21 results on '"WD, Western diet"'
Search Results
2. Novel Göttingen Miniswine Model of Heart Failure With Preserved Ejection Fraction Integrating Multiple Comorbidities
- Author
-
Hunter A Hidalgo, Amy Scarborough, Zhen Li, Thomas E. Sharp, David J. Polhemus, J. Stephen Jenkins, Timothy Matsuura, Jeffery D. Schumacher, Daniel P. Kelly, David J. Lefer, and Traci T. Goodchild
- Subjects
heart failure with preserved ejection fraction ,0301 basic medicine ,obesity ,medicine.medical_specialty ,animal models of human disease ,hypertension ,HDL, high-density lipoprotein ,DBP, diastolic blood pressure ,HFpEF, heart failure with preserved ejection fraction ,Disease ,DOCA, 11-deoxycorticosterone acetate ,030204 cardiovascular system & hematology ,Novel Translational Models ,metabolic syndrome ,03 medical and health sciences ,PCWP - Pulmonary capillary wedge pressure ,0302 clinical medicine ,LDL, low-density lipoprotein ,Internal medicine ,Western diet ,medicine ,EF, ejection fraction ,HFrEF, heart failure with reduced ejection fraction ,WD, Western diet ,business.industry ,SBP, systolic blood pressure ,DBP - Diastolic blood pressure ,medicine.disease ,PCWP, pulmonary capillary wedge pressure ,TC, total cholesterol ,LV, left ventricle ,030104 developmental biology ,Heart failure ,IVGTT, intravenous glucose tolerance test ,Cardiology ,EC50, half-maximal effective concentration ,Metabolic syndrome ,Cardiology and Cardiovascular Medicine ,Heart failure with preserved ejection fraction ,business ,Large animal - Abstract
Visual Abstract, Highlights • A large animal model that recapitulates the clinical heterogeneity of HFpEF has been developed. • Multiple comorbidities, including obesity, hypercholesterolemia, pre-diabetic phenotype, as well as pulmonary and systemic arterial hypertension, were induced in adult female Göttingen minipigs by mineralocorticoid excess and a diet high in cholesterol, fat, fructose, and salt. • Severity of each comorbidity was titrated, such that marked left ventricular and left atrial diastolic dysfunction, profound left ventricular concentric remodeling, impaired coronary vasorelaxation, and extensive multiorgan fibrosis were achieved over a relatively short time. • This novel heart failure model was generated in Göttingen minipigs to allow for translational studies of novel pharmacological agents and for human-sized device testing in adult-aged animals. • This novel “multihit” minipig heart failure model enabled investigations into the pathology of HFpEF without the confounding effects inherent with invasive surgical procedures or the administration of agents that induce hypertension and end-organ damage in an artificial manner., Summary A lack of preclinical large animal models of heart failure with preserved ejection fraction (HFpEF) that recapitulate this comorbid-laden syndrome has led to the inability to tease out mechanistic insights and to test novel therapeutic strategies. This study developed a large animal model that integrated multiple comorbid determinants of HFpEF in a miniswine breed that exhibited sensitivity to obesity, metabolic syndrome, and vascular disease with overt clinical signs of heart failure. The combination of a Western diet and 11-deoxycorticosterone acetate salt−induced hypertension in the Göttingen miniswine led to the development of a novel large animal model of HFpEF that exhibited multiorgan involvement and a full spectrum of comorbidities associated with human HFpEF.
- Published
- 2021
- Full Text
- View/download PDF
3. Sestrin Proteins Protect Against Lipotoxicity-Induced Oxidative Stress in the Liver via Suppression of C-Jun N-Terminal KinasesSummary
- Author
-
Menghao Huang, Kushan Chowdhury, Zhigang Fang, X. Charlie Dong, Ming O. Li, Suthat Liangpunsakul, and Hyeong-Geug Kim
- Subjects
0301 basic medicine ,Sestrins ,MPO, myeloperoxidase ,Mitogen-Activated Protein Kinase Kinase 7 ,RC799-869 ,DMSO, dimethyl sulfoxide ,FoxO, forkhead box O ,Sesn1/2/3, sestrin1/2/3 ,medicine.disease_cause ,TKO, triple knockout ,Mice ,PCR, polymerase chain reaction ,0302 clinical medicine ,GSH, glutathione ,Phosphorylation ,NF-κB, nuclear factor kappa B ,Original Research ,Cat, catalase ,Mice, Knockout ,Sestrin ,Gpx3, glutathione peroxidase 3 ,Il6, interleukin 6 ,WD, Western diet ,biology ,Chemistry ,Kinase ,Fatty liver ,Tgfbr1, transforming growth factor beta receptor 1 ,Gastroenterology ,Col1a1, collagen type 1 alpha 1 ,Nox1, NADPH oxidase 1 ,Diseases of the digestive system. Gastroenterology ,Dgat2, diacylglycerol O-acyltransferase 2 ,Cell biology ,Keap1, Kelch-like ECH-associated protein 1 ,C-Jun N-Terminal Kinase ,Liver ,Lipotoxicity ,c-Jun N-terminal kinases ,JNK, c-Jun N-terminal kinase ,030211 gastroenterology & hepatology ,Disease Susceptibility ,Signal transduction ,NASH, nonalcoholic steatohepatitis ,Gss, glutathione synthetase ,Pdgfrb, platelet-derived growth factor receptor beta ,Timp1, tissue inhibitor of metalloproteinase 1 ,Fasn, fatty acid synthase ,Fatty Liver Disease ,Sod, superoxide dismutase ,Acta2, smooth muscle actin alpha 2 ,ER, endoplasmic reticulum ,PA, palmitic acid ,03 medical and health sciences ,4-HNE, 4-hydroxynonenal ,MKK, mitogen-activated protein kinase kinase ,medicine ,Animals ,Humans ,Srebp, sterol regulatory element-binding protein ,Protein kinase A ,KD, knockdown ,MDA, malondialdehyde ,Inflammation ,KO, knockout ,Hepatology ,NRF2, nuclear factor erythroid 2-related like 2 ,JNK Mitogen-Activated Protein Kinases ,Lipid Metabolism ,medicine.disease ,Tgfb1, transforming growth factor beta 1 ,WT, wild-type ,mTORC, mechanistic target of rapamycin complex ,Fatty Liver ,AMPK, AMP-activated protein kinase ,Disease Models, Animal ,Oxidative Stress ,Scd1, stearoyl-CoA desaturase 1 ,030104 developmental biology ,Gene Expression Regulation ,Cytoprotection ,Hepatocytes ,biology.protein ,BSA, bovine serum albumin ,SD, standard deviation ,DHE, dihydroethidium ,PERK, protein kinase R-like endoplasmic reticulum kinase ,Tnf, tumor necrosis factor ,Biomarkers ,Oxidative stress - Abstract
Background & Aims Sestrin 1/2/3 (Sesn1/2/3) belong to a small family of proteins that have been implicated in the regulation of metabolic homeostasis and oxidative stress. However, the underlying mechanisms remain incompletely understood. The aim of this work was to illustrate the collective function of Sesn1/2/3 in the protection against hepatic lipotoxicity. Methods We used Sesn1/2/3 triple knockout (TKO) mouse and cell models to characterize oxidative stress and signal transduction under lipotoxic conditions. Biochemical, histologic, and physiological approaches were applied to illustrate the related processes. Results After feeding with a Western diet for 8 weeks, TKO mice developed remarkable metabolic associated fatty liver disease that was manifested by exacerbated hepatic steatosis, inflammation, and fibrosis compared with wild-type counterparts. Moreover, TKO mice exhibited higher levels of hepatic lipotoxicity and oxidative stress. Our biochemical data revealed a critical signaling node from sestrins to c-Jun N-terminal kinases (JNKs) in that sestrins interact with JNKs and mitogen-activated protein kinase kinase 7 and suppress the JNK phosphorylation and activity. In doing so, sestrins markedly reduced palmitate-induced lipotoxicity and oxidative stress in both mouse and human hepatocytes. Conclusions The data from this study suggest that Sesn1/2/3 play an important role in the protection against lipotoxicity-associated oxidative stress and related pathology in the liver., Graphical abstract
- Published
- 2021
4. Nonalcoholic Steatohepatitis and HCC in a Hyperphagic Mouse Accelerated by Western Diet
- Author
-
