1
|
Stapenhorst França F, Gensel JC. Redefining macrophage phenotypes after spinal cord injury: An open data approach. Exp Neurol 2025; 388:115222. [PMID: 40113007 DOI: 10.1016/j.expneurol.2025.115222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
Spinal cord injury (SCI) triggers intraspinal inflammation through an influx of blood-derived inflammatory cells such as neutrophils and monocyte-derived macrophages. Macrophages play a complex role in SCI pathophysiology ranging from potentiating secondary injury to facilitating recovery and wound healing. In vitro, macrophages have been classified as having a pro-inflammatory, M1 phenotype, or a regenerative, M2 phenotype. In vivo, however, studies suggest that macrophages exist in a spectrum of phenotypes and can shift from one phenotype to another. Single-cell RNA sequencing (scRNA-seq) allows us to assess immune cell heterogeneity in the spinal cord after injury, and several groups have created publicly available datasets containing valuable data for further exploration. In this study, we compared three different scRNA-seq datasets and analyzed macrophage heterogeneity after SCI based on cell clustering according to gene expression profiles. We analyzed data from 7 days post injury (dpi) in young female mice that received a mid-thoracic SCI contusion. Using the Seurat pipeline, we clustered cells, subsetted macrophages from microglia and other myeloid cells, and identified different macrophage populations. Using SingleR as a cross-dataset cluster comparison tool, we identified similarities in macrophage populations across datasets. To confirm and refine this analysis, we analyzed the top 10 differentially expressed genes for each population in each dataset. Most clusters identified in the SingleR analysis were confirmed to have a unique genetic signature and were consistently present in all datasets analyzed. Taken together, four distinct macrophage populations were consistently identified after SCI at 7 dpi in three datasets from independent research teams. Our identification of biologically conserved macrophage populations after SCI using an unbiased approach highlights the power of data sharing and open data in redefining macrophage heterogeneity.
Collapse
Affiliation(s)
- Fernanda Stapenhorst França
- Spinal Cord and Brain Injury Research Center and Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States.
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center and Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
2
|
Kanda D, Takumi T, Arikawa R, Anzaki K, Sonoda T, Ohmure K, Fukumoto D, Tokushige A, Ohishi M. Secondary rotational atherectomy is associated with reduced occurrence of prolonged ST-segment elevation following ablation. Intern Emerg Med 2023; 18:1995-2002. [PMID: 37566359 PMCID: PMC10543138 DOI: 10.1007/s11739-023-03385-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023]
Abstract
Elevation of the ST segment after percutaneous coronary intervention (PCI) using rotational atherectomy (RA) for severely calcified lesions often persists after disappearance of the slow-flow phenomenon on angiography. We investigated clinical factors relevant to prolonged ST-segment elevation following RA among 152 patients with stable angina undergoing elective PCI. PCI procedures were divided into two strategies, RA without (primary RA strategy) or with (secondary RA strategy) balloon dilatation before RA. Incidence of prolonged ST-segment elevation after disappearance of slow-flow phenomenon was higher in the 56 patients with primary RA strategy (13%) than in the 96 patients with secondary RA strategy (3%, p = 0.039). Univariate logistic regression analysis showed levels of low-density lipoprotein cholesterol (LDL-C) (odds ratio [OR] 0.95, 95% confidence interval [CI] 0.93-0.99; p = 0.013), levels of triglycerides (OR 0.97, 95%CI 0.94-0.99; p = 0.040), and secondary RA strategy (OR 0.23, 95% CI 0.05-0.85; p = 0.028) were inversely associated with occurrence of prolonged ST-segment elevation following ablation. However, hemodialysis, diabetes mellitus, left-ventricular ejection fraction, lesion length ≥ 20 mm, and burr size did not show significant associations. Multivariate logistic regression analysis modeling revealed that secondary RA strategy was significantly associated with the occurrence of prolonged ST-segment elevation (Model 1: OR 0.24, 95% CI 0.05-0.95, p = 0.042; Model 2: OR 0.17, 95% CI 0.03-0.68, p = 0.018; Model 3: OR 0.21, 95% CI 0.03-0.87, p = 0.041) even after adjusting for levels of LDL-C and triglycerides. Secondary RA strategy may be useful to reduce the occurrence of prolonged ST-segment elevation following RA.
Collapse
Affiliation(s)
- Daisuke Kanda
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8520, Japan.
| | - Takuro Takumi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8520, Japan
| | - Ryo Arikawa
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8520, Japan
| | - Kazuhiro Anzaki
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8520, Japan
| | - Takeshi Sonoda
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8520, Japan
| | - Kenta Ohmure
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8520, Japan
| | - Daichi Fukumoto
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8520, Japan
| | - Akihiro Tokushige
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8520, Japan
| | - Mitsuru Ohishi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8520, Japan
| |
Collapse
|
3
|
Crosstalk between neurological, cardiovascular, and lifestyle disorders: insulin and lipoproteins in the lead role. Pharmacol Rep 2022; 74:790-817. [PMID: 36149598 DOI: 10.1007/s43440-022-00417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/03/2022] [Accepted: 09/08/2022] [Indexed: 10/14/2022]
Abstract
Insulin resistance and impaired lipoprotein metabolism contribute to a plethora of metabolic and cardiovascular disorders. These alterations have been extensively linked with poor lifestyle choices, such as consumption of a high-fat diet, smoking, stress, and a redundant lifestyle. Moreover, these are also known to increase the co-morbidity of diseases like Type 2 diabetes mellitus and atherosclerosis. Under normal physiological conditions, insulin and lipoproteins exert a neuroprotective role in the central nervous system. However, the tripping of balance between the periphery and center may alter the normal functioning of the brain and lead to neurological disorders such as Alzheimer's disease, Parkinson's disease, stroke, depression, and multiple sclerosis. These neurological disorders are further characterized by certain behavioral and molecular changes that show consistent overlap with alteration in insulin and lipoprotein signaling pathways. Therefore, targeting these two mechanisms not only reveals a way to manage the co-morbidities associated with the circle of the metabolic, central nervous system, and cardiovascular disorders but also exclusively work as a disease-modifying therapy for neurological disorders. In this review, we summarize the role of insulin resistance and lipoproteins in the progression of various neurological conditions and discuss the therapeutic options currently in the clinical pipeline targeting these two mechanisms; in addition, challenges faced in designing these therapeutic approaches have also been touched upon briefly.
