Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Mol Metab ; 78: 101815, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37797918

ABSTRACT

BACKGROUND AND AIMS: Genome and epigenome wide association studies identified variants in carnitine palmitoyltransferase 1a (CPT1a) that associate with lipid traits. The goal of this study was to determine the role of liver-specific CPT1a on hepatic lipid metabolism. APPROACH AND RESULTS: Male and female liver-specific knockout (LKO) and littermate controls were placed on a low-fat or high-fat diet (60% kcal fat) for 15 weeks. Mice were necropsied after a 16 h fast, and tissues were collected for lipidomics, matrix-assisted laser desorption ionization mass spectrometry imaging, kinome analysis, RNA-sequencing, and protein expression by immunoblotting. Female LKO mice had increased serum alanine aminotransferase levels which were associated with greater deposition of hepatic lipids, while male mice were not affected by CPT1a deletion relative to male control mice. Mice with CPT1a deletion had reductions in DHA-containing phospholipids at the expense of monounsaturated fatty acids (MUFA)-containing phospholipids in whole liver and at the level of the lipid droplet (LD). Male and female LKO mice increased RNA levels of genes involved in LD lipolysis (Plin2, Cidec, G0S2) and in polyunsaturated fatty acid metabolism (Elovl5, Fads1, Elovl2), while only female LKO mice increased genes involved in inflammation (Ly6d, Mmp12, Cxcl2). Kinase profiling showed decreased protein kinase A activity, which coincided with increased PLIN2, PLIN5, and G0S2 protein levels and decreased triglyceride hydrolysis in LKO mice. CONCLUSIONS: Liver-specific deletion of CPT1a promotes sexually dimorphic steatotic liver disease (SLD) in mice, and here we have identified new mechanisms by which females are protected from HFD-induced liver injury.


Subject(s)
Docosahexaenoic Acids , Fatty Liver , Female , Male , Animals , Mice , Phospholipids , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Fatty Liver/metabolism , RNA
2.
bioRxiv ; 2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37645721

ABSTRACT

Background and Aims: Genome and epigenome wide association studies identified variants in carnitine palmitoyltransferase 1a (CPT1a) that associate with lipid traits. The goal of this study was to determine the impact by which liver-specific CPT1a deletion impacts hepatic lipid metabolism. Approach and Results: Six-to-eight-week old male and female liver-specific knockout (LKO) and littermate controls were placed on a low-fat or high-fat diet (HFD; 60% kcal fat) for 15 weeks. Mice were necropsied after a 16 hour fast, and tissues were collected for lipidomics, matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI), kinome analysis, RNA-sequencing, and protein expression by immunoblotting. Female LKO mice had increased serum alanine aminotransferase (ALT) levels which were associated with greater deposition of hepatic lipids, while male mice were not affected by CPT1a deletion relative to male control mice. Mice with CPT1a deletion had reductions in DHA-containing phospholipids at the expense of monounsaturated fatty acids (MUFA)-containing phospholipids in both whole liver and at the level of the lipid droplet (LD). Male and female LKO mice increased RNA levels of genes involved in LD lipolysis ( Plin2 , Cidec , G0S2 ) and in polyunsaturated fatty acid (PUFA) metabolism ( Elovl5, Fads1, Elovl2 ), while only female LKO mice increased genes involved in inflammation ( Ly6d, Mmp12, Cxcl2 ). Kinase profiling showed decreased protein kinase A (PKA) activity, which coincided with increased PLIN2, PLIN5, and G0S2 protein levels and decreased triglyceride hydrolysis in LKO mice. Conclusions: Liver-specific deletion of CPT1a promotes sexually dimorphic steatotic liver disease (SLD) in mice, and here we have identified new mechanisms by which females are protected from HFD-induced liver injury.

3.
Drug Metab Dispos ; 51(9): 1207-1215, 2023 09.
Article in English | MEDLINE | ID: mdl-37230767

ABSTRACT

Pregnane X receptor (PXR) is a xenobiotic receptor that can be activated by numerous chemicals including endogenous hormones, dietary steroids, pharmaceutical agents, and environmental chemicals. PXR has been established to function as a xenobiotic sensor to coordinately regulate xenobiotic metabolism by regulating the expression of many enzymes and transporters required for xenobiotic metabolism. Recent studies have implicated a potentially important role for PXR in obesity and metabolic disease beyond xenobiotic metabolism, but how PXR action in different tissues or cell types contributes to obesity and metabolic disorders remains elusive. To investigate the role of adipocyte PXR in obesity, we generated a novel adipocyte-specific PXR deficient mouse model (PXRΔAd). Notably, we found that loss of adipocyte PXR did not affect food intake, energy expenditure, and obesity in high-fat diet-fed male mice. PXRΔAd mice also had similar obesity-associated metabolic disorders including insulin resistance and hepatic steatosis as control littermates. PXR deficiency in adipocytes did not affect expression of key adipose genes in PXRΔAd mice. Our findings suggest that adipocyte PXR signaling may be dispensable in diet-induced obesity and metabolic disorders in mice. Further studies are needed to understand the role of PXR signaling in obesity and metabolic disorders in the future. SIGNIFICANCE STATEMENT: The authors demonstrate that deficiency of adipocyte pregnane X receptor (PXR) does not affect diet-induced obesity or metabolic disorders in mice and infers that adipocyte PXR signaling may not play a key role in diet-induced obesity. More studies are needed to understand the tissue-specific role of PXR in obesity.


