-alcoholic Fatty Liver Disease/pathology , Animals , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Liver Cirrhosis, Experimental/etiology , Liver Cirrhosis, Experimental/pathology , Rats , Rats, Sprague-Dawley
8.
Arch Toxicol ; 93(6): 1713-1725, 2019 06.
Article En | MEDLINE | ID: mdl-31004178

Previous epidemiological studies have suggested a link between high-cholesterol intake and liver disease progression, including hepatocellular carcinoma (HCC). However, the precise mechanism of hepatotoxicity and hepatocarcinogenesis caused by excessive cholesterol consumption remains unclear. We aimed to investigate the impact of dietary cholesterol using hepatitis C virus core gene transgenic (HCVcpTg) mice, which spontaneously developed HCC with age. Male HCVcpTg mice were treated for 15 months with either a control diet or an isocaloric diet containing 1.5% cholesterol, and liver phenotypes and tumor-associated signaling pathways were evaluated. The high-cholesterol diet-fed HCVcpTg mice exhibited a significantly higher incidence of liver tumors compared with the control diet mice (100% vs. 41%, P < 0.001). The diet induced steatohepatitis with pericellular fibrosis and evoked higher mRNA expression of pro-inflammatory and pro-fibrotic mediators along with enhanced hepatocyte proliferation and greater oxidative and endoplasmic reticulum stress in the liver. Moreover, long-term consumption of cholesterol-rich diet activated nuclear factor-kappa B (NF-κB) and p62/sequestosome 1 (Sqstm1)-nuclear factor erythroid 2 (NRF2) axis, enhanced fibrogenesis, and consequently accelerated hepatic tumorigenesis. In conclusion, these results demonstrate that a high-cholesterol diet facilitates liver tumorigenesis by inducing steatohepatitis, promoting hepatocyte division, and up-regulating cellular stress and pro-inflammatory NF-κB and detoxifying p62/Sqstm1-NRF2 signals. Therefore, high dietary cholesterol should be avoided in HCV-infected patients to prevent development of steatohepatitis, liver fibrosis, and HCC.


Cholesterol, Dietary/toxicity , Genes, Viral/genetics , Hepatitis C/genetics , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/genetics , Animals , Cell Proliferation/drug effects , Diet , Hepatocytes/drug effects , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Liver Function Tests , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
9.
FASEB J ; 33(6): 7301-7314, 2019 06.
Article En | MEDLINE | ID: mdl-30860872

Hypercholesterolemia is reported to increase reactive oxygen species (ROS) and to promote breast cancer progression. ROS play an important role in tumor biology, and xanthine oxidase (XO) is an enzyme that generates ROS. The effects of febuxostat (FBX), an XO inhibitor, on breast cancer cell migration under LDL stimulation in vitro and metastasis of breast cancer associated with hypercholesterolemia in vivo were studied. In vitro, FBX significantly inhibited LDL-induced ROS production and cell migration. Treatment of small interfering RNA against XO was consistent with the findings of FBX treatment. In vivo, a significant increase of tumor growth and pulmonary metastasis was observed in a xenograft mouse model with 4T1 cells on a high cholesterol diet (HCD), both of which were markedly inhibited by FBX or allopurinol treatment. Moreover, ERK represented the main target-signaling pathway that was affected by FBX treatment in a xenograft mouse model on an HCD evaluated by NanoString nCounter analysis. Consistently, MEK/ERK inhibitors directly decreased the LDL-induced cell migration in vitro. In conclusion, FBX mitigates breast cancer cell migration and pulmonary metastasis in the hyperlipidemic condition, associated with decreased ROS generation and MAPK phosphorylation. The inhibition of ERK pathways is likely to underlie the XO inhibitor-mediated suppression of breast cancer cell migration.-Oh, S.-H., Choi, S.-Y., Choi, H.-J., Ryu, H.-M., Kim, Y.-J., Jung, H.-Y., Cho, J.-H., Kim, C.-D., Park, S.-H., Kwon, T.-H., Kim, Y.-L. The emerging role of xanthine oxidase inhibition for suppression of breast cancer cell migration and metastasis associated with hypercholesterolemia.