Collapse
|
4
|
Macrophages take up VLDL-sized emulsion particles through caveolae-mediated endocytosis and excrete part of the internalized triglycerides as fatty acids. PLoS Biol 2022; 20:e3001516. [PMID: 36026438 PMCID: PMC9455861 DOI: 10.1371/journal.pbio.3001516] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/08/2022] [Accepted: 07/09/2022] [Indexed: 11/26/2022] Open
Abstract
Triglycerides are carried in the bloodstream as part of very low-density lipoproteins (VLDLs) and chylomicrons, which represent the triglyceride-rich lipoproteins. Triglyceride-rich lipoproteins and their remnants contribute to atherosclerosis, possibly by carrying remnant cholesterol and/or by exerting a proinflammatory effect on macrophages. Nevertheless, little is known about how macrophages process triglyceride-rich lipoproteins. Here, using VLDL-sized triglyceride-rich emulsion particles, we aimed to study the mechanism by which VLDL triglycerides are taken up, processed, and stored in macrophages. Our results show that macrophage uptake of VLDL-sized emulsion particles is dependent on lipoprotein lipase (LPL) and requires the lipoprotein-binding C-terminal domain but not the catalytic N-terminal domain of LPL. Subsequent internalization of VLDL-sized emulsion particles by macrophages is carried out by caveolae-mediated endocytosis, followed by triglyceride hydrolysis catalyzed by lysosomal acid lipase. It is shown that STARD3 is required for the transfer of lysosomal fatty acids to the ER for subsequent storage as triglycerides, while NPC1 likely is involved in promoting the extracellular efflux of fatty acids from lysosomes. Our data provide novel insights into how macrophages process VLDL triglycerides and suggest that macrophages have the remarkable capacity to excrete part of the internalized triglycerides as fatty acids. How do macrophages take up and process very low density lipoprotein (VLDL) particles? This study reveals that endocytic uptake of VLDLs depends on lipoprotein lipase and caveolae; internalized VLDLs are then processed by lysosomes, and the lipids are hydrolyzed and translocated to the ER for storage as triglycerides.
Collapse
|
5
|
Takahashi M, Yamamuro D, Wakabayashi T, Takei A, Takei S, Nagashima S, Okazaki H, Ebihara K, Yagyu H, Takayanagi Y, Onaka T, Goldberg IJ, Ishibashi S. Loss of myeloid lipoprotein lipase exacerbates adipose tissue fibrosis with collagen VI deposition and hyperlipidemia in leptin-deficient obese mice. J Biol Chem 2022; 298:102322. [PMID: 35926714 PMCID: PMC9440390 DOI: 10.1016/j.jbc.2022.102322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/20/2022] Open
Abstract
During obesity, tissue macrophages increase in number and become proinflammatory, thereby contributing to metabolic dysfunction. Lipoprotein lipase (LPL), which hydrolyzes triglyceride in lipoproteins, is secreted by macrophages. However, the role of macrophage-derived LPL in adipose tissue remodeling and lipoprotein metabolism is largely unknown. To clarify these issues, we crossed leptin-deficient Lepob/ob mice with mice lacking the Lpl gene in myeloid cells (Lplm−/m−) to generate Lplm−/m−;Lepob/ob mice. We found the weight of perigonadal white adipose tissue (WAT) was increased in Lplm−/m−;Lepob/ob mice compared with Lepob/ob mice due to substantial accumulation of both adipose tissue macrophages and collagen that surrounded necrotic adipocytes. In the fibrotic epidydimal WAT of Lplm−/m−;Lepob/ob mice, we observed an increase in collagen VI and high mobility group box 1, while α-smooth muscle cell actin, a marker of myofibroblasts, was almost undetectable, suggesting that the adipocytes were the major source of the collagens. Furthermore, the adipose tissue macrophages from Lplm−/m−;Lepob/ob mice showed increased expression of genes related to fibrosis and inflammation. In addition, we determined Lplm−/m−;Lepob/ob mice were more hypertriglyceridemic than Lepob/ob mice. Lplm−/m−;Lepob/ob mice also showed slower weight gain than Lepob/ob mice, which was primarily due to reduced food intake. In conclusion, we discovered that the loss of myeloid Lpl led to extensive fibrosis of perigonadal WAT and hypertriglyceridemia. In addition to illustrating an important role of macrophage LPL in regulation of circulating triglyceride levels, these data show that macrophage LPL protects against fibrosis in obese adipose tissues.
Collapse
Affiliation(s)
- Manabu Takahashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan.
| | - Daisuke Yamamuro
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Tetsuji Wakabayashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Akihito Takei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Shoko Takei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Shuichi Nagashima
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Hiroaki Okazaki
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Ken Ebihara
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Hiroaki Yagyu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Yuki Takayanagi
- Division of Brain and Neurophysiology, Department of Physiology, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Ira J Goldberg
- NYU-Langone Medical Center, 435 East 30(th) Street, SB617, New York, NY, 10016
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan.
| |
Collapse
|
6
|
Zhang BH, Yin F, Qiao YN, Guo SD. Triglyceride and Triglyceride-Rich Lipoproteins in Atherosclerosis. Front Mol Biosci 2022; 9:909151. [PMID: 35693558 PMCID: PMC9174947 DOI: 10.3389/fmolb.2022.909151] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/06/2022] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is still the leading cause of death globally, and atherosclerosis is the main pathological basis of CVDs. Low-density lipoprotein cholesterol (LDL-C) is a strong causal factor of atherosclerosis. However, the first-line lipid-lowering drugs, statins, only reduce approximately 30% of the CVD risk. Of note, atherosclerotic CVD (ASCVD) cannot be eliminated in a great number of patients even their LDL-C levels meet the recommended clinical goals. Previously, whether the elevated plasma level of triglyceride is causally associated with ASCVD has been controversial. Recent genetic and epidemiological studies have demonstrated that triglyceride and triglyceride-rich lipoprotein (TGRL) are the main causal risk factors of the residual ASCVD. TGRLs and their metabolites can promote atherosclerosis via modulating inflammation, oxidative stress, and formation of foam cells. In this article, we will make a short review of TG and TGRL metabolism, display evidence of association between TG and ASCVD, summarize the atherogenic factors of TGRLs and their metabolites, and discuss the current findings and advances in TG-lowering therapies. This review provides information useful for the researchers in the field of CVD as well as for pharmacologists and clinicians.