Subject(s)
Insulin Resistance , Receptors, Steroid , Male , Mice , Animals , Pregnane X Receptor/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Xenobiotics/metabolism , Obesity/etiology , Obesity/metabolism , Adipocytes/metabolism , Diet, High-Fat/adverse effects
4.
Metabolism ; 145: 155591, 2023 08.
Article in English | MEDLINE | ID: mdl-37230214

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a liver manifestation of metabolic syndrome, and is estimated to affect one billion individuals worldwide. An increased intake of a high-fat diet (HFD) and sugar-sweetened beverages are risk-factors for NAFLD development, but how their combined intake promotes progression to a more severe form of liver injury is unknown. Here we show that fructose metabolism via ketohexokinase (KHK) C isoform leads to unresolved endoplasmic reticulum (ER) stress when coupled with a HFD intake. Conversely, a liver-specific knockdown of KHK in mice consuming fructose on a HFD is adequate to improve the NAFLD activity score and exert a profound effect on the hepatic transcriptome. Overexpression of KHK-C in cultured hepatocytes is sufficient to induce ER stress in fructose free media. Upregulation of KHK-C is also observed in mice with genetically induced obesity or metabolic dysfunction, whereas KHK knockdown in these mice improves metabolic function. Additionally, in over 100 inbred strains of male or female mice hepatic KHK expression correlates positively with adiposity, insulin resistance, and liver triglycerides. Similarly, in 241 human subjects and their controls, hepatic Khk expression is upregulated in early, but not late stages of NAFLD. In summary, we describe a novel role of KHK-C in triggering ER stress, which offers a mechanistic understanding of how the combined intake of fructose and a HFD propagates the development of metabolic complications.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Female , Humans , Male , Mice , Diet, High-Fat/adverse effects , Fructokinases/genetics , Fructokinases/metabolism , Fructose/pharmacology , Lipogenesis/physiology , Liver/metabolism , Models, Genetic , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism
5.
bioRxiv ; 2023 Jan 27.
Article in English | MEDLINE | ID: mdl-36747758

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a liver manifestation of metabolic syndrome, and is estimated to affect one billion individuals worldwide. An increased intake of a high-fat diet (HFD) and sugar-sweetened beverages are risk-factors for NAFLD development, but how their combined intake promotes progression to a more severe form of liver injury is unknown. Here we show that fructose metabolism via ketohexokinase (KHK) C isoform increases endoplasmic reticulum (ER) stress in a dose dependent fashion, so when fructose is coupled with a HFD intake it leads to unresolved ER stress. Conversely, a liver-specific knockdown of KHK in C57BL/6J male mice consuming fructose on a HFD is adequate to improve the NAFLD activity score and exert a profound effect on the hepatic transcriptome. Overexpression of KHK-C in cultured hepatocytes is sufficient to induce ER stress in fructose free media. Upregulation of KHK-C is also observed in genetically obesity ob/ob, db/db and lipodystrophic FIRKO male mice, whereas KHK knockdown in these mice improves metabolic function. Additionally, in over 100 inbred strains of male or female mice hepatic KHK expression correlates positively with adiposity, insulin resistance, and liver triglycerides. Similarly, in 241 human subjects and their controls, hepatic Khk expression is upregulated in early, but not late stages of NAFLD. In summary, we describe a novel role of KHK-C in triggering ER stress, which offers a mechanistic understanding of how the combined intake of fructose and a HFD propagates the development of metabolic complications.