Anthony M. Diomino, David A. Brenner, Hyeok Choon Kwon, Gibraan Rahman, Sara Brin Rosenthal, Tatiana Kisseleva, Linshan Shang, Souradipta Ganguly, Ruoyu Wang, Debanjan Dhar, Rob Knight, Pejman Soorosh, Mojgan Hosseini, Yanhan Wang, Bernd Schnabl, and German R. Aleman Muench
- Subjects
Cirrhosis ,medicine.medical_treatment ,Cell Cycle Proteins ,RC799-869 ,Gastroenterology ,Mice ,0302 clinical medicine ,Fibrosis ,Aetiology ,Original Research ,Cancer ,Liver Disease ,Liver Neoplasms ,ILC, innate lymphoid cell ,Diseases of the digestive system. Gastroenterology ,Immunohistochemistry ,HSC, hepatic stellate cell ,DSI, distal small intestine ,Editorial ,FACS, fluorescence-activated cell sorter ,030211 gastroenterology & hepatology ,FITC, fluorescein isothiocyanate ,NASH, nonalcoholic steatohepatitis ,Western ,IHC, immunohistochemistry ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Knockout ,CVD, cardiovascular disease ,Hyperphagia ,digestive system ,03 medical and health sciences ,Humans ,RPCA, robust principal component analysis ,Dyslipidemias ,Animal ,nutritional and metabolic diseases ,Hepatocellular ,PE, Phycoerythrin ,Hepatocellular Carcinoma ,Gene signature ,medicine.disease ,WT, wild-type ,digestive system diseases ,IL, interleukin ,LBP, lipopolysaccharide binding protein ,030104 developmental biology ,NAFLD, nonalcoholic fatty liver disease ,MIP2, Macrophage Inflammatory Protein 2 ,Digestive Diseases ,Biomarkers ,Liver Inflammation ,Liver Cirrhosis ,0301 basic medicine ,Chemokine ,CXCL2, C-X-C motif chemokine ligand 2 ,Adipose tissue ,Nonalcoholic Steatohepatitis ,Gut Inflammation ,Oral and gastrointestinal ,Hepatitis ,Risk Factors ,Non-alcoholic Fatty Liver Disease ,2.1 Biological and endogenous factors ,Mice, Knockout ,TNF, tumor necrosis factor ,WD, Western diet ,biology ,rRNA, ribosomal RNA ,Cytokine ,Hepatocellular carcinoma ,LPS, lipopolysaccharide ,Disease Susceptibility ,medicine.symptom ,eWAT, white adipose tissue (epididymal fat) ,NASH Regression ,Biotechnology ,Liver Cancer ,APC, Allophycocyanin ,Chronic Liver Disease and Cirrhosis ,PBS, phosphate-buffered saline ,Inflammation ,CD-HFD, choline-deficient high-fat diet ,SI, small intestine ,Rare Diseases ,FBS, fetal bovine serum ,ALT, alanine aminotransferase ,Internal medicine ,medicine ,Animals ,IFN, interferon ,Obesity ,Nutrition ,Hepatology ,business.industry ,Gene Expression Profiling ,CKD, chronic kidney disease ,Carcinoma ,qRT-PCR, quantitative reverse-transcription polymerase chain reaction ,Diet ,Disease Models, Animal ,Good Health and Well Being ,Diet, Western ,Disease Models ,biology.protein ,Insulin Resistance ,HCC, hepatocellular carcinoma ,business - Abstract
Background & Aims How benign liver steatosis progresses to nonalcoholic steatohepatitis (NASH), fibrosis, and hepatocellular carcinoma (HCC) remains elusive. NASH progression entails diverse pathogenic mechanisms and relies on complex cross-talk between multiple tissues such as the gut, adipose tissues, liver, and the brain. Using a hyperphagic mouse fed with a Western diet (WD), we aimed to elucidate the cross-talk and kinetics of hepatic and extrahepatic alterations during NASH–HCC progression, as well as regression. Methods Hyperphagic mice lacking a functional Alms1 gene (Foz/Foz) and wild-type littermates were fed WD or standard chow for 12 weeks for NASH/fibrosis and for 24 weeks for HCC development. NASH regression was modeled by switching back to normal chow after NASH development. Results Foz+WD mice were steatotic within 1 to 2 weeks, developed NASH by 4 weeks, and grade 3 fibrosis by 12 weeks, accompanied by chronic kidney injury. Foz+WD mice that continued on WD progressed to cirrhosis and HCC within 24 weeks and had reduced survival as a result of cardiac dysfunction. However, NASH mice that were switched to normal chow showed NASH regression, improved survival, and did not develop HCC. Transcriptomic and histologic analyses of Foz/Foz NASH liver showed strong concordance with human NASH. NASH was preceded by an early disruption of gut barrier, microbial dysbiosis, lipopolysaccharide leakage, and intestinal inflammation. This led to acute-phase liver inflammation in Foz+WD mice, characterized by neutrophil infiltration and increased levels of several chemokines/cytokines. The liver cytokine/chemokine profile evolved as NASH progressed, with subsequent predominance by monocyte recruitment. Conclusions The Foz+WD model closely mimics the pathobiology and gene signature of human NASH with fibrosis and subsequent HCC. Foz+WD mice provide a robust and relevant preclinical model of NASH, NASH-associated HCC, chronic kidney injury, and heart failure., Supplemental Graphical Summary
- Published
- 2021
- Full Text
- View/download PDF
5. Antisense oligonucleotide–mediated inhibition of angiopoietin-like protein 3 increases reverse cholesterol transport in mice
- Author
-
Aaron J. Donner, Rosanne M. Crooke, Adam E. Mullick, Richard G. Lee, Mingxia Liu, and Thomas A. Bell
- Subjects
Endothelial lipase ,antisense oligonucleotide ,EL, endothelial lipase ,Biochemistry ,chemistry.chemical_compound ,Mice ,Endocrinology ,ANGPTL3 ,cardiovascular disease ,RCT, reverse cholesterol transport ,Hyperlipidemia ,hyperlipidemia ,LOF, loss-of-function ,chemistry.chemical_classification ,Mice, Knockout ,WD, Western diet ,Reverse cholesterol transport ,tg, transgenic ,lipids (amino acids, peptides, and proteins) ,ASCVD, atherosclerotic cardiovascular disease ,coronary artery disease ,Research Article ,medicine.medical_specialty ,HDL ,Transgene ,ASO, antisense oligonucleotide ,QD415-436 ,ANGPTL3, angiopoietin-like protein 3 ,TPC, total plasma cholesterol ,lipids ,Internal medicine ,medicine ,Animals ,Humans ,Loss function ,Angiopoietin-Like Protein 3 ,LSC, liquid scintillation counting ,3H-CHE, 3H-cholesteryl hexadecyl ether ,FCR, fractional catabolic rate ,Cholesterol ,Cholesterol, HDL ,cholesterol ,Biological Transport ,Cell Biology ,FHBL2, familial combined hypobetalipoproteinemia ,Oligonucleotides, Antisense ,medicine.disease ,reverse cholesterol transport ,Mice, Inbred C57BL ,Enzyme ,chemistry ,atherosclerosis - Abstract
Supported by an abundance of experimental and genetic evidence, angiopoietin-like protein 3 (ANGPTL3) has emerged as a promising therapeutic target for cardiovascular disease. ANGPTL3 is primarily produced by the liver and is a potent modulator of plasma lipids and lipoproteins. Experimental models and subjects with loss-of-function Angptl3 mutations typically present with lower levels of HDL-C than noncarriers. The effect of ANGPTL3 on HDL-C is typically attributed to its function as an inhibitor of the enzyme endothelial lipase. The ability to facilitate reverse cholesterol transport (RCT), the transport of cholesterol from peripheral tissues back to the liver, is a proposed antiatherogenic property of HDL. However, the effect of ANGPTL3 inhibition on RCT remains unclear. Here, we performed a series of dose-response and RCT studies using an Angptl3 antisense oligonucleotide (ASO) in mouse models with varying plasma lipid profiles ranging from moderately to severely hyperlipidemic. Angptl3 ASO-mediated reduction in HDL-C was limited to the model with moderate lipidemia, where the majority of plasma cholesterol was associated with HDL. Surprisingly, regardless of the effect on HDL-C, treatment with the Angptl3 ASO enhanced RCT in all models tested. The observations from the RCT assays were confirmed in HDL clearance studies, where mice treated with the Angptl3 ASO displayed increased plasma clearance and hepatic uptake of labeled HDL. The results from our studies suggest that inhibition of ANGPTL3 not only reduces levels of proatherogenic lipids but also improves HDL-mediated RCT.