Collapse
Affiliation(s)
| | | | - Ya-Nan Qiao
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
7
|
Saraswathi V, Kumar N, Ai W, Gopal T, Bhatt S, Harris EN, Talmon GA, Desouza CV. Myristic Acid Supplementation Aggravates High Fat Diet-Induced Adipose Inflammation and Systemic Insulin Resistance in Mice. Biomolecules 2022; 12:739. [PMID: 35740864 PMCID: PMC9220168 DOI: 10.3390/biom12060739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 12/12/2022] Open
Abstract
Saturated fatty acids (SFAs) are considered to be detrimental to human health. One of the SFAs, myristic acid (MA), is known to exert a hypercholesterolemic effect in mice as well as humans. However, its effects on altering adipose tissue (AT) inflammation and systemic insulin resistance (IR) in obesity are still unclear. Here, we sought to determine the effects of a high fat (HF) diet supplemented with MA on obesity-associated metabolic disorders in mice. Wild-type C57BL/6 mice were fed a HF diet in the presence or absence of 3% MA for 12 weeks. Plasma lipids, plasma adipokines, AT inflammation, systemic IR, glucose homeostasis, and hepatic steatosis were assessed. The body weight and visceral adipose tissue (VAT) mass were significantly higher in mice receiving the HF+MA diet compared to HF diet-fed controls. Plasma total cholesterol levels were marginally increased in HF+MA-fed mice compared to controls. Fasting blood glucose was comparable between HF and HF+MA-fed mice. Interestingly, the plasma insulin and HOMA-IR index, a measure of insulin resistance, were significantly higher in HF+MA-fed mice compared to HF controls. Macrophage and inflammatory markers were significantly elevated in the AT and AT-derived stromal vascular cells upon MA feeding. Moreover, the level of circulating resistin, an adipokine promoting insulin resistance, was significantly higher in HF+MA-fed mice compared with HF controls. The insulin tolerance test revealed that the IR was higher in mice receiving the MA supplementation compared to HF controls. Moreover, the glucose tolerance test showed impairment in systemic glucose homeostasis in MA-fed mice. Analyses of liver samples showed a trend towards an increase in liver TG upon MA feeding. However, markers of oxidative stress and inflammation were reduced in the liver of mice fed an MA diet compared to controls. Taken together, our data suggest that chronic administration of MA in diet exacerbates obesity-associated insulin resistance and this effect is mediated in part, via increased AT inflammation and increased secretion of resistin.
Collapse
Affiliation(s)
- Viswanathan Saraswathi
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.K.); (W.A.); (T.G.); (S.B.); (C.V.D.)
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Narendra Kumar
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.K.); (W.A.); (T.G.); (S.B.); (C.V.D.)
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Weilun Ai
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.K.); (W.A.); (T.G.); (S.B.); (C.V.D.)
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Thiyagarajan Gopal
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.K.); (W.A.); (T.G.); (S.B.); (C.V.D.)
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Saumya Bhatt
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.K.); (W.A.); (T.G.); (S.B.); (C.V.D.)
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Edward N. Harris
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA;
| | - Geoffrey A. Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Cyrus V. Desouza
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.K.); (W.A.); (T.G.); (S.B.); (C.V.D.)
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Hypertriglyceridemia is a common dyslipidemia associated with an increased risk of cardiovascular disease and pancreatitis. Severe hypertriglyceridemia may sometimes be a monogenic condition. However, in the vast majority of patients, hypertriglyceridemia is due to the cumulative effect of multiple genetic risk variants along with lifestyle factors, medications, and disease conditions that elevate triglyceride levels. In this review, we will summarize recent progress in the understanding of the genetic basis of hypertriglyceridemia. RECENT FINDINGS More than 300 genetic loci have been identified for association with triglyceride levels in large genome-wide association studies. Studies combining the loci into polygenic scores have demonstrated that some hypertriglyceridemia phenotypes previously attributed to monogenic inheritance have a polygenic basis. The new genetic discoveries have opened avenues for the development of more effective triglyceride-lowering treatments and raised interest towards genetic screening and tailored treatments against hypertriglyceridemia. The discovery of multiple genetic loci associated with elevated triglyceride levels has led to improved understanding of the genetic basis of hypertriglyceridemia and opened new translational opportunities.
Collapse
Affiliation(s)
- Germán D. Carrasquilla
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Mærsk Building, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Malene Revsbech Christiansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Mærsk Building, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Tuomas O. Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Mærsk Building, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
9
|
Raposeiras-Roubin S, Rosselló X, Oliva B, Fernández-Friera L, Mendiguren JM, Andrés V, Bueno H, Sanz J, Martínez de Vega V, Abu-Assi E, Iñiguez A, Fernández-Ortiz A, Ibáñez B, Fuster V. Triglycerides and Residual Atherosclerotic Risk. J Am Coll Cardiol 2021; 77:3031-3041. [PMID: 34140107 PMCID: PMC8215641 DOI: 10.1016/j.jacc.2021.04.059] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/05/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Even when low-density lipoprotein-cholesterol (LDL-C) levels are lower than guideline thresholds, a residual risk of atherosclerosis remains. It is unknown whether triglyceride (TG) levels are associated with subclinical atherosclerosis and vascular inflammation regardless of LDL-C. OBJECTIVES This study sought to assess the association between serum TG levels and early atherosclerosis and vascular inflammation in apparently healthy individuals. METHODS An observational, longitudinal, and prospective cohort study, including 3,754 middle-aged individuals with low to moderate cardiovascular risk from the PESA (Progression of Early Subclinical Atherosclerosis) study who were consecutively recruited between June 2010 and February 2014, was conducted. Peripheral atherosclerotic plaques were assessed by 2-dimensional vascular ultrasound, and coronary artery calcification (CAC) was assessed by noncontrast computed tomography, whereas vascular inflammation was assessed by fluorine-18 fluorodeoxyglucose uptake on positron emission tomography. RESULTS Atherosclerotic plaques and CAC were observed in 58.0% and 16.8% of participants, respectively, whereas vascular inflammation was evident in 46.7% of evaluated participants. After multivariate adjustment, TG levels ≥150 mg/dl showed an association with subclinical noncoronary atherosclerosis (odds ratio [OR]: 1.35; 95% confidence interval [CI]: 1.08 to 1.68; p = 0.008). This association was significant for groups with high LDL-C (OR: 1.42; 95% CI: 1.11 to 1.80; p = 0.005) and normal LDL-C (OR: 1.85; 95% CI: 1.08 to 3.18; p = 0.008). No association was found between TG level and CAC score. TG levels ≥150 mg/dl were significantly associated with the presence of arterial inflammation (OR: 2.09; 95% CI: 1.29 to 3.40; p = 0.003). CONCLUSIONS In individuals with low to moderate cardiovascular risk, hypertriglyceridemia was associated with subclinical atherosclerosis and vascular inflammation, even in participants with normal LDL-C levels. (Progression of Early Subclinical Atherosclerosis [PESA]; NCT01410318).