6.
J Hepatol ; 79(1): 25-42, 2023 07.
Article in English | MEDLINE | ID: mdl-36822479

ABSTRACT

BACKGROUND & AIMS: The consumption of sugar and a high-fat diet (HFD) promotes the development of obesity and metabolic dysfunction. Despite their well-known synergy, the mechanisms by which sugar worsens the outcomes associated with a HFD are largely elusive. METHODS: Six-week-old, male, C57Bl/6 J mice were fed either chow or a HFD and were provided with regular, fructose- or glucose-sweetened water. Moreover, cultured AML12 hepatocytes were engineered to overexpress ketohexokinase-C (KHK-C) using a lentivirus vector, while CRISPR-Cas9 was used to knockdown CPT1α. The cell culture experiments were complemented with in vivo studies using mice with hepatic overexpression of KHK-C and in mice with liver-specific CPT1α knockout. We used comprehensive metabolomics, electron microscopy, mitochondrial substrate phenotyping, proteomics and acetylome analysis to investigate underlying mechanisms. RESULTS: Fructose supplementation in mice fed normal chow and fructose or glucose supplementation in mice fed a HFD increase KHK-C, an enzyme that catalyzes the first step of fructolysis. Elevated KHK-C is associated with an increase in lipogenic proteins, such as ACLY, without affecting their mRNA expression. An increase in KHK-C also correlates with acetylation of CPT1α at K508, and lower CPT1α protein in vivo. In vitro, KHK-C overexpression lowers CPT1α and increases triglyceride accumulation. The effects of KHK-C are, in part, replicated by a knockdown of CPT1α. An increase in KHK-C correlates negatively with CPT1α protein levels in mice fed sugar and a HFD, but also in genetically obese db/db and lipodystrophic FIRKO mice. Mechanistically, overexpression of KHK-C in vitro increases global protein acetylation and decreases levels of the major cytoplasmic deacetylase, SIRT2. CONCLUSIONS: KHK-C-induced acetylation is a novel mechanism by which dietary fructose augments lipogenesis and decreases fatty acid oxidation to promote the development of metabolic complications. IMPACT AND IMPLICATIONS: Fructose is a highly lipogenic nutrient whose negative consequences have been largely attributed to increased de novo lipogenesis. Herein, we show that fructose upregulates ketohexokinase, which in turn modifies global protein acetylation, including acetylation of CPT1a, to decrease fatty acid oxidation. Our findings broaden the impact of dietary sugar beyond its lipogenic role and have implications on drug development aimed at reducing the harmful effects attributed to sugar metabolism.


Subject(s)
Carnitine O-Palmitoyltransferase , Liver , Male , Mice , Animals , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/pharmacology , Acetylation , Liver/metabolism , Obesity/metabolism , Glucose/metabolism , Diet, High-Fat/adverse effects , Fatty Acids/metabolism , Fructose/metabolism , Fructokinases/genetics , Fructokinases/metabolism
7.
J Nutr Biochem ; 114: 109224, 2023 04.
Article in English | MEDLINE | ID: mdl-36403701

ABSTRACT

Increased fructose intake from sugar-sweetened beverages and highly processed sweets is a well-recognized risk factor for the development of obesity and its complications. Fructose strongly supports lipogenesis on a normal chow diet by providing both, a substrate for lipid synthesis and activation of lipogenic transcription factors. However, the negative health consequences of dietary sugar are best observed with the concomitant intake of a HFD. Indeed, the most commonly used obesogenic research diets, such as "Western diet", contain both fructose and a high amount of fat. In spite of its common use, how the combined intake of fructose and fat synergistically supports development of metabolic complications is not fully elucidated. Here we present the preponderance of evidence that fructose consumption decreases oxidation of dietary fat in human and animal studies. We provide a detailed review of the mitochondrial ß-oxidation pathway. Fructose affects hepatic activation of fatty acyl-CoAs, decreases acylcarnitine production and impairs the carnitine shuttle. Mechanistically, fructose suppresses transcriptional activity of PPARα and its target CPT1α, the rate limiting enzyme of acylcarnitine production. These effects of fructose may be, in part, mediated by protein acetylation. Acetylation of PGC1α, a co-activator of PPARα and acetylation of CPT1α, in part, account for fructose-impaired acylcarnitine production. Interestingly, metabolic effects of fructose in the liver can be largely overcome by carnitine supplementation. In summary, fructose decreases oxidation of dietary fat in the liver, in part, by impairing acylcarnitine production, offering one explanation for the synergistic effects of these nutrients on the development of metabolic complications, such as NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Humans , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Fructose/metabolism , PPAR alpha/metabolism , Liver/metabolism , Carnitine/metabolism , Diet, Western/adverse effects , Dietary Fats/pharmacology , Diet, High-Fat
8.
STAR Protoc ; 2(3): 100731, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34409309

ABSTRACT

Ketohexokinase (KHK) catalyzes the first step of fructose metabolism. Inhibitors of KHK enzymatic activity are being evaluated in clinical trials for the treatment of non-alcoholic fatty liver disease (NAFLD) and diabetes. Here, we present a luminescence-based protocol to quantify KHK activity. The accuracy of this technique has been validated using knockdown and overexpression of KHK in vivo and in vitro. The specificity of the assay has been verified using 3-O-methyl-D-fructose, a non-metabolizable analog of fructose, heat inactivation of hexokinases, and depletion of potassium. For complete details on the use of this protocol, please refer to Damen et al. (2021).