- Published
- 2021
6. Making the Best of a Competition: the CREB3L3–SREBP Axis in Arteriosclerosis
- Author
-
Tirthadipa Pradhan-Sundd
- Subjects
Male ,Arteriosclerosis ,SREBP, sterol regulatory element-binding protein ,CREB3L3 ,FGF21, fibroblast growth factor 21 ,Bioinformatics ,CREB3L3, cAMP responsive element-binding protein 3 like 3 ,Mice ,Enterohepatic Circulation ,Cyclic AMP Response Element-Binding Protein ,Insig, insulin-induced gene ,Original Research ,GFP, green fluorescent protein ,Mice, Knockout ,Sterol Regulatory Element Binding Proteins ,TBA, total bile acid ,WD, Western diet ,TG, triglyceride ,Chemistry ,Gastroenterology ,Editorial ,Hyperlipidemia ,LPL, lipoprotein lipase ,Female ,lipids (amino acids, peptides, and proteins) ,Sterol Regulatory Element Binding Protein 1 ,HDL, high-density lipoprotein ,Hyperlipidemias ,SREBP ,ER, endoplasmic reticulum ,Competition (economics) ,Tg, transgenic ,FFA, free fatty acid ,medicine ,Animals ,Humans ,lcsh:RC799-869 ,KO, knockout ,PPARα, peroxisome proliferator-activated receptor alpha ,Hepatology ,PLA, proximity ligation assay ,S1P, site-1 protease ,Lipogenesis ,SREBF, sterol regulatory element-binding factor ,VLDL, very-low-density lipoprotein ,Atherosclerosis ,Lipid Metabolism ,medicine.disease ,Sterol regulatory element-binding protein ,Mice, Inbred C57BL ,TC, total cholesterol ,Gene Expression Regulation ,Receptors, LDL ,Apo, apolipoprotein ,HPLC, high-performance liquid chromatography ,HSV, herpes simplex virus ,lcsh:Diseases of the digestive system. Gastroenterology ,LXR, liver X receptor - Abstract
Background & Aims cAMP responsive element-binding protein 3 like 3 (CREB3L3) is a membrane-bound transcription factor involved in the maintenance of lipid metabolism in the liver and small intestine. CREB3L3 controls hepatic triglyceride and glucose metabolism by activating plasma fibroblast growth factor 21 (FGF21) and lipoprotein lipase. In this study, we intended to clarify its effect on atherosclerosis. Methods CREB3L3-deficifient, liver-specific CREB3L3 knockout, intestine-specific CREB3L3 knockout, both liver- and intestine-specific CREB3L3 knockout, and liver CREB3L3 transgenic mice were crossed with LDLR−/− mice. These mice were fed with a Western diet to develop atherosclerosis. Results CREB3L3 ablation in LDLR−/− mice exacerbated hyperlipidemia with accumulation of remnant APOB-containing lipoprotein. This led to the development of enhanced aortic atheroma formation, the extent of which was additive between liver- and intestine-specific deletion. Conversely, hepatic nuclear CREB3L3 overexpression markedly suppressed atherosclerosis with amelioration of hyperlipidemia. CREB3L3 directly up-regulates anti-atherogenic FGF21 and APOA4. In contrast, it antagonizes hepatic sterol regulatory element-binding protein (SREBP)–mediated lipogenic and cholesterogenic genes and regulates intestinal liver X receptor–regulated genes involved in the transport of cholesterol. CREB3L3 deficiency results in the accumulation of nuclear SREBP proteins. Because both transcriptional factors share the cleavage system for nuclear transactivation, full-length CREB3L3 and SREBPs in the endoplasmic reticulum (ER) functionally inhibit each other. CREB3L3 promotes the formation of the SREBP-insulin induced gene 1 complex to suppress SREBPs for ER-Golgi transport, resulting in ER retention and inhibition of proteolytic activation at the Golgi and vice versa. Conclusions CREB3L3 has multi-potent protective effects against atherosclerosis owing to new mechanistic interaction between CREB3L3 and SREBPs under atherogenic conditions., Graphical abstract
- Published
- 2021
- Full Text
- View/download PDF
7. SIRT6 Protects Against Liver Fibrosis by Deacetylation and Suppression of SMAD3 in Hepatic Stellate CellsSummary
- Author
-
Romil Saxena, Wenjie Cai, Yang Zhang, Menghao Huang, Xiaolin Zhong, Kushan Chowdhury, Robert F. Schwabe, Suthat Liangpunsakul, Hyeong-Geug Kim, and X. Charlie Dong
- Subjects
Liver Cirrhosis ,0301 basic medicine ,Steatosis ,Nonalcoholic Steatohepatitis ,Pathogenesis ,PCR, polymerase chain reaction ,0302 clinical medicine ,SMAD3, SMAD family member 3 ,Fibrosis ,Sirtuin 6 ,Sirtuins ,Original Research ,GFP, green fluorescent protein ,WD, Western diet ,Fatty liver ,Gastroenterology ,SIRT6, sirtuin 6 ,HSC, hepatic stellate cell ,mRNA, messenger RNA ,ChIP, chromatin immunoprecipitation ,Sirtuin ,shRNA, short hairpin RNA ,TGFB, transforming growth factor β ,030211 gastroenterology & hepatology ,Lrat, lecithin retinol acyltransferase ,medicine.symptom ,NASH, nonalcoholic steatohepatitis ,SIRT6 ,Deacetylation ,PBS, phosphate-buffered saline ,Inflammation ,Biology ,03 medical and health sciences ,ALT, alanine aminotransferase ,Tg, transgenic ,Hepatic Stellate Cells ,medicine ,Humans ,lcsh:RC799-869 ,KO, knockout ,Hepatology ,medicine.disease ,WT, wild-type ,digestive system diseases ,030104 developmental biology ,Cancer research ,biology.protein ,Hepatic stellate cell ,DMEM, Dulbecco’s modified Eagle medium ,lcsh:Diseases of the digestive system. Gastroenterology ,NAFLD, nonalcoholic fatty liver disease ,HA, hemagglutinin ,Transforming growth factor - Abstract
Background & Aims Nonalcoholic steatohepatitis (NASH) is a chronic liver disease that is manifested clinically by an increase in hepatic triglycerides, inflammation, and fibrosis. The pathogenesis of NASH remains incompletely understood. Sirtuin 6 (Sirt6), a nicotinamide adenine dinucleotide–dependent deacetylase, has been implicated in fatty liver disease; however, the underlying molecular mechanisms in the NASH pathogenesis are elusive. The aims of this study were to elucidate the role of hepatic Sirt6 in NASH. Methods Wild-type, liver-specific Sirt6 knockout (KO), hepatic stellate cell (HSC)-specific Sirt6 knockout (HSC-KO), and Sirt6 transgenic mice were subjected to a Western diet for 4 weeks. Hepatic phenotypes were characterized and underlying mechanisms were investigated. Results Remarkably, both the liver-KO and HSC-KO mice developed much worse NASH than the wild-type mice, whereas the transgenic mice were protected from the diet-induced NASH. Our cell signaling analysis showed that Sirt6 negatively regulates the transforming growth factor β–Smad family member 3 (Smad3) pathway. Biochemical analysis showed a physical interaction between Sirt6 and Smad3 in hepatic stellate cells. Moreover, our molecular data further showed that Sirt6 deacetylated Smad3 at key lysine residues K333 and K378, and attenuated its transcriptional activity induced by transforming growth factor β in hepatic stellate cells. Conclusions Our data suggest that SIRT6 plays a critical role in the protection against NASH development and it may serve as a potential therapeutic target for NASH., Graphical abstract
- Published
- 2020
8. miR-22 inhibition reduces hepatic steatosis via FGF21 and FGFR1 induction
- Author
-
Yu-Jui Yvonne Wan, Mohamed R. Ali, Prasant Kuma Jena, Ying Hu, Lili Sheng, and Hui Xin Liu
- Subjects