Breast Neoplasms/enzymology , Hypercholesterolemia/complications , Lung Neoplasms/secondary , Neoplasm Proteins/antagonists & inhibitors , Xanthine Oxidase/antagonists & inhibitors , Allopurinol/pharmacology , Allopurinol/therapeutic use , Animals , Breast Neoplasms/complications , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cholesterol, Dietary/toxicity , Disease Progression , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Febuxostat/pharmacology , Febuxostat/therapeutic use , Female , Flavonoids/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/etiology , Lung Neoplasms/prevention & control , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplasm Proteins/genetics , RNA, Small Interfering/pharmacology , Random Allocation , Reactive Oxygen Species , Time-Lapse Imaging , Xanthine Oxidase/genetics , Xenograft Model Antitumor Assays
10.
Arterioscler Thromb Vasc Biol ; 39(3): 432-445, 2019 03.
Article En | MEDLINE | ID: mdl-30626205

Objective- Vascular smooth muscle cell (VSMC) transformation to an osteochondrogenic phenotype is an initial step toward arterial calcification, which is highly correlated with cardiovascular disease-related morbidity and mortality. TLR2 (Toll-like receptor 2) plays a pathogenic role in the development of vascular diseases, but its regulation in calcification of arteries and VSMCs remains unclear. We postulate that TLR2-mediated inflammation participates in mediating atherosclerotic arterial calcification and VSMC calcification. Approach and Results- We found that ApoE-/- Tlr2-/- genotype in mice suppressed high-fat diet-induced atherosclerotic plaques formation during initiation but progressively lost its preventative capacity, compared with ApoE-/- mice. However, TLR2 deficiency prohibited high-fat diet-induced advanced atherosclerotic calcification, chondrogenic metaplasia, and OPG (osteoprotegerin) downregulation in the calcified lesions. Incubation of VSMCs in a calcifying medium revealed that TLR2 agonists significantly increased VSMC calcification and chondrogenic differentiation. Furthermore, TLR2 deficiency suppressed TLR2 agonist-mediated VSMC chondrogenic differentiation and consequent calcification, which were triggered via the concerted actions of IL (interleukin)-6-mediated RANKL (receptor activator of nuclear factor κB ligand) induction and OPG suppression. Inhibition experiments with pharmacological inhibitors demonstrated that IL-6-mediated RANKL induction is signaled by p38 and ERK1/2 (extracellular signal-regulated kinase 1/2) pathways, whereas the OPG is suppressed via NF-κB (nuclear factor κB) dependent signaling mediated by ERK1/2. Conclusions- We concluded that on ligand binding, TLR2 activates p38 and ERK1/2 signaling to selectively modulate the upregulation of IL-6-mediated RANKL and downregulation of OPG. These signaling pathways act in concert to induce chondrogenic transdifferentiation of VSMCs, which in turn leads to vascular calcification during the pathogenesis of atherosclerosis.


Atherosclerosis/pathology , Calcinosis/metabolism , Calcinosis/pathology , Chondrogenesis/physiology , Interleukin-6/physiology , MAP Kinase Signaling System , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Osteoprotegerin/biosynthesis , RANK Ligand/biosynthesis , Toll-Like Receptor 2/physiology , Animals , Aortic Diseases/etiology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Calcinosis/genetics , Cells, Cultured , Cholesterol, Dietary/toxicity , Diet, High-Fat/adverse effects , Dietary Fats/toxicity , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Osteoprotegerin/genetics , RANK Ligand/genetics , Random Allocation
11.
Sci Rep ; 8(1): 14515, 2018 09 28.
Article En | MEDLINE | ID: mdl-30266983