Collapse
Affiliation(s)
- Sergio Raposeiras-Roubin
- National Center for Cardiovascular Research (CNIC), Madrid, Spain; Department of Cardiology, Álvaro Cunqueiro University Hospital, Vigo, Spain. https://twitter.com/Borjaibanez1
| | - Xavier Rosselló
- National Center for Cardiovascular Research (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red Consortium for Cardiovascular Diseases (CIBERCV), Madrid, Spain; Department of Cardiology, Son Espases University Hospital, Palma de Mallorca, Spain
| | - Belén Oliva
- National Center for Cardiovascular Research (CNIC), Madrid, Spain
| | - Leticia Fernández-Friera
- National Center for Cardiovascular Research (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red Consortium for Cardiovascular Diseases (CIBERCV), Madrid, Spain; Comprehensive Cardiovascular Diseases Center, HM Montepríncipe University Hospital, Madrid, Spain
| | | | - Vicente Andrés
- National Center for Cardiovascular Research (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red Consortium for Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Héctor Bueno
- National Center for Cardiovascular Research (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red Consortium for Cardiovascular Diseases (CIBERCV), Madrid, Spain; Health Research Institute, October 12 Hospital (imas12), Madrid, Spain
| | - Javier Sanz
- National Center for Cardiovascular Research (CNIC), Madrid, Spain; Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Vicente Martínez de Vega
- National Center for Cardiovascular Research (CNIC), Madrid, Spain; Quirón University Hospital, Madrid, Spain
| | - Emad Abu-Assi
- Department of Cardiology, Álvaro Cunqueiro University Hospital, Vigo, Spain; Centro de Investigación Biomédica en Red Consortium for Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Andrés Iñiguez
- Department of Cardiology, Álvaro Cunqueiro University Hospital, Vigo, Spain; Centro de Investigación Biomédica en Red Consortium for Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Antonio Fernández-Ortiz
- National Center for Cardiovascular Research (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red Consortium for Cardiovascular Diseases (CIBERCV), Madrid, Spain; Health Research Institute Clínico San Carlos Hospital, Complutense University, Madrid, Spain
| | - Borja Ibáñez
- National Center for Cardiovascular Research (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red Consortium for Cardiovascular Diseases (CIBERCV), Madrid, Spain; Health Research Institute, Fundación Jiménez Díaz University Hospital, Madrid, Spain.
| | - Valentin Fuster
- National Center for Cardiovascular Research (CNIC), Madrid, Spain; Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
10
|
Kluge S, Schubert M, Börmel L, Lorkowski S. The vitamin E long-chain metabolite α-13'-COOH affects macrophage foam cell formation via modulation of the lipoprotein lipase system. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158875. [PMID: 33421592 DOI: 10.1016/j.bbalip.2021.158875] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/01/2020] [Accepted: 01/02/2021] [Indexed: 12/14/2022]
Abstract
The α-tocopherol-derived long-chain metabolite (α-LCM) α-13'-carboxychromanol (α-13'-COOH) is formed via enzymatic degradation of α-tocopherol (α-TOH) in the liver. In the last decade, α-13'-COOH has emerged as a new regulatory metabolite revealing more potent or even different effects compared with its vitamin precursor α-TOH. The detection of α-13'-COOH in human serum has further strengthened the concept of its physiological relevance as a potential regulatory molecule. Here, we present a new facet on the interaction of α-13'-COOH with macrophage foam cell formation. We found that α-13'-COOH (5 μM) increases angiopoietin-like 4 (ANGPTL4) mRNA expression in human THP-1 macrophages in a time- and dose-dependent manner, while α-TOH (100 μM) showed no effects. Interestingly, the mRNA level of lipoprotein lipase (LPL) was not influenced by α-13'-COOH, but α-TOH treatment led to a reduction of LPL mRNA expression. Both compounds also revealed different effects on protein level: while α-13'-COOH reduced the secreted amount of LPL protein via induction of ANGPTL4 cleavage, i.e. activation, the secreted amount of LPL in the α-TOH-treated samples was diminished due to the inhibition of mRNA expression. In line with this, both compounds reduced the catalytic activity of LPL. However, α-13'-COOH but not α-TOH attenuated VLDL-induced lipid accumulation by 35%. In conclusion, only α-13'-COOH revealed possible antiatherogenic effects due to the reduction of VLDL-induced foam cell formation in THP-1 macrophages. Our results provide further evidence for the role of α-13'-COOH as a functional metabolite of its vitamin E precursor.
Collapse
Affiliation(s)
- Stefan Kluge
- Institute of Nutritional Sciences, Friedrich Schiller University, Jena, Germany
| | - Martin Schubert
- Institute of Nutritional Sciences, Friedrich Schiller University, Jena, Germany
| | - Lisa Börmel
- Institute of Nutritional Sciences, Friedrich Schiller University, Jena, Germany
| | - Stefan Lorkowski
- Institute of Nutritional Sciences, Friedrich Schiller University, Jena, Germany; Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany.