FGF21 ,Steatosis ,insulin tolerance test ,ALT ,ITT ,HDAC, histone deacetylase ,PFUs ,Pharmacology ,PPREs ,sirtuin 1 ,0302 clinical medicine ,obeticholic acid ,HDAC ,Aetiology ,PHHs ,Western diet ,RARβ, retinoic acid receptor β ,0303 health sciences ,Liver Disease ,Gastroenterology ,3. Good health ,FXR ,030220 oncology & carcinogenesis ,alcoholic steatosis ,CD, control diet ,PPREs, peroxisome proliferative-response elements ,non-alcoholic steatohepatitis ,PHHs, primary human hepatocytes ,RT-PCR ,03 medical and health sciences ,peroxisome proliferative-response elements ,FXR, farnesoid X receptor ,SIRT1 ,OCA ,NPCs, non-parenchymal cells ,ALP, alkaline phosphatase ,RT-PCR, reverse transcription PCR ,medicine.disease ,3' untranslated region ,PFUs, plaque-forming units ,primary human hepatocytes ,chemistry ,histone deacetylase ,Metabolic syndrome ,Digestive Diseases ,GLP-1 ,farnesoid X receptor ,retinoic acid receptor β ,FGF21, fibroblast growth factor 21 ,chemistry.chemical_compound ,non-parenchymal cells ,reverse transcription PCR ,2.1 Biological and endogenous factors ,Immunology and Allergy ,WD, Western diet ,Diabetes ,Fatty liver ,lipopolysaccharide ,Obeticholic acid ,NPCs ,CD ,OCA, obeticholic acid ,PGC1α, PPAR-activated receptor-γ coactivator-1α ,plaque-forming units ,LPS, lipopolysaccharide ,alkaline phosphatase ,Research Article ,Biotechnology ,LPS ,SIRT1, sirtuin 1 ,glucagon-like peptide ,alanine aminotransferase ,Chronic Liver Disease and Cirrhosis ,fibroblast growth factor 21 ,PPAR-activated receptor-γ coactivator-1α ,metabolic syndrome ,Insulin resistance ,WD ,ALT, alanine aminotransferase ,PGC1α ,GLP-1, glucagon-like peptide ,Internal Medicine ,medicine ,insulin sensitivity ,lcsh:RC799-869 ,Metabolic and endocrine ,ITT, insulin tolerance test ,Nutrition ,030304 developmental biology ,RARβ ,Hepatology ,3'-UTR, 3' untranslated region ,AMPK ,control diet ,ALP ,Farnesoid X receptor ,lcsh:Diseases of the digestive system. Gastroenterology ,3'-UTR - Abstract
Background & Aims Metabolism supports cell proliferation and growth. Surprisingly, the tumor suppressor miR-22 is induced by metabolic stimulators like bile acids. Thus, this study examines whether miR-22 could be a metabolic silencer. Methods The relationship between miR-22 and the expression of fibroblast growth factor 21 (FGF21) and its receptor FGFR1 was studied in cells and fatty livers obtained from patients and mouse models. We evaluated the effect of an miR-22 inhibitor alone and in combination with obeticholic acid (OCA) for the treatment of steatosis. Results The levels of miR-22 were inversely correlated with those of FGF21, FGFR1, and PGC1α in human and mouse fatty livers, suggesting that hepatic miR-22 acts as a metabolic silencer. Indeed, miR-22 reduced FGFR1 by direct targeting and decreased FGF21 by reducing the recruitment of PPARα and PGC1α to their binding motifs. In contrast, an miR-22 inhibitor increases hepatic FGF21 and FGFR1, leading to AMPK and ERK1/2 activation, which was effective in treating alcoholic steatosis in mouse models. The farnesoid x receptor-agonist OCA induced FGF21 and FGFR1, as well as their inhibitor miR-22. An miR-22 inhibitor and OCA were effective in treating diet-induced steatosis, both alone and in combination. The combined treatment was the most effective at improving insulin sensitivity, releasing glucagon-like peptide 1, and reducing hepatic triglyceride in obese mice. Conclusion The simultaneous induction of miR-22, FGF21 and FGFR1 by metabolic stimulators may maintain FGF21 homeostasis and restrict ERK1/2 activation. Reducing miR-22 enhances hepatic FGF21 and activates AMPK, which could be a novel approach to treat steatosis and insulin resistance. Lay summary This study examines the metabolic role of a tumor suppressor, miR-22, that can be induced by metabolic stimulators such as bile acids. Our novel data revealed that the metabolic silencing effect of miR-22 occurs as a result of reductions in metabolic stimulators, which likely contribute to the development of fatty liver. Consistent with this finding, an miR-22 inhibitor effectively reversed both alcohol- and diet-induced fatty liver; miR-22 inhibition is a promising therapeutic option which could be used in combination with obeticholic acid., Graphical abstract, Highlights • Human and mouse fatty livers have elevated miR-22, but reduced FGF21, FGFR1, and PGC1α. • FGFR1 is a novel target of miR-22. • MiR-22 inhibits FGF21 expression by reducing recruitment of PPARα and PGC1α to their binding motifs. • Hepatic miR-22 silencing could be a novel approach to treat metabolic diseases including steatosis. • miR-22 inhibitor improves the efficacy of FGF21 activators such as obeticholic acid.
- Published
- 2020
9. The GLP-1 Analogs Liraglutide and Semaglutide Reduce Atherosclerosis in ApoE−/− and LDLr−/− Mice by a Mechanism That Includes Inflammatory Pathways
- Author
-
Robert Augustin, Lotte Bjerre Knudsen, Klaus Stensgaard Frederiksen, Jane Nøhr, Ib Klewe, Joseph Polex-Wolf, Günaj Rakipovski, Bidda Rolin, Camilla Ingvorsen, and Jacob Hecksher-Sørensen
- Subjects
Apolipoprotein E ,lcsh:Diseases of the circulatory (Cardiovascular) system ,obesity ,030209 endocrinology & metabolism ,Inflammation ,Type 2 diabetes ,030204 cardiovascular system & hematology ,Pharmacology ,Systemic inflammation ,03 medical and health sciences ,PRECLINICAL RESEARCH ,0302 clinical medicine ,Diabetes mellitus ,LDL, low-density lipoprotein ,TIMP, tissue inhibitor of metalloproteinases ,medicine ,IFN, interferon ,TNF, tumor necrosis factor ,WD, Western diet ,diabetes ,GLP, glucagon-like peptide ,business.industry ,Liraglutide ,Semaglutide ,medicine.disease ,IL, interleukin ,MMP, matrix metalloproteinase ,lcsh:RC666-701 ,inflammation ,LDL receptor ,CD163, cluster of differentiation 163 molecule ,OPN, osteopontin ,RNA, ribonucleic acid ,LPS, lipopolysaccharide ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,atherosclerosis ,Cardiology and Cardiovascular Medicine ,business ,GLP-1 ,NASH, nonalcoholic steatohepatitis ,medicine.drug - Abstract
Visual Abstract, Highlights • The GLP-1RAs liraglutide and semaglutide reduce cardiovascular risk in type 2 diabetes patients. • In ApoE−/− mice and LDLr−/− mice, liraglutide and semaglutide treatment significantly attenuated plaque lesion development, in part independently of body weight and cholesterol lowering. • Semaglutide decreased levels of plasma markers of systemic inflammation in an acute inflammation model (lipopolysaccharide), and transcriptomic analysis of aortic atherosclerotic tissue revealed that multiple inflammatory pathways were down-regulated by semaglutide., Summary The glucagon-like peptide-1 receptor agonists (GLP-1RAs) liraglutide and semaglutide reduce cardiovascular risk in type 2 diabetes patients. The mode of action is suggested to occur through modified atherosclerotic progression. In this study, both of the compounds significantly attenuated plaque lesion development in apolipoprotein E-deficient (ApoE−/−) mice and low-density lipoprotein receptor-deficient (LDLr−/−) mice. This attenuation was partly independent of weight and cholesterol lowering. In aortic tissue, exposure to a Western diet alters expression of genes in pathways relevant to the pathogenesis of atherosclerosis, including leukocyte recruitment, leukocyte rolling, adhesion/extravasation, cholesterol metabolism, lipid-mediated signaling, extracellular matrix protein turnover, and plaque hemorrhage. Treatment with semaglutide significantly reversed these changes. These data suggest GLP-1RAs affect atherosclerosis through an anti-inflammatory mechanism.