By using near-infrared fluorescent protein (iRFP)-expressing hematopoietic cells, we established a novel, quantitative, in vivo, noninvasive atherosclerosis imaging system. This murine atherosclerosis imaging approach targets macrophages expressing iRFP in plaques. Low-density lipoprotein receptor-deficient (LDLR-/-) mice transplanted with beta-actin promoter-derived iRFP transgenic (TG) mouse bone marrow (BM) cells (iRFP → LDLR-/-) were used. Atherosclerosis was induced by a nonfluorescent 1.25% cholesterol diet (HCD). Atherosclerosis was compared among the three differently induced mouse groups. iRFP → LDLR-/- mice fed a normal diet (ND) and LDLR-/- mice transplanted with wild-type (WT) BM cells were used as controls. The in vivo imaging system (IVIS) detected an enhanced iRFP signal in the thoracic aorta of HCD-fed iRFP → LDLR-/- mice, whereas iRFP signals were not observed in the control mice. Time-course imaging showed a gradual increase in the signal area, which was correlated with atherosclerotic plaque progression. Oil red O (ORO) staining of aortas and histological analysis of plaques confirmed that the detected signal was strictly emitted from plaque-positive areas of the aorta. Our new murine atherosclerosis imaging system can noninvasively image atherosclerotic plaques in the aorta and generate longitudinal data, validating the ability of the system to monitor lesion progression.


Aortic Diseases/diagnostic imaging , Atherosclerosis/diagnostic imaging , Luminescent Measurements/methods , Luminescent Proteins/analysis , Optical Imaging/methods , Plaque, Atherosclerotic/diagnostic imaging , Actins/genetics , Animals , Aortic Diseases/genetics , Atherosclerosis/etiology , Atherosclerosis/genetics , Azo Compounds , Bone Marrow Transplantation , Cholesterol, Dietary/toxicity , Coloring Agents , Flow Cytometry , Genes, Reporter , Genes, Synthetic , Luminescent Proteins/genetics , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/ultrastructure , Mice , Mice, Transgenic , Microscopy, Fluorescence , Promoter Regions, Genetic , Receptors, LDL/deficiency , Recombinant Proteins/analysis , Recombinant Proteins/genetics
12.
Biochem Biophys Res Commun ; 485(2): 550-555, 2017 04 01.
Article En | MEDLINE | ID: mdl-28088517

Although chronic kidney disease (CKD) is strongly associated with onsets of cardiovascular disease (CVD), the pathogenic mechanism between these diseases has not been fully understood. To develop and validate new therapeutic strategies for this complication, appropriate experimental models that reflect the complexity of the underlying pathophysiology are needed. The Osborne-Mendel (OM) rat was identified as an atherosclerosis-prone and a premature-death rat strain among 16 inbred rat strains when fed high-cholesterol containing diet. When fed high-cholesterol diet, OM rats showed simultaneous occurrence of aortic aneurysm, aortic dissection, peripheral artery occlusion, and left atrial thrombosis. OM rats had significantly lower max dP/dt and higher min dP/dt than F344 rats did, indicating impaired left ventricle contractility and relaxation. OM rats developed renal dysfunction, showing increased urinary albumin excretion. OM rats also showed mild hypertension, decreased endothelial function, and enhanced coagulation and platelet aggregation, compared with F344 rats. We now report that OM rat would be a novel spontaneous animal model which simultaneously demonstrates cardiac and renal dysfunction, and CVD events. This model could be a useful model for the pre-clinical testing of pharmacological therapies and could provide new insight into potential targets and pathways for the treatment of CKD and CVD.