Subject(s)
Enzyme Assays/methods , Fructokinases/metabolism , Fructose/metabolism , Luminescent Measurements/methods , Animals , Carbohydrate Metabolism , Fructokinases/antagonists & inhibitors , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Luminescence , Mice , Non-alcoholic Fatty Liver Disease
9.
Int J Obes (Lond) ; 45(11): 2377-2387, 2021 11.
Article in English | MEDLINE | ID: mdl-34302121

ABSTRACT

OBJECTIVE: The risks of excess sugar intake in addition to high-fat diet consumption on immunopathogenesis of obesity-associated metabolic diseases are poorly defined. Interleukin-4 (IL-4) and IL-13 signaling via IL-4Rα regulates adipose tissue lipolysis, insulin sensitivity, and liver fibrosis in obesity. However, the contribution of IL-4Rα to sugar rich diet-driven obesity and metabolic sequelae remains unknown. METHODS: WT, IL-4Rα-deficient (IL-4Rα-/-) and STAT6-deficient mice (STAT6-/-) male mice were fed low-fat chow, high fat (HF) or HF plus high carbohydrate (HC/fructose) diet (HF + HC). Analysis included quantification of: (i) body weight, adiposity, energy expenditure, fructose metabolism, fatty acid oxidation/synthesis, glucose dysmetabolism and hepatocellular damage; (ii) the contribution of the hematopoietic or non-hematopoietic IL-4Rα expression; and (iii) the relevance of IL-4Rα downstream canonical STAT6 signaling pathway in this setting. RESULTS: We show that IL-4Rα regulated HF + HC diet-driven weight gain, whole body adiposity, adipose tissue inflammatory gene expression, energy expenditure, locomotor activity, glucose metabolism, hepatic steatosis, hepatic inflammatory gene expression and hepatocellular damage. These effects were potentially, and in part, dependent on non-hematopoietic IL-4Rα expression but were independent of direct STAT6 activation. Mechanistically, hepatic ketohexokinase-A and C expression was dependent on IL-4Rα, as it was reduced in IL-4Rα-deficient mice. KHK activity was also affected by HF + HC dietary challenge. Further, reduced expression/activity of KHK in IL-4Rα mice had a significant effect on fatty acid oxidation and fatty acid synthesis pathways. CONCLUSION: Our findings highlight potential contribution of non-hematopoietic IL-4Rα activation of a non-canonical signaling pathway that regulates the HF + HC diet-driven induction of obesity and severity of obesity-associated sequelae.


Subject(s)
Energy Metabolism/physiology , Interleukin-4/metabolism , Obesity/metabolism , Animals , Disease Models, Animal , Fructose/adverse effects , Insulin Resistance/physiology , Interleukin-4/analysis , Mice , Obesity/immunology
11.
J Lipid Res ; 61(5): 696-706, 2020 05.
Article in English | MEDLINE | ID: mdl-32170024

ABSTRACT

The pregnane X receptor (PXR) is a nuclear receptor that can be activated by numerous drugs and xenobiotic chemicals. PXR thereby functions as a xenobiotic sensor to coordinately regulate host responses to xenobiotics by transcriptionally regulating many genes involved in xenobiotic metabolism. We have previously reported that PXR has pro-atherogenic effects in animal models, but how PXR contributes to atherosclerosis development in different tissues or cell types remains elusive. In this study, we generated an LDL receptor-deficient mouse model with myeloid-specific PXR deficiency (PXRΔMyeLDLR-/-) to elucidate the role of macrophage PXR signaling in atherogenesis. The myeloid PXR deficiency did not affect metabolic phenotypes and plasma lipid profiles, but PXRΔMyeLDLR-/- mice had significantly decreased atherosclerosis at both aortic root and brachiocephalic arteries compared with control littermates. Interestingly, the PXR deletion did not affect macrophage adhesion and migration properties, but reduced lipid accumulation and foam cell formation in the macrophages. PXR deficiency also led to decreased expression of the scavenger receptor CD36 and impaired lipid uptake in macrophages of the PXRΔMyeLDLR-/- mice. Further, RNA-Seq analysis indicated that treatment with a prototypical PXR ligand affects the expression of many atherosclerosis-related genes in macrophages in vitro. These findings reveal a pivotal role of myeloid PXR signaling in atherosclerosis development and suggest that PXR may be a potential therapeutic target in atherosclerosis management.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/metabolism , Macrophages/metabolism , Pregnane X Receptor/deficiency , Receptors, LDL/deficiency , Animals , CD36 Antigens/metabolism , Foam Cells/cytology , Foam Cells/metabolism , Gene Expression Regulation , Lipids/blood , Mice , Phenotype
12.
J Am Heart Assoc ; 8(12): e012009, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31203708