| |
Collapse
|
11
|
Lauric Acid versus Palmitic Acid: Effects on Adipose Tissue Inflammation, Insulin Resistance, and Non-Alcoholic Fatty Liver Disease in Obesity. BIOLOGY 2020; 9:biology9110346. [PMID: 33105887 PMCID: PMC7690582 DOI: 10.3390/biology9110346] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 12/21/2022]
Abstract
Simple Summary The aim of this study was to compare the effect of palmitic acid (PA), a long-chain fatty acid, and lauric acid (LA), a medium-chain fatty acid, on obesity-related metabolic disorders. We used a mouse model of diet-induced obesity and fed them a modified high fat diet supplemented with 3% PA or LA for 12 wk. An LA diet led to an increase in visceral fat mass with a reduction in inflammation compared to the PA diet. We also noted that PA significantly increased systemic insulin resistance whereas LA showed only a trend towards an increase compared to lean control mice. The expression of a protein involved in muscle glucose uptake was higher in LA-treated mice compared to the PA-treated group, indicating improved muscle glucose uptake in LA-fed mice. Analysis of liver samples showed that hepatic steatosis was higher in both PA and LA-fed mice compared to lean controls. Markers of liver inflammation were not altered significantly in mice receiving PA or LA. Our data suggest that compared to PA, LA exerts less adverse effects on metabolic disorders and this could be due to the differential effects of these fatty acids in fat and muscle. Abstract Coconut oil, rich in medium-chain saturated fatty acids (MCSFA), in particular, lauric acid (LA), is known to exert beneficial metabolic effects. Although LA is the most abundant saturated fatty acid in coconut oil, the specific role of LA in altering obesity-related metabolic disorders remains unknown. Here, we examined the effects of supplementing a high fat (HF) diet with purified LA on obesity-associated metabolic derangements in comparison with palmitic acid (PA), a long-chain saturated fatty acid. Male C57BL/6 mice were fed a control chow diet (CD) or an HF diet supplemented with 3% LA (HF + LA) or PA (HF + PA) for 12 wk. Markers of adipose tissue (AT) inflammation, systemic insulin resistance (IR), and hepatic steatosis, were assessed. The body weight and total fat mass were significantly higher in both HF + LA and HF + PA diet-fed groups compared to CD controls. However, the visceral adipose tissue (VAT) mass was significantly higher (p < 0.001) in HF + LA-fed mice compared to both CD as well as HF + PA-fed mice. Interestingly, markers of AT inflammation were promoted to a lesser extent in HF + LA-fed mice compared to HF + PA-fed mice. Thus, immunohistochemical analysis of VAT showed an increase in MCP-1 and IL-6 staining in HF + PA-fed mice but not in HF + LA-fed mice compared to CD controls. Further, the mRNA levels of macrophage and inflammatory markers were significantly higher in HF + PA-fed mice (p < 0.001) whereas these markers were increased to a lesser extent in HF + LA-fed group. Of note, the insulin tolerance test revealed that IR was significantly increased only in HF + PA-fed mice but not in HF + LA-fed group compared to CD controls. While liver triglycerides were increased significantly in both HF + PA and HF + LA-fed mice, liver weight and plasma markers of liver injury such as alanine aminotransferase and aspartate aminotransferase were increased significantly only in HF + PA-fed mice but not in HF + LA-fed mice. Taken together, our data suggest that although both LA and PA increased AT inflammation, systemic IR, and liver injury, the extent of metabolic derangements caused by LA was less compared to PA in the setting of high fat feeding.
Collapse
|
12
|
Usui N, Iwata K, Miyachi T, Takagai S, Wakusawa K, Nara T, Tsuchiya KJ, Matsumoto K, Kurita D, Kameno Y, Wakuda T, Takebayashi K, Iwata Y, Fujioka T, Hirai T, Toyoshima M, Ohnishi T, Toyota T, Maekawa M, Yoshikawa T, Maekawa M, Nakamura K, Tsujii M, Sugiyama T, Mori N, Matsuzaki H. VLDL-specific increases of fatty acids in autism spectrum disorder correlate with social interaction. EBioMedicine 2020; 58:102917. [PMID: 32739868 PMCID: PMC7393524 DOI: 10.1016/j.ebiom.2020.102917] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Abnormalities of lipid metabolism contributing to the autism spectrum disorder (ASD) pathogenesis have been suggested, but the mechanisms are not fully understood. We aimed to characterize the lipid metabolism in ASD and to explore a biomarker for clinical evaluation. METHODS An age-matched case-control study was designed. Lipidomics was conducted using the plasma samples from 30 children with ASD compared to 30 typical developmental control (TD) children. Large-scale lipoprotein analyses were also conducted using the serum samples from 152 children with ASD compared to 122 TD children. Data comparing ASD to TD subjects were evaluated using univariate (Mann-Whitney test) and multivariate analyses (conditional logistic regression analysis) for main analyses using cofounders (diagnosis, sex, age, height, weight, and BMI), Spearman rank correlation coefficient, and discriminant analyses. FINDINGS Forty-eight significant metabolites involved in lipid biosynthesis and metabolism, oxidative stress, and synaptic function were identified in the plasma of ASD children by lipidomics. Among these, increased fatty acids (FAs), such as omega-3 (n-3) and omega-6 (n-6), showed correlations with clinical social interaction score and ASD diagnosis. Specific reductions of very-low-density lipoprotein (VLDL) and apoprotein B (APOB) in serum of ASD children also were found by large-scale lipoprotein analysis. VLDL-specific reduction in ASD was correlated with APOB, indicating VLDL-specific dyslipidaemia associated with APOB in ASD children. INTERPRETATION Our results demonstrated that the increases in FAs correlated positively with social interaction are due to VLDL-specific degradation, providing novel insights into the lipid metabolism underlying ASD pathophysiology. FUNDING This study was supported mainly by MEXT, Japan.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Research Center for Child Mental Development, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan; Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan; Center for Medical Research and Education, and Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| | - Keiko Iwata
- Research Center for Child Mental Development, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan; Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan
| | - Taishi Miyachi
- Department of Pediatrics, Nagoya City University Medical School, Aichi 467-8601, Japan
| | - Shu Takagai
- Department of Child and Adolescent Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Keisuke Wakusawa
- Department of Rehabilitation, Miyagi Children's Hospital, Miyagi 989-3126, Japan
| | - Takahiro Nara
- Department of Rehabilitation, Miyagi Children's Hospital, Miyagi 989-3126, Japan
| | - Kenji J Tsuchiya
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Kaori Matsumoto
- Graduate School of Psychology, Kanazawa Institute of Technology, Ishikawa 921-8054, Japan
| | - Daisuke Kurita
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Yosuke Kameno
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Tomoyasu Wakuda
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Kiyokazu Takebayashi
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Yasuhide Iwata
- Department of Psychiatry and Neurology, Fukude Nishi Hospital, Shizuoka 437-1216, Japan
| | - Toru Fujioka
- Research Center for Child Mental Development, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan; Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan
| | - Takaharu Hirai
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; Department of Community Health Nursing, School of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Manabu Toyoshima
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Tetsuo Ohnishi
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Tomoko Toyota
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Motoko Maekawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Masato Maekawa
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Kazuhiko Nakamura
- Department of Psychiatry, Hirosaki University School of Medicine, Aomori 036-8562, Japan
| | - Masatsugu Tsujii
- School of Contemporary Sociology, Chukyo University, Aichi 470-0393, Japan
| | - Toshiro Sugiyama
- Research Center for Child Mental Development, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan
| | - Norio Mori
- Department of Psychiatry and Neurology, Fukude Nishi Hospital, Shizuoka 437-1216, Japan
| | - Hideo Matsuzaki
- Research Center for Child Mental Development, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan; Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan.