Aortic Aneurysm/physiopathology , Arterial Occlusive Diseases/physiopathology , Heart Diseases/physiopathology , Kidney Diseases/physiopathology , Peripheral Arterial Disease/physiopathology , Thrombosis/physiopathology , Animals , Aortic Aneurysm/etiology , Arterial Occlusive Diseases/etiology , Blood Pressure/drug effects , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/toxicity , Diet, High-Fat/adverse effects , Disease Models, Animal , Heart Atria , Heart Diseases/etiology , Heart Rate/drug effects , Humans , Kidney Diseases/etiology , Male , Peripheral Arterial Disease/etiology , Rats, Inbred F344 , Rats, Inbred Strains , Species Specificity , Survival Analysis , Thrombosis/etiology , Time Factors
13.
Oxid Med Cell Longev ; 2016: 9895176, 2016.
Article En | MEDLINE | ID: mdl-27635189

Nonalcoholic steatohepatitis is one of the leading causes of liver disease. Dietary factors determine the clinical presentation of steatohepatitis and can influence the progression of related diseases. Cholesterol has emerged as a critical player in the disease and hence consumption of cholesterol-enriched diets can lead to a progressive form of the disease. The aim was to investigate the impact of liver cholesterol overload on the progression of the obstructive cholestasis in mice subjected to bile duct ligation surgery. Mice were fed with a high cholesterol diet for two days and then were subjected to surgery procedure; histological, biochemical, and molecular analyses were conducted to address the effect of cholesterol in liver damage. Mice under the diet were more susceptible to damage. Results show that cholesterol fed mice exhibited increased apoptosis and oxidative stress as well as reduction in cell proliferation. Mortality following surgery was higher in HC fed mice. Liver cholesterol impairs the repair of liver during obstructive cholestasis and aggravates the disease with early fatal consequences; these effects were strongly associated with oxidative stress.


Cholestasis/etiology , Cholesterol, Dietary/toxicity , Liver/pathology , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Bile Ducts/surgery , Bilirubin/analysis , Caspase 3/metabolism , Cholestasis/pathology , Cholesterol/analysis , Fatty Liver/etiology , Glutathione/analysis , Immunohistochemistry , Jaundice/etiology , Ki-67 Antigen/metabolism , Liver/drug effects , Liver/enzymology , Liver Function Tests , Mice , Mice, Inbred C57BL , Mortality, Premature , Reactive Oxygen Species/metabolism , Triglycerides/analysis
14.
Toxicology ; 361-362: 39-48, 2016 06 15.
Article En | MEDLINE | ID: mdl-27394961

Recent studies confirmed a critical importance of c-Met signaling for liver regeneration by modulating redox balance. Here we used liver-specific conditional knockout mice (MetKO) and a nutritional model of hepatic steatosis to address the role of c-Met in cholesterol-mediated liver toxicity. Liver injury was assessed by histopathology and plasma enzymes levels. Global transcriptomic changes were examined by gene expression microarray, and key molecules involved in liver damage and lipid homeostasis were evaluated by Western blotting. Loss of c-Met signaling amplified the extent of liver injury in MetKO mice fed with high-cholesterol diet for 30days as evidenced by upregulation of liver enzymes and increased synthesis of total bile acids, aggravated inflammatory response and enhanced intrahepatic lipid deposition. Global transcriptomic changes confirmed the enrichment of networks involved in steatosis and cholestasis. In addition, signaling pathways related to glutathione and lipid metabolism, oxidative stress and mitochondria dysfunction were significantly affected by the loss of c-Met function. Mechanistically, exacerbation of oxidative stress in MetKO livers was corroborated by increased lipid and protein oxidation. Western blot analysis further revealed suppression of Erk, NF-kB and Nrf2 survival pathways and downstream target genes (e.g. cyclin D1, SOD1, gamma-GCS), as well as up-regulation of proapoptotic signaling (e.g. p53, caspase 3). Consistent with the observed steatotic and cholestatic phenotype, nuclear receptors RAR, RXR showed increased activation while expression levels of CAR, FXR and PPAR-alpha were decreased in MetKO. Collectively, our data provide evidence for the critical involvement of c-Met signaling in cholesterol and bile acids toxicity.