ABSTRACT

Background Obesity-associated chronic inflammation has been known to contribute to atherosclerosis development, but the underlying mechanisms remain elusive. Recent studies have revealed novel functions of IKK ß (inhibitor of NF -κB [nuclear factor κB] kinase ß), a key coordinator of inflammation through activation of NF -κB, in atherosclerosis and adipose tissue development. However, it is not clear whether IKK ß signaling in adipocytes can also affect atherogenesis. This study aims to investigate the impact of adipocyte IKK ß expression on atherosclerosis development in lean and obese LDLR (low-density lipoprotein receptor)-deficient ( LDLR -/-) mice. Methods and Results To define the role of adipocyte IKK ß in atherogenesis, we generated adipocyte-specific IKK ß-deficient LDLR -/- ( IKK ßΔAd LDLR -/-) mice. Targeted deletion of IKK ß in adipocytes did not affect adiposity and atherosclerosis in lean LDLR -/- mice when fed a low-fat diet. In response to high-fat feeding, however, IKK ßΔAd LDLR -/- mice had defective adipose remodeling and increased adipose tissue and systemic inflammation. Deficiency of adipocyte IKK ß did not affect atherosclerotic lesion sizes but resulted in enhanced lesional inflammation and increased plaque vulnerability in obese IKK ßΔAd LDLR -/- mice. Conclusions These data demonstrate that adipocyte IKK ß signaling affects the evolution of atherosclerosis plaque vulnerability in obese LDLR -/- mice. This study suggests that the functions of IKK ß signaling in atherogenesis are complex, and IKK ß in different cell types or tissues may have different effects on atherosclerosis development.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Adipocytes/enzymology , I-kappa B Kinase/deficiency , Plaque, Atherosclerotic/etiology , Animals , Male , Mice, Obese
13.
JCI Insight ; 4(3)2019 Feb 07.
Article in English | MEDLINE | ID: mdl-30728326

ABSTRACT

Quetiapine, one of the most prescribed atypical antipsychotics, has been associated with hyperlipidemia and an increased risk for cardiovascular disease in patients, but the underlying mechanisms remain unknown. Here, we identified quetiapine as a potent and selective agonist for pregnane X receptor (PXR), a key nuclear receptor that regulates xenobiotic metabolism in the liver and intestine. Recent studies have indicated that PXR also plays an important role in lipid homeostasis. We generated potentially novel tissue-specific PXR-KO mice and demonstrated that quetiapine induced hyperlipidemia by activating intestinal PXR signaling. Quetiapine-mediated PXR activation stimulated the intestinal expression of cholesterol transporter Niemann-Pick C1-Like 1 (NPC1L1) and microsomal triglyceride transfer protein (MTP), leading to increased intestinal lipid absorption. While NPC1L1 is a known PXR target gene, we identified a DR-1-type PXR-response element in the MTP promoter and established MTP as a potentially novel transcriptional target of PXR. Quetiapine's effects on PXR-mediated gene expression and cholesterol uptake were also confirmed in cultured murine enteroids and human intestinal cells. Our findings suggest a potential role of PXR in mediating adverse effects of quetiapine in humans and provide mechanistic insights for certain atypical antipsychotic-associated dyslipidemia.

14.
J Hepatol ; 70(5): 930-940, 2019 05.
Article in English | MEDLINE | ID: mdl-30677459

ABSTRACT

BACKGROUND & AIMS: The most prescribed non-nucleoside reverse transcriptase inhibitor, efavirenz, has been associated with elevated risk of dyslipidemia and hepatic steatosis in HIV-infected patients but the underlying mechanisms remain elusive. Herein, we investigated the role of pregnane X receptor (PXR) in mediating the adverse effects of efavirenz on lipid homeostasis. METHODS: Cell-based reporter assays, primary cell culture, and multiple mouse models including conditional knockout and humanized mice were combined to study the impact of efavirenz on PXR activities and lipid homeostasis in vitro and in vivo. A novel liver-specific Pxr knockout mouse model was also generated to determine the contribution of hepatic PXR signaling to efavirenz-elicited dyslipidemia and hepatic steatosis. RESULTS: We found that efavirenz is a potent PXR-selective agonist that can efficiently activate PXR and induce its target gene expression in vitro and in vivo. Treatment with efavirenz-induced hypercholesterolemia and hepatic steatosis in mice but deficiency of hepatic PXR abolished these adverse effects. Interestingly, efavirenz-mediated PXR activation regulated the expression of several key hepatic lipogenic genes including fatty acid transporter CD36 and cholesterol biosynthesis enzyme squalene epoxidase (SQLE), leading to increased lipid uptake and cholesterol biosynthesis in hepatic cells. While CD36 is a known PXR target gene, we identified a DR-2-type of PXR-response element in the SQLE promoter and established SQLE as a direct transcriptional target of PXR. Since PXR exhibits considerable differences in its pharmacology across species, we also confirmed these findings in PXR-humanized mice and human primary hepatocytes. CONCLUSIONS: The widely prescribed antiretroviral drug efavirenz induces hypercholesterolemia and hepatic steatosis by activating PXR signaling. Activation of PXR should be taken into consideration for patients undergoing long-term treatment with PXR agonistic antiretroviral drugs. LAY SUMMARY: Efavirenz is widely prescribed for HIV-infected patients but has some side effects. It can increase lipid levels in patients' blood and liver. Here we show that efavirenz can activate a unique liver protein called PXR which mediates the adverse effects of efavirenz on lipid levels in mouse models.