| |
Collapse
|
13
|
Tekavec S, Sorčan T, Giacca M, Režen T. VLDL and HDL attenuate endoplasmic reticulum and metabolic stress in HL-1 cardiomyocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158713. [PMID: 32330663 DOI: 10.1016/j.bbalip.2020.158713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/06/2020] [Accepted: 04/13/2020] [Indexed: 11/17/2022]
Abstract
Lipoproteins have a vital role in the development of metabolic and cardiovascular diseases ranging from protective to deleterious effects on target tissues. VLDL has been shown to induce lipotoxic lipid accumulation and exert a variety of negative effects on cardiomyocytes. Lipotoxicity and endoplasmic reticulum (ER) stress are proposed to be the mediators of damaging effects of metabolic diseases on cardiovascular system. We treated cardiomyocytes with lipoproteins to evaluate the adaptability of these cells to metabolic stress induced by starvation and excess of lipoproteins, and to evaluate the effect of lipoproteins and lipid accumulation on ER stress. VLDL reversed metabolic stress induced by starvation, while HDL did not. VLDL induced dose-dependent lipid accumulation in cardiomyocytes, which however did not result in reduced cell viability or induction of ER stress. Moreover, VLDL or HDL pre-treatment reduced ER stress in cardiomyocytes induced by tunicamycin and palmitic acid as measured by the expression of ER stress markers, even in conditions of increased lipid accumulation. VLDL and HDL induced activation of pro-survival ERK1/2 in cardiomyocytes; however, this activation was not involved in the protection against ER stress. Additionally, we observed that LDLR and VLDLR are regulated differently by lipoproteins and cellular stress, as lipoproteins induced VLDLR protein independently of the level of lipid accumulation. We conclude that VLDL is not a priori detrimental for cardiomyocytes and can even have beneficial effects, enabling cell survival under starvation and attenuating ER stress.
Collapse
Affiliation(s)
- Sara Tekavec
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tjaša Sorčan
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Tadeja Režen
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
14
|
Tran V, De Silva TM, Sobey CG, Lim K, Drummond GR, Vinh A, Jelinic M. The Vascular Consequences of Metabolic Syndrome: Rodent Models, Endothelial Dysfunction, and Current Therapies. Front Pharmacol 2020; 11:148. [PMID: 32194403 PMCID: PMC7064630 DOI: 10.3389/fphar.2020.00148] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/04/2020] [Indexed: 12/30/2022] Open
Abstract
Metabolic syndrome is characterized by visceral obesity, dyslipidemia, hyperglycemia and hypertension, and affects over one billion people. Independently, the components of metabolic syndrome each have the potential to affect the endothelium to cause vascular dysfunction and disrupt vascular homeostasis. Rodent models of metabolic syndrome have significantly advanced our understanding of this multifactorial condition. In this mini-review we compare the currently available rodent models of metabolic syndrome and consider their limitations. We also discuss the numerous mechanisms by which metabolic abnormalities cause endothelial dysfunction and highlight some common pathophysiologies including reduced nitric oxide production, increased reactive oxygen species and increased production of vasoconstrictors. Additionally, we explore some of the current therapeutics for the comorbidities of metabolic syndrome and consider how these benefit the vasculature.
Collapse
Affiliation(s)
- Vivian Tran
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Kyungjoon Lim
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Antony Vinh
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Maria Jelinic
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
15
|
Chang HR, Josefs T, Scerbo D, Gumaste N, Hu Y, Huggins LA, Barett T, Chiang S, Grossman J, Bagdasarov S, Fisher EA, Goldberg IJ. Role of LpL (Lipoprotein Lipase) in Macrophage Polarization In Vitro and In Vivo. Arterioscler Thromb Vasc Biol 2019; 39:1967-1985. [PMID: 31434492 PMCID: PMC6761022 DOI: 10.1161/atvbaha.119.312389] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Fatty acid uptake and oxidation characterize the metabolism of alternatively activated macrophage polarization in vitro, but the in vivo biology is less clear. We assessed the roles of LpL (lipoprotein lipase)-mediated lipid uptake in macrophage polarization in vitro and in several important tissues in vivo. Approach and Results: We created mice with both global and myeloid-cell specific LpL deficiency. LpL deficiency in the presence of VLDL (very low-density lipoproteins) altered gene expression of bone marrow-derived macrophages and led to reduced lipid uptake but an increase in some anti- and some proinflammatory markers. However, LpL deficiency did not alter lipid accumulation or gene expression in circulating monocytes nor did it change the ratio of Ly6Chigh/Ly6Clow. In adipose tissue, less macrophage lipid accumulation was found with global but not myeloid-specific LpL deficiency. Neither deletion affected the expression of inflammatory genes. Global LpL deficiency also reduced the numbers of elicited peritoneal macrophages. Finally, we assessed gene expression in macrophages from atherosclerotic lesions during regression; LpL deficiency did not affect the polarity of plaque macrophages. CONCLUSIONS The phenotypic changes observed in macrophages upon deletion of Lpl in vitro is not mimicked in tissue macrophages.