Cholestasis, Intrahepatic/chemically induced , Cholestasis, Intrahepatic/metabolism , Hepatocytes/drug effects , Lipids/toxicity , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Animals , Cell Survival/drug effects , Cell Survival/genetics , Cholesterol, Dietary/toxicity , Glutathione/metabolism , Lipid Metabolism/drug effects , Lipid Peroxidation , Liver Function Tests , Mice , Mice, Knockout , Signal Transduction
15.
PLoS One ; 11(1): e0147469, 2016.
Article En | MEDLINE | ID: mdl-26807847

BACKGROUND: Pork is an essential component of the diet that has been linked with major degenerative diseases and development of non-alcoholic steatohepatitis (NASH). Previous studies have. Previous studies have demonstrated the in vitro antioxidant activity of silicon (Si). Furthermore, when Si is added to restructured pork (RP) strongly counterbalances the negative effect of high-cholesterol-ingestion, acting as an active hypocholesterolemic and hypolipemic dietary ingredient in aged rats. OBJECTIVE: This study was designed to evaluate the effects of Si vs hydroxytyrosol (HxT) RP on liver antioxidant defense in aged rats fed cholesterol-enriched high saturated/high cholesterol diets as a NASH model. METHODS: Four diets were prepared: Control RP diet (C) with non-added cholesterol; Cholesterol-enriched high-saturated/high-cholesterol control RP diet (CHOL-C) with added cholesterol and cholic acid; Si- or HxT-RP cholesterol-enriched high-saturated/high-cholesterol diets (CHOL-Si and CHOL-HxT). Groups of six male Wistar rats (1-yr old) were fed these modified diets for eight weeks. Total cholesterol, hepatosomatic index, liver Nrf2 and antioxidant (CAT, SOD, GSH, GSSG, GR, GPx) markers were determined. RESULTS: Both CHOL-Si and CHOL-HxT diets enhanced the liver antioxidant status, reduced hepatosomatic index and increased SOD actvity. Hydrogen peroxide removal seemed to be involved, explaining that the value of redox index was even lower than C without changing the CAT activity. CHOL-Si results were quite better than CHOL-HxT in most measured parameters. CONCLUSIONS: Our study suggests that Si incorporated into RP matrix was able to counterbalance, more efficiently than HxT, the deleterious effect of consuming a high-saturated/high-cholesterol diet, by improving the liver antioxidant defenses in the context of NASH.


Antioxidants/therapeutic use , Dietary Fats/toxicity , Liver/metabolism , Meat , Non-alcoholic Fatty Liver Disease/prevention & control , Phenylethyl Alcohol/analogs & derivatives , Silicon Dioxide/therapeutic use , Aging , Animal Feed/adverse effects , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacology , Body Weight/drug effects , Catalase/blood , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/toxicity , Cholic Acid/administration & dosage , Cholic Acid/toxicity , Colloids , Dietary Fats/administration & dosage , Drug Evaluation, Preclinical , Fatty Acids/administration & dosage , Fatty Acids/toxicity , Glutathione/blood , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/pharmacology , Hypolipidemic Agents/therapeutic use , Male , Meat/adverse effects , NF-E2-Related Factor 2/blood , Non-alcoholic Fatty Liver Disease/etiology , Oxidation-Reduction , Phenylethyl Alcohol/administration & dosage , Phenylethyl Alcohol/pharmacology , Phenylethyl Alcohol/therapeutic use , Rats , Rats, Wistar , Silicon Dioxide/administration & dosage , Silicon Dioxide/pharmacology , Superoxide Dismutase/blood , Sus scrofa , Swine
16.
PLoS One ; 10(10): e0141464, 2015.
Article En | MEDLINE | ID: mdl-26517374