Subject(s)
Benzoxazines/adverse effects , Fatty Liver/chemically induced , Hypercholesterolemia/chemically induced , Pregnane X Receptor/agonists , Reverse Transcriptase Inhibitors/adverse effects , Alkynes , Animals , CD36 Antigens/physiology , Cholesterol/biosynthesis , Cyclopropanes , Hepatocytes/metabolism , Humans , Mice , Mice, Inbred C57BL , Pregnane X Receptor/physiology , Signal Transduction/physiology , Squalene Monooxygenase/physiology
15.
Arterioscler Thromb Vasc Biol ; 38(7): 1468-1478, 2018 07.
Article in English | MEDLINE | ID: mdl-29724817

ABSTRACT

OBJECTIVE: The Wnt/ß-catenin signaling is an ancient and evolutionarily conserved pathway that regulates essential aspects of cell differentiation, proliferation, migration and polarity. Canonical Wnt/ß-catenin signaling has also been implicated in the pathogenesis of atherosclerosis. Macrophage is one of the major cell types involved in the initiation and progression of atherosclerosis, but the role of macrophage ß-catenin in atherosclerosis remains elusive. This study aims to investigate the impact of ß-catenin expression on macrophage functions and atherosclerosis development. APPROACH AND RESULTS: To investigate the role of macrophage canonical Wnt/ß-catenin signaling in atherogenesis, we generated ß-cateninΔmyeLDLR-/- mice (low-density lipoprotein receptor-deficient mice with myeloid-specific ß-catenin deficiency). As expected, deletion of ß-catenin decreased macrophage adhesion and migration properties in vitro. However, deficiency of ß-catenin significantly increased atherosclerotic lesion areas in the aortic root of LDLR-/- (low-density lipoprotein receptor-deficient) mice without affecting the plasma lipid levels and atherosclerotic plaque composition. Mechanistic studies revealed that ß-catenin can regulate activation of STAT (signal transducer and activator of transcription) pathway in macrophages, and ablation of ß-catenin resulted in STAT3 downregulation and STAT1 activation, leading to elevated macrophage inflammatory responses and increased atherosclerosis. CONCLUSIONS: This study demonstrates a critical role of myeloid ß-catenin expression in atherosclerosis by modulating macrophage inflammatory responses.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Macrophages/metabolism , Plaque, Atherosclerotic , Receptors, LDL/deficiency , beta Catenin/deficiency , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Adhesion , Cell Movement , Diet, High-Fat , Disease Models, Animal , Disease Progression , Lipids/blood , Macrophages/pathology , Male , Mice , Mice, Knockout , RAW 264.7 Cells , Receptors, LDL/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Time Factors , Wnt Signaling Pathway , beta Catenin/genetics
16.
Endocrinology ; 159(4): 1595-1608, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29425287

ABSTRACT

Bisphenol A (BPA) is a base chemical used extensively in numerous consumer products, and human exposure to BPA is ubiquitous. Higher BPA exposure has been associated with an increased risk of atherosclerosis and cardiovascular disease (CVD) in multiple human population-based studies. However, the underlying mechanisms responsible for the associations remain elusive. We previously reported that BPA activates the xenobiotic receptor pregnane X receptor (PXR), which has proatherogenic effects in animal models. Because BPA is a potent agonist for human PXR but does not affect rodent PXR activity, a suitable PXR-humanized apolipoprotein E-deficient (huPXR•ApoE-/-) mouse model was developed to study BPA's atherogenic effects. Chronic BPA exposure increased atherosclerosis in the huPXR•ApoE-/- mice. We report that BPA exposure can also activate human PXR signaling in the heart tubes of huPXR•ApoE-/- embryos, and perinatal BPA exposure exacerbated atherosclerosis in adult male huPXR•ApoE-/- offspring. However, atherosclerosis development in female offspring was not affected by perinatal BPA exposure. Perinatal BPA exposure did not affect plasma lipid levels but increased aortic and atherosclerotic lesional fatty acid transporter CD36 expression in male huPXR•ApoE-/- offspring. Mechanistically, PXR epigenetically regulated CD36 expression by increasing H3K4me3 levels and decreasing H3K27me3 levels in the CD36 promoter in response to perinatal BPA exposure. The findings from the present study contribute to our understanding of the association between BPA exposure and increased atherosclerosis or CVD risk in humans, and activation of human PXR should be considered for future BPA risk assessment.