- Published
- 2018
10. Can healthy lifestyle reduce disease progression of Alzheimer’s during a global pandemic of COVID-19?
- Author
-
Kiran Ali, P. Hemachandra Reddy, Albin John, and Harrison Marsh
- Subjects
COX, cytochrome c oxidase activity ,Aging ,HIF-α, hypoxia inducible factor-Iα ,PS2, presenilin 2 ,Review ,Aβ, amyloid beta ,Disease ,Biochemistry ,Diabetes mellitus ,RAAS, renin-angiotensin-aldosterone system ,ROS, reactive oxidative species ,Pandemic ,Vector (molecular biology) ,TMPRSS2, transmembrane protease serine 2 ,APP, amyloid beta precursor protein ,TNF-α, tumor necrosis factor α ,COVID-19, coronavirus disease 2019 ,PS1, presenilin 1 ,WD, Western diet ,ACE2, angiotensin converting enzyme 2 ,MFRTA, mitochondrial free radical theory of aging ,Neurology ,MCI, mild cognitive impairment ,Hypertension ,Disease Progression ,APOE4, apolipoprotein E4 genotype ,ORF, open-reading frames ,SCD, sickle cell disease ,Alzheimer's disease ,Alzheimer’s disease ,PARP, poly-ADP-ribose polymerase ,Biotechnology ,medicine.medical_specialty ,T2D, type 2 diabetes ,AD, Alzheimer’s disease ,BMI, body metabolic index ,CNS, central nervous system ,SARS-CoV-2, severe acute respiratory syndrome coronavirus 2 ,CypD, cyclophilin D ,MitomiRs, mitochondrial microRNAs ,MtDNA, mitochondrial DNA ,NPIQ, neuropsychiatric Inventory-Questionnaire ,Alzheimer Disease ,medicine ,Humans ,Dementia ,IFN, interferon ,Obesity ,Healthy Lifestyle ,Intensive care medicine ,MHV, mouse hepatitis virus ,Pandemics ,Molecular Biology ,Aged ,NO, nitric oxide ,SARS-CoV-2 ,business.industry ,Amyloid beta ,COVID-19 ,Lifestyle ,medicine.disease ,DMV, double membrane vesicles ,Pneumonia ,TyG, triglyceride and glucose index ,Amyloid precursor protein ,business - Abstract
The novel coronavirus disease 2019 (COVID-19) has pushed the medical system to its breaking point. While the virus does not discriminate, the elderly and those with comorbidities, including hypertension severe obesity, diabetes mellitus, coronary disease, pneumonia and dementia, are at a greater risk for adverse outcomes due to COVID-19. While many people navigate their new normal, the question of what the long-lasting effects of the pandemic may be, lingers. To investigate how vulnerable populations are affected by the pandemic, we focused on Alzheimer's disease, a vector to understanding how the virus has impacted AD progression and risk via aging. By assessing the effect of COVID-19 on AD patients, we explore genetics, metabolism, and lifestyle factors in both COVID-19 and Alzheimer's disease that can work synergistically to precipitate adverse outcomes. This article also discusses how age-related conditions and/or age-related comorbidities susceptible to COVID-19. We also discuss possible healthy lifestyle factors reduce and/or combat COVID-19 now and in the future.
- Published
- 2021
- Full Text
- View/download PDF
11. Development of a non-alcoholic steatohepatitis model with rapid accumulation of fibrosis, and its treatment using mesenchymal stem cells and their small extracellular vesicles
- Author
-
Takayoshi Suganami, Shuji Terai, Shunsuke Nojiri, Masahiro Ogawa, Tomoaki Yoshida, Atsunori Tsuchiya, Suguru Takeuchi, Takayuki Watanabe, Yoshihiro Ogawa, Masaaki Takamura, and Michiko Itoh
- Subjects
0301 basic medicine ,NAFLD, non-alcoholic fatty liver disease ,Cirrhosis ,MC4R, melanocortin-4 receptor-deficient ,Lipopolysaccharide ,ALT, alanine transaminase ,NASH, non-alcoholic steatohepatitis ,Biomedical Engineering ,Adipose tissue ,MSC, mesenchymal stem cell ,Small extracellular vesicles ,Pharmacology ,hCLS, hepatic crown-like structure ,Biomaterials ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,MCD, methionine-choline-deficient diet ,Fibrosis ,sEV, small extracellular vesicle ,medicine ,lcsh:QH573-671 ,Receptor ,Non-alcoholic steatohepatitis ,lcsh:R5-920 ,WD, Western diet ,biology ,business.industry ,lcsh:Cytology ,ND, normal diet ,Mesenchymal stem cell ,medicine.disease ,030104 developmental biology ,chemistry ,Alanine transaminase ,biology.protein ,Mesenchymal stem cells ,Original Article ,Steatohepatitis ,business ,lcsh:Medicine (General) ,HCC, hepatocellular carcinoma ,T-bil, total bilirubin ,T-cho, total cholesterol ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Introduction Currently, there are no approved drugs for treating non-alcoholic steatohepatitis (NASH); however, mesenchymal stem cells (MSCs) and their small extracellular vesicles (sEVs), which possess immunomodulatory activities, are potential candidates. This study aimed to develop a mouse model of NASH with rapid accumulation of fibrosis using the pre-established melanocortin type-4 receptor knockout (Mc4r-KO) NASH mouse model and lipopolysaccharide (LPS), and to evaluate the therapeutic effect of MSCs and their sEVs. Methods Mc4r-KO mice (8 weeks old, male) were fed a western diet (WD) for 8 weeks. Next, the mice were intraperitoneally injected with lipopolysaccharide (LPS) twice a week for 4 weeks while continuing the WD. To confirm the therapeutic effect of MSCs and sEVs, human adipose tissue-derived MSCs or their sEVs were administered 12 weeks after initiation of the WD, and serum testing, quantitative analysis of fibrosis, and quantitative reverse transcription-polymerase chain reaction qRT-PCR were performed. Results By providing a WD combined with LPS treatment, we successfully developed a NASH model with rapid accumulation of fibrosis. Both human MSCs and their sEVs decreased serum alanine transaminase levels and inflammatory markers based on qRT-PCR. Histological analysis showed that MSC or sEV treatment did not affect fat accumulation. However, an improvement in fibrosis in the groups treated with MSCs and their sEVs was observed. Furthermore, after administering MSCs and sEVs, there was a significant increase in anti-inflammatory macrophages in the liver. Conclusion We successfully developed a NASH model with rapid accumulation of fibrosis and confirmed the anti-inflammatory and anti-fibrotic effects of MSCs and their sEVs, which may be options for future therapy., Graphical abstract Image 1, Highlights • A new animal model of NASH that resembles the human disease was developed. • The new model showed characteristic pathological features of NASH. • Treatment with MSCs and MSC-derived sEVs lowered fibrosis in the animal model. • MSCs and MSC-derived sEVs may be developed as potential therapeutics for NASH.