Abnormal expression of thymic stromal lymphopoietin (TSLP) and its receptor (TSLPR) was found in patients with acute coronary syndrome. Ticagrelor, an oral platelet ADP P2Y12 receptor antagonist, is widely used in these patients. The aim of this study was to verify whether different doses of ticagrelor regulated plaque progression and platelet activity by modulating TSLP/TSLPR. Seventy-five ApoE-/- mice were randomly divided into five groups: (1) high-cholesterol diet (HCD, n = 15); (2) HCD plus ticagrelor 25 mg/kg/d (T1, n = 15); (3) HCD plus ticagrelor 50 mg/kg/d (T2, n = 15); (4) HCD plus ticagrelor 100 mg/kg/d (T3, n = 15); and (5) a normal diet group (ND, n = 15). At day 0 and at week 16, blood lipids and serum TSLP levels, expression of TSLPR, CD62, and CD63, platelet aggregation, platelet ATP release, PI3K/Akt signaling pathway, and plaque morphology were assessed. HCD increased TSLPR expression and atherosclerosis progression but high-dose ticagrelor (100 mg/kg) moderated this trend. TSLPR was positively correlated with Akt1, platelet aggregation, corrected plaque area, and vulnerability index in the T3 group (P<0.01). In conclusion, low-dose ticagrelor only inhibited platelet activity. Besides this inhibition, high-dose ticagrelor modulated platelet activity and atherosclerosis mediated by TSLPR, potentially through the PI3K/Akt signal pathway.


Adenosine/analogs & derivatives , Atherosclerosis/drug therapy , Cytokines/physiology , Immunoglobulins/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Cytokine/drug effects , Adenosine/administration & dosage , Adenosine/pharmacology , Animals , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Atherosclerosis/blood , Atherosclerosis/prevention & control , Cholesterol, Dietary/toxicity , Cytokines/blood , Dose-Response Relationship, Drug , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/complications , Hyperlipoproteinemia Type II/drug therapy , Immunoglobulins/biosynthesis , Immunoglobulins/physiology , Lipids/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/analysis , Phosphatidylinositol 3-Kinases/physiology , Platelet Aggregation Inhibitors/administration & dosage , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/physiology , Purinergic P2Y Receptor Antagonists/administration & dosage , Random Allocation , Receptors, Cytokine/biosynthesis , Receptors, Cytokine/physiology , Signal Transduction/drug effects , Tetraspanin 30/blood , Ticagrelor , Thymic Stromal Lymphopoietin
17.
Naunyn Schmiedebergs Arch Pharmacol ; 388(12): 1333-44, 2015 Dec.
Article En | MEDLINE | ID: mdl-26341793

Flavocoxid is a mixed extract containing baicalin and catechin, and it acts as a dual balanced inhibitor of cyclooxygenase-1 (COX-1) and COX-2 peroxidase enzyme activities with a significant inhibition of 5-lipoxygenase (5-LOX) enzyme activity in vitro. Flavocoxid downregulates gene or protein expression of several inflammatory markers and exerts also strong antioxidant activity in several experimental models. Inflammation and oxidative stress contribute in the pathogenesis of atherosclerosis. In the present study, an experimental rabbit model of hypercholesterolemia was developed and the effects of flavocoxid were evaluated. Rabbits were divided into four groups-normal control, high-cholesterol-diet (HCD)-fed group, HCD plus flavocoxid (20 mg/kg/day), or HCD plus atorvastatin (10 mg/kg/day). Blood samples were collected at the end of the experiment for measuring serum total cholesterol (TC), triglycerides (TGs), high-density lipoprotein cholesterol (HDL-C), C-reactive protein (CRP), malondialdehyde (MDA), reduced glutathione (GSH), and superoxide dismutase (SOD). In addition, the aorta was removed for measurement of antioxidant status, vascular reactivity, and intima/media (I/M) ratio. Elevated levels of serum TC, TGs, LDL-C, and CRP were measured in HCD group. Moreover, HCD caused a significant increase in serum and aortic MDA concomitantly with a reduction in serum and aortic GSH and SOD. Immunohistochemical staining of aortic specimens from HCD-fed rabbits revealed high expression levels of both tumor necrosis factor-alpha (TNF-α) and nuclear factor (NF)-κB. Rabbits in flavocoxid group showed significantly lower levels of serum CRP, serum, and aortic MDA and higher levels of serum HDL-C, serum, and aortic GSH and SOD compared to HCD group. HCD-induced elevations in serum TC and LDL-C did not significantly affected by flavocoxid treatment. Additionally, flavocoxid significantly enhanced rabbit aortic endothelium-dependent relaxation to acetylcholine and decreased the elevated I/M ratio. This effect was confirmed by histopathological examination of the aorta. Moreover, flavocoxid effectively suppresses the release of inflammatory markers. In conclusion, these findings demonstrated that flavocoxid would be useful in preventing oxidative stress, inflammation, and vascular dysfunction induced by HCD.


Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Catechin/therapeutic use , Cholesterol, Dietary/toxicity , Hypercholesterolemia/drug therapy , Animals , Cholesterol, Dietary/administration & dosage , Drug Combinations , Hypercholesterolemia/blood , Hypercholesterolemia/chemically induced , Male , Rabbits , Treatment Outcome
18.
Biomaterials ; 64: 125-135, 2015 Sep.
Article En | MEDLINE | ID: mdl-26111596

D-Threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (D-PDMP), a glycosphingolipid synthesis inhibitor, holds promise for the treatment of atherosclerosis and cardiac hypertrophy but rapid in vivo clearance has severely hindered translation to the clinic. To overcome this impediment, we used a materials-based delivery strategy wherein D-PDMP was encapsulated within a biodegradable polymer composed of poly ethylene glycol (PEG) and sebacic acid (SA). PEG-SA was formulated into nanoparticles that were doped with (125)I-labeled PEG to allow in vivo bio-distribution and release kinetics of D-PDMP to be determined by using γ-scintigraphy and subsequently, by mass spectrometry. Polymer-encapsulation increased the residence time of D-PDMP in the body of a treated mouse from less than one hour to at least four hours (and up to 48 h or longer). This substantially increased in vivo longevity provided by polymer encapsulation resulted in an order of magnitude gain in efficacy for interfering with atherosclerosis and cardiac hypertrophy in apoE-/- mice fed a high fat and high cholesterol (HFHC) diet. These results establish that D-PDMP encapsulated in a biodegradable polymer provides a superior mode of delivery compared to unconjugated D-PDMP by way of increased gastrointestinal absorption and increased residence time thus providing this otherwise rapidly cleared compound with therapeutic relevance in interfering with atherosclerosis, cardiac hypertrophy, and probably other diseases associated with the deleterious effects of abnormally high glycosphingolipid biosynthesis or deficient catabolism.


Atherosclerosis/drug therapy , Cardiomegaly/drug therapy , Morpholines/administration & dosage , Animals , Aortic Diseases/blood , Aortic Diseases/drug therapy , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/blood , Atherosclerosis/prevention & control , Capsules , Cardiomegaly/blood , Cholesterol, Dietary/toxicity , Decanoic Acids , Delayed-Action Preparations , Dicarboxylic Acids , Diet, Atherogenic , Drug Evaluation, Preclinical , Heart Ventricles/pathology , Inactivation, Metabolic , Iodine Radioisotopes/analysis , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Morpholines/pharmacokinetics , Nanoparticles/administration & dosage , Polyethylene Glycols , Tissue Distribution , Vascular Stiffness/drug effects
19.
Immunol Cell Biol ; 93(7): 653-61, 2015 Aug.
Article En | MEDLINE | ID: mdl-25748163