Subject(s)
Aorta/drug effects , Atherosclerosis/metabolism , Benzhydryl Compounds/administration & dosage , Environmental Pollutants/administration & dosage , Phenols/administration & dosage , Receptors, Steroid/metabolism , Signal Transduction/drug effects , Animals , Aorta/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cholesterol/blood , Gene Expression Regulation/drug effects , Male , Mice , Mice, Knockout , Pregnane X Receptor
17.
JCI Insight ; 3(2)2018 01 25.
Article in English | MEDLINE | ID: mdl-29367460

ABSTRACT

Mesenchymal stem cells (MSCs) can give rise to both adipocytes and osteoblasts, but the molecular mechanisms underlying MSC fate determination remain poorly understood. IκB kinase ß (IKKß), a central coordinator of inflammation and immune responses through activation of NF-κB, has been implicated as a critical molecular link between obesity and metabolic disorders. Here, we show that IKKß can reciprocally regulate adipocyte and osteoblast differentiation of murine and human MSCs through an NF-κB-independent mechanism. IKKß is a ß-catenin kinase that phosphorylates the conserved degron motif of ß-catenin to prime it for ß-TrCP-mediated ubiquitination and degradation, thereby increasing adipogenesis and inhibiting osteogenesis in MSCs. Animal studies demonstrated that deficiency of IKKß in BM mesenchymal stromal cells increased bone mass and decreased BM adipocyte formation in adult mice. In humans, IKKß expression in adipose tissue was also positively associated with increased adiposity and elevated ß-catenin phosphorylation. These findings suggest IKKß as a key molecular switch that regulates MSC fate, and they provide potentially novel mechanistic insights into the understanding of the cross-regulation between the evolutionarily conserved IKKß and Wnt/ß-catenin signaling pathways. The IKKß-Wnt axis we uncovered may also have important implications for development, homeostasis, and disease pathogenesis.


Subject(s)
Cell Differentiation/physiology , I-kappa B Kinase/metabolism , Mesenchymal Stem Cells/metabolism , Obesity/pathology , beta Catenin/metabolism , Abdominal Fat/pathology , Adipocytes/physiology , Adipogenesis/physiology , Adult , Animals , Biopsy , Cells, Cultured , Female , Humans , I-kappa B Kinase/analysis , I-kappa B Kinase/genetics , Male , Mice , Mice, Transgenic , Middle Aged , Models, Animal , Obesity/blood , Osteoblasts/physiology , Osteogenesis/physiology , Phosphorylation/physiology , Primary Cell Culture , Proteolysis , Ubiquitination/physiology , Wnt Signaling Pathway/physiology , beta-Transducin Repeat-Containing Proteins/metabolism
18.
Knee Surg Relat Res ; 29(4): 307-315, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29046046

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the anatomical similarity of three-dimensional (3D) morphometric parameters between right and left knees. MATERIALS AND METHODS: Ten fresh-frozen paired cadaveric knees were tested. Following dissection, footprint areas of the anterior cruciate ligament (ACL) and posterior cruciate ligament (PCL) were measured. Surface scanning was performed using a 3D scanner. Scanned data were reproduced and morphometric parameters were measured on specialized software. After making mirror models, we compared footprint center positions of the ACL and PCL of both sides and calculated the average deviation of 3D alignment between the right- and left-side models. RESULTS: No significant side-to-side differences were found in any morphometric parameters. Bony shapes displayed a side-to-side difference of 〈1 mm. Distal femoral and proximal tibial volumes did not present side-to-side differences, either; the average 3D deviations of alignment between the right and left sides were 0.8±0.4/1.1±0.6 mm (distal femur/proximal tibia). Center-to-center distances between the right and left ACL footprints were 2.6/2.7 mm (femur/tibia) for the anteromedial bundle and 2.4/2.8 mm for the posterolateral bundle. They were 1.9/1.5 mm for the anterolateral bundle and 2.2/1.8 mm for the posteromedial bundle of the PCL. CONCLUSIONS: There was a remarkable 3D morphometric similarity between right and left knees. Our results might support the concept of obtaining morphologic reference data from the uninvolved contralateral knee.