Collapse
Affiliation(s)
- Hye Rim Chang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Tatjana Josefs
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Diego Scerbo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Namrata Gumaste
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Yunying Hu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Lesley-Ann Huggins
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Tessa Barett
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York; Division of Vascular Surgery, Department of Surgery, New York University School of Medicine, New York, New York
| | - Stephanie Chiang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Jennifer Grossman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Svetlana Bagdasarov
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Edward A. Fisher
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| |
Collapse
|
16
|
A third of nonfasting plasma cholesterol is in remnant lipoproteins: Lipoprotein subclass profiling in 9293 individuals. Atherosclerosis 2019; 286:97-104. [DOI: 10.1016/j.atherosclerosis.2019.05.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/26/2019] [Accepted: 05/08/2019] [Indexed: 12/30/2022]
|
17
|
Chan SMH, Selemidis S, Bozinovski S, Vlahos R. Pathobiological mechanisms underlying metabolic syndrome (MetS) in chronic obstructive pulmonary disease (COPD): clinical significance and therapeutic strategies. Pharmacol Ther 2019; 198:160-188. [PMID: 30822464 PMCID: PMC7112632 DOI: 10.1016/j.pharmthera.2019.02.013] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major incurable global health burden and is currently the 4th largest cause of death in the world. Importantly, much of the disease burden and health care utilisation in COPD is associated with the management of its comorbidities (e.g. skeletal muscle wasting, ischemic heart disease, cognitive dysfunction) and infective viral and bacterial acute exacerbations (AECOPD). Current pharmacological treatments for COPD are relatively ineffective and the development of effective therapies has been severely hampered by the lack of understanding of the mechanisms and mediators underlying COPD. Since comorbidities have a tremendous impact on the prognosis and severity of COPD, the 2015 American Thoracic Society/European Respiratory Society (ATS/ERS) Research Statement on COPD urgently called for studies to elucidate the pathobiological mechanisms linking COPD to its comorbidities. It is now emerging that up to 50% of COPD patients have metabolic syndrome (MetS) as a comorbidity. It is currently not clear whether metabolic syndrome is an independent co-existing condition or a direct consequence of the progressive lung pathology in COPD patients. As MetS has important clinical implications on COPD outcomes, identification of disease mechanisms linking COPD to MetS is the key to effective therapy. In this comprehensive review, we discuss the potential mechanisms linking MetS to COPD and hence plausible therapeutic strategies to treat this debilitating comorbidity of COPD.
Collapse
Affiliation(s)
- Stanley M H Chan
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia.
| |
Collapse
|
18
|
Lipoprotein markers associated with disability from multiple sclerosis. Sci Rep 2018; 8:17026. [PMID: 30451923 PMCID: PMC6242870 DOI: 10.1038/s41598-018-35232-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/18/2018] [Indexed: 12/25/2022] Open
Abstract
Altered lipid metabolism is a feature of chronic inflammatory disorders. Increased plasma lipids and lipoproteins have been associated with multiple sclerosis (MS) disease activity. Our objective was to characterise the specific lipids and associated plasma lipoproteins increased in MS and to test for an association with disability. Plasma samples were collected from 27 RRMS patients (median EDSS, 1.5, range 1–7) and 31 healthy controls. Concentrations of lipids within lipoprotein sub-classes were determined from NMR spectra. Plasma cytokines were measured using the MesoScale Discovery V-PLEX kit. Associations were tested using multivariate linear regression. Differences between the patient and volunteer groups were found for lipids within VLDL and HDL lipoprotein sub-fractions (p < 0.05). Multivariate regression demonstrated a high correlation between lipids within VLDL sub-classes and the Expanded Disability Status Scale (EDSS) (p < 0.05). An optimal model for EDSS included free cholesterol carried by VLDL-2, gender and age (R2 = 0.38, p < 0.05). Free cholesterol carried by VLDL-2 was highly correlated with plasma cytokines CCL-17 and IL-7 (R2 = 0.78, p < 0.0001). These results highlight relationships between disability, inflammatory responses and systemic lipid metabolism in RRMS. Altered lipid metabolism with systemic inflammation may contribute to immune activation.
Collapse
|
19
|
|
20
|
Priyadarshini S, Pradhan B, Aich P. Role of murine macrophage in temporal regulation of cortisol- and serotonin-induced adipogenesis in pre-adipocytes when grown together. Biol Open 2018; 7:bio.034629. [PMID: 30082376 PMCID: PMC6124570 DOI: 10.1242/bio.034629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Regulation of adipogenesis, the root cause for obesity, is very poorly understood. However, studies have presented evidence of immuno-metabolic regulation of adipose tissue during periods of chronic psychological stress, leading to adverse conditions related to stress manifestation, including visceral obesity and atherosclerosis. Despite pronounced association of hormonal markers of stress with dys-regulated metabolic states, the contributing signalling events are yet to be established. It is apparent that to understand contributing signalling events we need a model. Although an in vivo model is preferred, it is difficult to establish. The current report, therefore, presents an in vitro model system for the simulation of adipose tissue in a chronic stress micro-environment by growing pre-adipocytes with macrophages in the presence and absence of stress hormones. In this report, effects of cortisol and serotonin on the kinetics of immune and metabolic changes in adipocytes and macrophage (alone and co-cultured) was studied through whole genome transcriptome profiling. A transition from pro- to anti-inflammatory response in the immune profile of pre-adipocytes, with increasing time in co-culture with macrophages, was observed. This transition was reversed by stress hormones cortisol and/or serotonin. Summary: Stress-induced obesity is poorly understood in vivo at the molecular level. The current report established a novel molecular basis of adipogenesis in vitro.