Inflammation is triggered after invasion or injury to restore homeostasis. Although the activation of Wnt/ß-catenin signaling is one of the first molecular responses to cellular damage, its role in inflammation is still unclear. It was our hypothesis that the low-density lipoprotein (LDL) receptor-related protein 5 (LRP5) and the canonical Wnt signaling pathway are modulators of inflammatory mechanisms. Wild-type (WT) and LRP5(-/-) mice were fed a hypercholesterolemic (HC) diet to trigger dislipidemia and chronic inflammation. Diets were supplemented with plant sterol esters (PSEs) to induce LDL cholesterol lowering and the reduction of inflammation. HC WT mice showed increased serum cholesterol levels that correlated with increased Lrp5 and Wnt/ß-catenin gene expression while in the HC LRP5(-/-) mice Wnt/ß-catenin pathway was shut down. Functionally, HC induced pro-inflammatory gene expression in LRP5(-/-) mice, suggesting an inhibitory role of the Wnt pathway in inflammation. Dietary PSE administration downregulated serum cholesterol levels in WT and LRP5(-/-) mice. Furthermore, in WT mice PSE increased anti-inflammatory genes expression and inhibited Wnt/ß-catenin activation. Hepatic gene expression of Vldlr, Lrp2 and Lrp6 was increased after HC feeding in WT mice but not in LRP5(-/-) mice, suggesting a role for these receptors in the clearance of plasmatic lipoproteins. Finally, an antiatherogenic role for LRP5 was demonstrated as HC LRP5(-/-) mice developed larger aortic atherosclerotic lesions than WT mice. Our results show an anti-inflammatory, pro-survival role for LRP5 and the Wnt signaling pathway in peripheral blood leukocytes.


Cholesterol/blood , Hypercholesterolemia/blood , Leukocytes/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Wnt Signaling Pathway , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/therapy , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/therapy , Cholesterol, Dietary/toxicity , Cholesterol, HDL/blood , Humans , Hypercholesterolemia/diet therapy , Jejunum/metabolism , Lipoproteins/biosynthesis , Lipoproteins/genetics , Liver/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/deficiency , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Phytosterols/therapeutic use , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Spleen/metabolism
20.
Climacteric ; 17 Suppl 2: 60-5, 2014 Dec.
Article En | MEDLINE | ID: mdl-25320023

Despite increased survivorship among patients, breast cancer remains the most common cancer among women and is the second leading cause of cancer death in women. The magnitude of this problem provides a strong impetus for new chemopreventative strategies and/or lifestyle changes that reduce cancer incidence. It is of significance, therefore, that several studies positively correlate obesity to the development of breast cancer. Importantly, obesity is also highly associated with elevated cholesterol, and cholesterol itself is a risk factor for breast cancer. Furthermore, patients taking statins demonstrate a lower breast cancer incidence and decreased recurrence. The recent observation that 27-hydroxycholesterol (27HC) is produced in a stoichiometric manner from cholesterol, together with our recent demonstration that it exerts partial agonist activity on both the estrogen and liver X receptors, suggested a potential mechanistic link between hyper-cholesterolemia and breast cancer incidence. Using genetic and pharmacological approaches, we have recently shown that elevation of circulating 27HC significantly increases tumor growth and metastasis in murine models of breast cancer. Further, we have demonstrated in appropriate animal models that the impact of high-fat diet on tumor pathogenesis can be mitigated by statins or by small molecule inhibitors of CYP27A1. These findings suggest that pharmacological or dietary modifications that lower total cholesterol, and by inference 27HC, are likely to reduce the impact of obesity/metabolic syndrome on breast cancer incidence.


Breast Neoplasms/etiology , Cholesterol, Dietary/toxicity , Receptors, Estrogen/metabolism , Animals , Autocrine Communication/immunology , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Cholesterol/blood , Cholesterol, Dietary/blood , Disease Models, Animal , Estrogen Receptor alpha/metabolism , Female , Humans , Hydroxycholesterols/blood , Hydroxycholesterols/chemical synthesis , Hyperlipidemias/complications , Hyperlipidemias/drug therapy , Incidence , Neoplasm Recurrence, Local , Obesity/complications , Paracrine Communication/immunology , Risk Factors , Selective Estrogen Receptor Modulators/metabolism
...