- Published
- 2019
12. Western Diet-Fed, Aortic-Banded Ossabaw Swine: A Preclinical Model of Cardio-Metabolic Heart Failure
- Author
-
R. John Solaro, Chad M. Warren, Chen Gao, Timothy L. Domeier, Stephanie C. Ferreira-Nichols, Jan R. Ivey, Maike Krenz, Linda Alex, Jaume Padilla, John L. Jones, Yibin Wang, Thomas J. Jurrissen, Christoph Rau, R. Scott Rector, Christopher P. Baines, Jenna C. Edwards, Paula J. Klutho, T. Dylan Olver, David A. Ford, Kerry S. McDonald, Craig A. Emter, Adam B. Veteto, and Pamela K. Thorne
- Subjects
0301 basic medicine ,lcsh:Diseases of the circulatory (Cardiovascular) system ,HF ,EDPVR, end-diastolic pressure−volume relationship ,heart failure ,Disease ,030204 cardiovascular system & hematology ,Cardiorespiratory Medicine and Haematology ,Cardiovascular ,HF, heart failure ,PRECLINICAL RESEARCH ,0302 clinical medicine ,end-diastolic pressure−volume relationship ,Western diet ,LV ,2.1 Biological and endogenous factors ,heart failure with reduced ejection fraction ,Aetiology ,ejection fraction ,integrative pathophysiology ,IL1RL1, interleukin 1 receptor-like 1 ,2. Zero hunger ,HF - Heart failure ,nuclear factor ,Western Diet ,preclinical model of cardiovascular disease ,Phenotype ,3. Good health ,Heart Disease ,WD, Western Diet ,5.1 Pharmaceuticals ,CON, control ,Cardiology ,PTX3, pentraxin-3 ,cardio-metabolic disease ,Development of treatments and therapeutic interventions ,NF, nuclear factor ,Cardiology and Cardiovascular Medicine ,heart failure with preserved ejection fraction ,medicine.medical_specialty ,left ventricle ,NF ,Clinical Sciences ,HFpEF, heart failure with preserved ejection fraction ,03 medical and health sciences ,AB, aortic-banded ,WD ,Cardio metabolic ,Internal medicine ,Genetics ,medicine ,EF, ejection fraction ,HFrEF, heart failure with reduced ejection fraction ,CON ,Nutrition ,PTX3 ,aortic-banded ,business.industry ,HFrEF ,medicine.disease ,HFpEF ,AB ,EF ,Patient population ,030104 developmental biology ,LV, left ventricle ,lcsh:RC666-701 ,Heart failure ,pentraxin-3 ,interleukin 1 receptor-like 1 ,business ,control ,IL1RL1 ,EDPVR - Abstract
Visual Abstract, Highlights • The combination of a Western-diet and aortic banding results in a cardio-metabolic heart failure phenotype in Ossabaw swine. • Ossabaw swine with cardio-metabolic heart failure display cardiac dysfunction at the whole heart and cellular levels. • The left ventricle transcriptome showed gene signatures consistent with pro-inflammatory heart failure. • Cardio-metabolic heart failure was coupled with microvascular dysfunction in the heart, skeletal muscle, and brain. • Ossabaw swine with cardio-metabolic heart failure are sedentary and obese with liver dysfunction., Summary The development of new treatments for heart failure lack animal models that encompass the increasingly heterogeneous disease profile of this patient population. This report provides evidence supporting the hypothesis that Western Diet−fed, aortic-banded Ossabaw swine display an integrated physiological, morphological, and genetic phenotype evocative of cardio-metabolic heart failure. This new preclinical animal model displays a distinctive constellation of findings that are conceivably useful to extending the understanding of how pre-existing cardio-metabolic syndrome can contribute to developing HF.
- Published
- 2019
13. Inhibition of citrate cotransporter Slc13a5/mINDY by RNAi improves hepatic insulin sensitivity and prevents diet-induced non-alcoholic fatty liver disease in mice
- Author
-
Hartmut Ruetten, Sebastian Brachs, Dieter Schmoll, Bodo Brunner, Joachim Spranger, Angelika F. Winkel, Kerstin Jahn-Hofmann, Andreas L. Birkenfeld, Hui Tang, and Daniel Margerie
- Subjects
0301 basic medicine ,HFD, high-fat diet ,Steatosis ,p, perirenal ,Lipid accumulation ,White adipose tissue ,SKM, skeletal muscle ,600 Technik, Medizin, angewandte Wissenschaften::610 Medizin und Gesundheit ,WD, western diet ,chemistry.chemical_compound ,EGP, endogenous glucose production ,GIR, glucose infusion rate ,Mice ,INDY/Slc13a5 ,Non-alcoholic Fatty Liver Disease ,Citrate transport ,IEX, anion-exchange high-performance liquid chromatography ,Citrates ,2-DG, 2-Deoxy-d-glucose ,RER, respiratory exchange ratio ,Dicarboxylic Acid Transporters ,mINDY, Slc13a5/SLC13A5 ,Symporters ,Fatty liver ,WAT, white adipose tissue ,siINDY, mINDY-specific siRNA ,Original Article ,RNA Interference ,e, epididymal ,medicine.medical_specialty ,lcsh:Internal medicine ,TCA, tricarboxylic acid ,Biology ,INDY, ‘I'm not dead Yet’ ,Citric Acid ,03 medical and health sciences ,SCR, non-silencing scrambled control siRNA ,Insulin resistance ,s, subcutaneous ,FLD, fatty liver disease ,Internal medicine ,medicine ,Animals ,lcsh:RC31-1245 ,Molecular Biology ,KO, knockout ,Triglyceride ,solute carrier family 13, member 5 ,EE, energy expenditure ,Lipid metabolism ,Cell Biology ,HE clamp, hyperinsulinemic-euglycemic clamp ,medicine.disease ,Lipid Metabolism ,Diet ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,chemistry ,FA, fatty acids ,siRNA ,ORO, oil red O ,2-Deoxy-D-glucose ,T2D, type-2 diabetes - Abstract
Objective Non-alcoholic fatty liver disease is a world-wide health concern and risk factor for cardio-metabolic diseases. Citrate uptake modifies intracellular hepatic energy metabolism and is controlled by the conserved sodium-dicarboxylate cotransporter solute carrier family 13 member 5 (SLC13A5, mammalian homolog of INDY: mINDY). In Drosophila melanogaster and Caenorhabditis elegans INDY reduction decreased whole-body lipid accumulation. Genetic deletion of Slc13a5 in mice protected from diet-induced adiposity and insulin resistance. We hypothesized that inducible hepatic mINDY inhibition should prevent the development of fatty liver and hepatic insulin resistance. Methods Adult C57BL/6J mice were fed a Western diet (60% kcal from fat, 21% kcal from carbohydrate) ad libitum. Knockdown of mINDY was induced by weekly injection of a chemically modified, liver-selective siRNA for 8 weeks. Mice were metabolically characterized and the effect of mINDY suppression on glucose tolerance as well as insulin sensitivity was assessed with an ipGTT and a hyperinsulinemic-euglycemic clamp. Hepatic lipid accumulation was determined by biochemical measurements and histochemistry. Results Within the 8 week intervention, hepatic mINDY expression was suppressed by a liver-selective siRNA by over 60%. mINDY knockdown improved hepatic insulin sensitivity (i.e. insulin-induced suppression of endogenous glucose production) of C57BL/6J mice in the hyperinsulinemic-euglycemic clamp. Moreover, the siRNA-mediated mINDY inhibition prevented neutral lipid storage and triglyceride accumulation in the liver, while we found no effect on body weight. Conclusions We show that inducible mINDY inhibition improved hepatic insulin sensitivity and prevented diet-induced non-alcoholic fatty liver disease in adult C57BL6/J mice. These effects did not depend on changes of body weight or body composition., Graphical abstract, Highlights • mINDY/Slc13a5 knockdown was induced by liver-selective siRNA in mice. • Liver-selective knockdown of mINDY improved hepatic insulin sensitivity. • Liver-selective knockdown of mINDY prevented steatosis hepatis.