19.
Am J Sports Med ; 45(10): 2345-2354, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28581816

ABSTRACT

BACKGROUND: There is considerable debate on the recovery of rotator cuff muscle atrophy after rotator cuff repair. PURPOSE: To evaluate the serial changes in supraspinatus muscle volume after rotator cuff repair by using semiautomatic segmentation software and to determine the relationship with functional outcomes. STUDY DESIGN: Case series; Level of evidence, 4. METHODS: Seventy-four patients (mean age, 62.8 ± 8.8 years) who underwent arthroscopic rotator cuff repair and obtained 3 consecutive (preoperatively, immediately postoperatively, and later postoperatively [≥1 year postoperatively]) magnetic resonance imaging (MRI) scans having complete Y-views were included. We generated a 3-dimensional (3D) reconstructed model of the supraspinatus muscle by using in-house semiautomatic segmentation software (ITK-SNAP) and calculated both the 2-dimensional (2D) cross-sectional area and 3D volume of the muscle in 3 different views (Y-view, 1 cm medial to the Y-view [Y+1 view], and 2 cm medial to the Y-view [Y+2 view]) at the 3 time points. The area and volume changes at each time point were evaluated according to repair integrity. Later postoperative volumes were compared with immediately postoperative volumes, and their relationship with various clinical factors and the effect of higher volume increases on range of motion, muscle power, and visual analog scale pain and American Shoulder and Elbow Surgeons scores were evaluated. RESULTS: The interrater reliabilities were excellent for all measurements. Areas and volumes increased immediately postoperatively as compared with preoperatively; however, only volumes on the Y+1 view and Y+2 view significantly increased later postoperatively as compared with immediately postoperatively ( P < .05). There were 9 patients with healing failure, and area and volume changes were significantly less later postoperatively compared with immediately postoperatively at all measurement points in these patients ( P < .05). After omitting the patients with healing failure, volume increases later postoperatively became more prominent ( P < .05) in the order of the Y+2 view, Y+1 view, and Y-view. Volume increases were higher in patients who healed successfully with larger tears ( P = .040). Higher volume increases were associated only with an increase in abduction power ( P = .029) and not with other outcomes. CONCLUSION: The supraspinatus muscle volume increased immediately postoperatively and continuously for at least 1 year after surgery. The increase was evident in patients who had larger tears and healed successfully and when measured toward the more medial portion of the supraspinatus muscle. The volume increases were associated with an increase in shoulder abduction power.


Subject(s)
Muscles/physiopathology , Rotator Cuff Injuries/surgery , Adult , Aged , Aged, 80 and over , Arthroplasty , Arthroscopy , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Postoperative Period , Range of Motion, Articular/physiology , Rotator Cuff/diagnostic imaging , Rotator Cuff/physiopathology , Rotator Cuff/surgery , Rotator Cuff Injuries/diagnostic imaging , Rotator Cuff Injuries/physiopathology , Treatment Outcome
20.
Comput Methods Programs Biomed ; 140: 165-174, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28254072

ABSTRACT

BACKGROUND AND OBJECTIVES: With significant increase in the number of people suffering from shoulder problems, the automatic image segmentation of the supraspinatus (one of the shoulder muscles) has become necessary for efficient and deliberate diagnosis and surgery. In this study, we developed an automatic segmentation method to extract the three-dimensional (3D) configuration of the supraspinatus, and we compared our segmentation results with reference segmentations obtained by experts. METHODS: We developed a two-stage active contour segmentation method using the level sets approach to automatically extract the supraspinatus configuration. In the first stage, a trial segmentation based on intensity and an internal shape fitting technique were performed. In the second stage, the undesired image portions of the trial segmentation were automatically identified by comparing the trial segmentation with the fitted shape, and then corrected by forcing the contour to stop evolution in the over-segmented region and pass through undesired edges in the under-segmented region. RESULTS: The proposed method was found to provide highly accurate results when compared with the reference segmentations. This comparison was made on the basis of four measurements: accuracy (0.995 ± 0.001), Dice similarity coefficients (0.951 ± 0.011), average distance (0.440 ± 0.086mm), and maximal distance (3.045 ± 0.433mm). The proposed method could generate regular surfaces of the 3D supraspinatus. CONCLUSIONS: The proposed automatic segmentation method provides a patient-specific tool to accurately extract the 3D configuration of the supraspinatus.


Subject(s)
Magnetic Resonance Imaging/methods , Rotator Cuff/diagnostic imaging , Automation , Humans , Imaging, Three-Dimensional
SELECTION OF CITATIONS
SEARCH DETAIL
...