- Published
- 2016
- Full Text
- View/download PDF
14. Sexually dimorphic brain fatty acid composition in low and high fat diet-fed mice
- Author
-
Carlos Rodriguez-Navas, Eugenia Morselli, and Deborah J. Clegg
- Subjects
0301 basic medicine ,Linoleic acid ,DHA, docosahexaenoic acid ,AA, arachidonic acid ,ACC, acetyl-CoA carboxylase ,WD, western diet ,Palmitic acid ,chemistry.chemical_compound ,0302 clinical medicine ,Western diet ,chemistry.chemical_classification ,biology ,UnsatFAs, unsaturated fatty acids ,FAS, fatty acid synthase ,BBB, blood brain barrier ,Fatty acid synthase ,Biochemistry ,Docosahexaenoic acid ,MSFD2a, membrane protein major facilitator super family domain containing 2a ,SatFAs, saturated fatty acids ,M, male ,Arachidonic acid ,Original Article ,FFAs, free fatty acids ,Polyunsaturated fatty acid ,N43 ,medicine.medical_specialty ,lcsh:Internal medicine ,ω6-fatty acids ,NF-κB, Nuclear Factor-κ Beta ,IL6, interleukin 6 ,CNS, central nervous system ,Fatty acid-binding protein ,03 medical and health sciences ,PA, palmitic acid ,FAT/CD36, fatty acid transporter ,Internal medicine ,medicine ,Obesity ,lcsh:RC31-1245 ,Molecular Biology ,LA, linoleic acid ,B2m, beta-2 microglobulin ,PUFAs, polyunsaturated fatty acids ,Fatty acid ,F, female ,Cell Biology ,FAs, fatty acids ,FABP, fatty acid binding protein ,TNFα, Tumor Necrosis Factor α ,FATP1, fatty acid transport protein 1 ,MUFAs, monounsaturated fatty acids ,WT, wild-type ,030104 developmental biology ,Endocrinology ,chemistry ,TFAs, total fatty acids ,Central nervous system ,MCD, malonyl-CoA decarboxylase ,biology.protein ,BSA, bovine serum albumin ,030217 neurology & neurosurgery ,C, Chow diet - Abstract
Objective In this study, we analyzed the fatty acid profile of brains and plasma from male and female mice fed chow or a western-style high fat diet (WD) for 16 weeks to determine if males and females process fatty acids differently. Based on the differences in fatty acids observed in vivo, we performed in vitro experiments on N43 hypothalamic neuronal cells to begin to elucidate how the fatty acid milieu may impact brain inflammation. Methods Using a comprehensive mass spectrometry fatty acid analysis, which includes a profile for 52 different fatty acid isomers, we assayed the plasma and brain fatty acid composition of age-matched male and female mice maintained on chow or a WD. Additionally, using the same techniques, we determined the fatty acid composition of N43 hypothalamic cells following exposure to palmitic and linoleic acid, alone or in combination. Results Our data demonstrate there is a sexual dimorphism in brain fatty acid content both following the consumption of the chow diet, as well as the WD, with males having an increased percentage of saturated fatty acids and reductions in ω6-polyunsaturated fatty acids when compared to females. Interestingly, we did not observe a sexual dimorphism in fatty acid content in the plasma of the same mice. Furthermore, exposure of N43 cells to the ω6-PUFA linoleic acid, which is higher in female brains when compared to males, reduces palmitic acid-induced inflammation. Conclusions Our data suggest male and female brains, and not plasma, differ in their fatty acid profile. This is the first time, to our knowledge, lipidomic analyses has been used to directly test the hypothesis there is a sexual dimorphism in brain and plasma fatty acid composition following consumption of the chow diet, as well as following exposure to the WD., Highlights • There is a sexual dimorphism in brain lipid composition. • Brains of male mice following high fat diet exposure are enriched in saturated fatty acids. • Male brains are depleted of polyunsaturated fatty acids following high fat diet exposure. • Linoleic acid inhibits palmitic acid-induced inflammation in hypothalamic cells.
- Published
- 2016
15. Novel Göttingen Miniswine Model of Heart Failure With Preserved Ejection Fraction Integrating Multiple Comorbidities.
- Author
-
Sharp TE 3rd, Scarborough AL, Li Z, Polhemus DJ, Hidalgo HA, Schumacher JD, Matsuura TR, Jenkins JS, Kelly DP, Goodchild TT, and Lefer DJ
- Abstract
A lack of preclinical large animal models of heart failure with preserved ejection fraction (HFpEF) that recapitulate this comorbid-laden syndrome has led to the inability to tease out mechanistic insights and to test novel therapeutic strategies. This study developed a large animal model that integrated multiple comorbid determinants of HFpEF in a miniswine breed that exhibited sensitivity to obesity, metabolic syndrome, and vascular disease with overt clinical signs of heart failure. The combination of a Western diet and 11-deoxycorticosterone acetate salt-induced hypertension in the Göttingen miniswine led to the development of a novel large animal model of HFpEF that exhibited multiorgan involvement and a full spectrum of comorbidities associated with human HFpEF., Competing Interests: Dr. Kelly is supported in part by the National Institutes of Health (R01 HL128349 and R01 HL151345). Dr. Matsuura is supported by the National Institute of Health (T32HL007843). Dr. Lefer was supported in part by the National Institutes of Health (1R01 HL146098, 1R56HL137711, 1R01 HL146514, and 1R01 HL151398). Drs. Sharp, Goodchild, and Lefer have a pending patent on the composition and methods for modeling HFpEF in Göttingen minipigs. All other authors have reported that they have no relationships relevant to the contents of this paper to disclose., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
16. Development of a non-alcoholic steatohepatitis model with rapid accumulation of fibrosis, and its treatment using mesenchymal stem cells and their small extracellular vesicles.
- Author
-
Watanabe T, Tsuchiya A, Takeuchi S, Nojiri S, Yoshida T, Ogawa M, Itoh M, Takamura M, Suganami T, Ogawa Y, and Terai S
- Abstract
Introduction: Currently, there are no approved drugs for treating non-alcoholic steatohepatitis (NASH); however, mesenchymal stem cells (MSCs) and their small extracellular vesicles (sEVs), which possess immunomodulatory activities, are potential candidates. This study aimed to develop a mouse model of NASH with rapid accumulation of fibrosis using the pre-established melanocortin type-4 receptor knockout ( Mc4r -KO) NASH mouse model and lipopolysaccharide (LPS), and to evaluate the therapeutic effect of MSCs and their sEVs., Methods: Mc4r -KO mice (8 weeks old, male) were fed a western diet (WD) for 8 weeks. Next, the mice were intraperitoneally injected with lipopolysaccharide (LPS) twice a week for 4 weeks while continuing the WD. To confirm the therapeutic effect of MSCs and sEVs, human adipose tissue-derived MSCs or their sEVs were administered 12 weeks after initiation of the WD, and serum testing, quantitative analysis of fibrosis, and quantitative reverse transcription-polymerase chain reaction qRT-PCR were performed., Results: By providing a WD combined with LPS treatment, we successfully developed a NASH model with rapid accumulation of fibrosis. Both human MSCs and their sEVs decreased serum alanine transaminase levels and inflammatory markers based on qRT-PCR. Histological analysis showed that MSC or sEV treatment did not affect fat accumulation. However, an improvement in fibrosis in the groups treated with MSCs and their sEVs was observed. Furthermore, after administering MSCs and sEVs, there was a significant increase in anti-inflammatory macrophages in the liver., Conclusion: We successfully developed a NASH model with rapid accumulation of fibrosis and confirmed the anti-inflammatory and anti-fibrotic effects of MSCs and their sEVs, which may be options for future therapy., Competing Interests: The authors declare no conflict of interest., (© 2020 The Japanese Society for Regenerative Medicine. Production and hosting by Elsevier B.V.)
- Published
- 2020
- Full Text
- View/download PDF
17. miR-22 inhibition reduces hepatic steatosis via FGF21 and FGFR1 induction.
- Author
-
Hu Y, Liu HX, Jena PK, Sheng L, Ali MR, and Wan YY
- Abstract
Background & Aims: Metabolism supports cell proliferation and growth. Surprisingly, the tumor suppressor miR-22 is induced by metabolic stimulators like bile acids. Thus, this study examines whether miR-22 could be a metabolic silencer., Methods: The relationship between miR-22 and the expression of fibroblast growth factor 21 (FGF21) and its receptor FGFR1 was studied in cells and fatty livers obtained from patients and mouse models. We evaluated the effect of an miR-22 inhibitor alone and in combination with obeticholic acid (OCA) for the treatment of steatosis., Results: The levels of miR-22 were inversely correlated with those of FGF21 , FGFR1, and PGC1α in human and mouse fatty livers, suggesting that hepatic miR-22 acts as a metabolic silencer. Indeed, miR-22 reduced FGFR1 by direct targeting and decreased FGF21 by reducing the recruitment of PPARα and PGC1α to their binding motifs. In contrast, an miR-22 inhibitor increases hepatic FGF21 and FGFR1, leading to AMPK and ERK1/2 activation, which was effective in treating alcoholic steatosis in mouse models. The farnesoid x receptor-agonist OCA induced FGF21 and FGFR1, as well as their inhibitor miR-22 . An miR-22 inhibitor and OCA were effective in treating diet-induced steatosis, both alone and in combination. The combined treatment was the most effective at improving insulin sensitivity, releasing glucagon-like peptide 1, and reducing hepatic triglyceride in obese mice., Conclusion: The simultaneous induction of miR-22 , FGF21 and FGFR1 by metabolic stimulators may maintain FGF21 homeostasis and restrict ERK1/2 activation. Reducing miR-22 enhances hepatic FGF21 and activates AMPK, which could be a novel approach to treat steatosis and insulin resistance., Lay Summary: This study examines the metabolic role of a tumor suppressor, miR-22 , that can be induced by metabolic stimulators such as bile acids. Our novel data revealed that the metabolic silencing effect of miR-22 occurs as a result of reductions in metabolic stimulators, which likely contribute to the development of fatty liver. Consistent with this finding, an miR-22 inhibitor effectively reversed both alcohol- and diet-induced fatty liver; miR-22 inhibition is a promising therapeutic option which could be used in combination with obeticholic acid., Competing Interests: The authors declare no conflicts of interest that pertain to this work. Please refer to the accompanying ICMJE disclosure forms for further details., (© 2020 The Author(s).)
- Published
- 2020
- Full Text
- View/download PDF
18. Western Diet-Fed, Aortic-Banded Ossabaw Swine: A Preclinical Model of Cardio-Metabolic Heart Failure.
- Author
-
Olver TD, Edwards JC, Jurrissen TJ, Veteto AB, Jones JL, Gao C, Rau C, Warren CM, Klutho PJ, Alex L, Ferreira-Nichols SC, Ivey JR, Thorne PK, McDonald KS, Krenz M, Baines CP, Solaro RJ, Wang Y, Ford DA, Domeier TL, Padilla J, Rector RS, and Emter CA
- Abstract
The development of new treatments for heart failure lack animal models that encompass the increasingly heterogeneous disease profile of this patient population. This report provides evidence supporting the hypothesis that Western Diet-fed, aortic-banded Ossabaw swine display an integrated physiological, morphological, and genetic phenotype evocative of cardio-metabolic heart failure. This new preclinical animal model displays a distinctive constellation of findings that are conceivably useful to extending the understanding of how pre-existing cardio-metabolic syndrome can contribute to developing HF.
- Published
- 2019
- Full Text
- View/download PDF
19. The GLP-1 Analogs Liraglutide and Semaglutide Reduce Atherosclerosis in ApoE -/- and LDLr -/- Mice by a Mechanism That Includes Inflammatory Pathways.
- Author
-
Rakipovski G, Rolin B, Nøhr J, Klewe I, Frederiksen KS, Augustin R, Hecksher-Sørensen J, Ingvorsen C, Polex-Wolf J, and Knudsen LB
- Abstract
The glucagon-like peptide-1 receptor agonists (GLP-1RAs) liraglutide and semaglutide reduce cardiovascular risk in type 2 diabetes patients. The mode of action is suggested to occur through modified atherosclerotic progression. In this study, both of the compounds significantly attenuated plaque lesion development in apolipoprotein E-deficient (ApoE
-/- ) mice and low-density lipoprotein receptor-deficient (LDLr-/- ) mice. This attenuation was partly independent of weight and cholesterol lowering. In aortic tissue, exposure to a Western diet alters expression of genes in pathways relevant to the pathogenesis of atherosclerosis, including leukocyte recruitment, leukocyte rolling, adhesion/extravasation, cholesterol metabolism, lipid-mediated signaling, extracellular matrix protein turnover, and plaque hemorrhage. Treatment with semaglutide significantly reversed these changes. These data suggest GLP-1RAs affect atherosclerosis through an anti-inflammatory mechanism.- Published
- 2018
- Full Text
- View/download PDF
20. Inhibition of citrate cotransporter Slc13a5/mINDY by RNAi improves hepatic insulin sensitivity and prevents diet-induced non-alcoholic fatty liver disease in mice.
- Author
-
Brachs S, Winkel AF, Tang H, Birkenfeld AL, Brunner B, Jahn-Hofmann K, Margerie D, Ruetten H, Schmoll D, and Spranger J
- Subjects
- Animals, Citrates, Citric Acid, Diet, Mice, Mice, Inbred C57BL, Dicarboxylic Acid Transporters physiology, Insulin Resistance, Lipid Metabolism, Non-alcoholic Fatty Liver Disease, RNA Interference, Symporters physiology
- Abstract
Objective: Non-alcoholic fatty liver disease is a world-wide health concern and risk factor for cardio-metabolic diseases. Citrate uptake modifies intracellular hepatic energy metabolism and is controlled by the conserved sodium-dicarboxylate cotransporter solute carrier family 13 member 5 (SLC13A5, mammalian homolog of INDY: mINDY). In Drosophila melanogast er and Caenorhabditis elegans INDY reduction decreased whole-body lipid accumulation. Genetic deletion of Slc13a5 in mice protected from diet-induced adiposity and insulin resistance. We hypothesized that inducible hepatic mINDY inhibition should prevent the development of fatty liver and hepatic insulin resistance., Methods: Adult C57BL/6J mice were fed a Western diet (60% kcal from fat, 21% kcal from carbohydrate) ad libitum. Knockdown of mINDY was induced by weekly injection of a chemically modified, liver-selective siRNA for 8 weeks. Mice were metabolically characterized and the effect of mINDY suppression on glucose tolerance as well as insulin sensitivity was assessed with an ipGTT and a hyperinsulinemic-euglycemic clamp. Hepatic lipid accumulation was determined by biochemical measurements and histochemistry., Results: Within the 8 week intervention, hepatic mINDY expression was suppressed by a liver-selective siRNA by over 60%. mINDY knockdown improved hepatic insulin sensitivity (i.e. insulin-induced suppression of endogenous glucose production) of C57BL/6J mice in the hyperinsulinemic-euglycemic clamp. Moreover, the siRNA-mediated mINDY inhibition prevented neutral lipid storage and triglyceride accumulation in the liver, while we found no effect on body weight., Conclusions: We show that inducible mINDY inhibition improved hepatic insulin sensitivity and prevented diet-induced non-alcoholic fatty liver disease in adult C57BL6/J mice. These effects did not depend on changes of body weight or body composition.
- Published
- 2016
- Full Text
- View/download PDF
21. Sexually dimorphic brain fatty acid composition in low and high fat diet-fed mice.
- Author
-
Rodriguez-Navas C, Morselli E, and Clegg DJ
- Abstract
Objective: In this study, we analyzed the fatty acid profile of brains and plasma from male and female mice fed chow or a western-style high fat diet (WD) for 16 weeks to determine if males and females process fatty acids differently. Based on the differences in fatty acids observed in vivo, we performed in vitro experiments on N43 hypothalamic neuronal cells to begin to elucidate how the fatty acid milieu may impact brain inflammation., Methods: Using a comprehensive mass spectrometry fatty acid analysis, which includes a profile for 52 different fatty acid isomers, we assayed the plasma and brain fatty acid composition of age-matched male and female mice maintained on chow or a WD. Additionally, using the same techniques, we determined the fatty acid composition of N43 hypothalamic cells following exposure to palmitic and linoleic acid, alone or in combination., Results: Our data demonstrate there is a sexual dimorphism in brain fatty acid content both following the consumption of the chow diet, as well as the WD, with males having an increased percentage of saturated fatty acids and reductions in ω6-polyunsaturated fatty acids when compared to females. Interestingly, we did not observe a sexual dimorphism in fatty acid content in the plasma of the same mice. Furthermore, exposure of N43 cells to the ω6-PUFA linoleic acid, which is higher in female brains when compared to males, reduces palmitic acid-induced inflammation., Conclusions: Our data suggest male and female brains, and not plasma, differ in their fatty acid profile. This is the first time, to our knowledge, lipidomic analyses has been used to directly test the hypothesis there is a sexual dimorphism in brain and plasma fatty acid composition following consumption of the chow diet, as well as following exposure to the WD.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.