Collapse
Affiliation(s)
- Sushri Priyadarshini
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO- Bhimpur-Padanpur, Via- Jatni, District:- Khurda, 752050, Odisha, India
| | - Biswaranjan Pradhan
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO- Bhimpur-Padanpur, Via- Jatni, District:- Khurda, 752050, Odisha, India
| | - Palok Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO- Bhimpur-Padanpur, Via- Jatni, District:- Khurda, 752050, Odisha, India
| |
Collapse
|
21
|
Watanabe N, S Sawada S, Shimada K, Lee IM, Gando Y, Momma H, Kawakami R, Miyachi M, Hagi Y, Kinugawa C, Okamoto T, Tsukamoto K, N Blair S. Relationship between Cardiorespiratory Fitness and Non-High-Density Lipoprotein Cholesterol: A Cohort Study. J Atheroscler Thromb 2018; 25:1196-1205. [PMID: 30089756 PMCID: PMC6249362 DOI: 10.5551/jat.43851] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Aim: Recent studies have suggested that non-high-density lipoprotein cholesterol (non-HDL-C) may be a good marker of coronary heart disease and cardiovascular disease risk. Therefore, we investigated the relationship between cardiorespiratory fitness (CRF) and non-HDL-C. Methods: We evaluated CRF and the incidence of high level of non-HDL-C in 4,067 Japanese men without dyslipidemia. The participants were given a submaximal exercise test, a medical examination, and questionnaires on their health habits in 1986. A cycle ergometer was used to measure the CRF and maximal oxygen uptake was estimated. The incidence of a high level of non-HDL -C (≥ 170 mg/dL) from 1986 to 2006 was ascertained based on the fasting blood levels. A high level of non-HDL-C was found in 1,482 participants during the follow-up. Cox proportional hazard models were used to obtain the hazard ratios (HRs) and 95% confidence intervals (CIs) for the incidence of a high level of non-HDL-C. Results: Following age adjustment, and using the lowest CRF group (quartile I) as reference, the HRs and 95% CIs for quartiles II through IV were: 1.00 (95% CI: 0.87–1.15), 0.87 (95% CI: 0.76–1.00), and 0.70 (95% CI: 0.60–0.81), respectively (P for trend < 0.001). After additional adjustment for body mass index, systolic blood pressure, smoking, alcohol intake, and family history of dyslipidemia, the HRs and 95% CIs were: 1.05 (95% CI: 0.92–1.21), 0.94 (95% CI: 0.81–1.08), and 0.79 (95% CI: 0.67–0.92), respectively (P for trend = 0.001). Conclusions: These results suggest that there is an inverse relationship between CRF levels and the incidence of a high level of non-HDL-C in Japanese men.
Collapse
Affiliation(s)
- Natsumi Watanabe
- Graduate School of Health and Sports Science, Juntendo University
| | | | - Kazunori Shimada
- Graduate School of Health and Sports Science, Juntendo University.,Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine
| | - I-Min Lee
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School.,Department of Epidemiology, Harvard T.H. Chan School of Public Health
| | - Yuko Gando
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition
| | - Haruki Momma
- Division of Biomedical Engineering for Health and Welfare, Tohoku University Graduate School of Biomedical Engineering
| | | | - Motohiko Miyachi
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition
| | - Yumiko Hagi
- Department of Sports & Leisure Management, Tokai University
| | | | | | | | - Steven N Blair
- Arnold School of Public Health, University of South Carolina
| |
Collapse
|
22
|
Abstract
Adipose tissue is a special tissue environment due to its high lipid content. Adipose tissue macrophages (ATMs) help maintain adipose tissue homeostasis in steady state by clearing dead adipocytes. However, adipose tissue changes drastically during obesity, resulting in a state of chronic low grade inflammation and a shift in the adipose immune landscape. In this review we will discuss how these changes influence the polarization of ATMs.
Collapse
Affiliation(s)
- Leen Catrysse
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.
| |
Collapse
|
23
|
Varbo A, Freiberg JJ, Nordestgaard BG. Remnant Cholesterol and Myocardial Infarction in Normal Weight, Overweight, and Obese Individuals from the Copenhagen General Population Study. Clin Chem 2018; 64:219-230. [DOI: 10.1373/clinchem.2017.279463] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/21/2017] [Indexed: 11/06/2022]
Abstract
Abstract
BACKGROUND
We tested whether high remnant cholesterol is associated with high myocardial infarction risk, independent of whether an individual is normal weight, overweight, or obese.
METHODS
A total of 106216 individuals from the Copenhagen General Population Study were followed for up to 11 years, during which 1565 experienced a myocardial infarction. Individuals were grouped by clinically meaningful remnant cholesterol concentrations of <0.5 mmol/L (19 mg/dL), 0.5 to 0.99 mmol/L (19–38 mg/dL), 1.0 to 1.49 mmol/L (39–58 mg/dL), and ≥1.5 mmol/L (58 mg/dL), and by body mass index (BMI) of <18.5 kg/m2 (underweight), 18.5 to 24.9 kg/m2 (normal weight), 25 to 29.9 kg/m2 (overweight), and ≥30 kg/m2 (obese).
RESULTS
Median calculated remnant cholesterol was 0.40 mmol/L [interquartile range (IQR), 0.30–0.55 mmol/L] [15 mg/dL (12–21 mg/dL)] for underweight, 0.50 mmol/L (IQR, 0.37–0.71 mmol/L) [19 mg/dL (14–27 mg/dL)] for normal weight, 0.70 mmol/L (IQR, 0.49–1.00 mmol/L) [27 mg/dL (19–39 mg/dL)] for overweight, and 0.85 mmol/L (IQR, 0.61–1.20 mmol/L) [(33 mg/dL (24–46 mg/dL)] for obese individuals. On continuous scales, remnant cholesterol was positively correlated with BMI until reaching a plateau of approximately 1 mmol/L (39 mg/dL) at BMI >35 kg/m2. R2 from an unadjusted linear regression for the correlation between calculated remnant cholesterol and BMI was 12%. Stepwise higher remnant cholesterol was associated with stepwise higher myocardial infarction risk in a similar pattern for normal weight, overweight, and obese individuals. When compared with individuals with remnant cholesterol <0.5 mmol/L (19 mg/dL), individuals with remnant cholesterol ≥1.5 mmol/L (58 mg/dL) had hazard ratios for myocardial infarction of 2.0 (95% CI, 1.3–3.2) for normal weight, 1.9 (95% CI, 1.4–2.6) for overweight, and 2.3 (95% CI, 1.4–3.5) for obese individuals. Directly measured remnant cholesterol increased 0.91 mmol/L (95% CI, 0.89–0.94 mmol/L) [35 mg/dL (34–36 mg/dL)] per 1 mmol/L (39 mg/dL) increase in calculated remnant cholesterol.
CONCLUSIONS
Remnant cholesterol and BMI were positively correlated; however, high remnant cholesterol was associated with higher myocardial infarction risk across the examined BMI subcategories, indicating that remnant cholesterol is a risk factor for myocardial infarction independent of overweight and obesity.
Collapse
Affiliation(s)
- Anette Varbo
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Jacob J Freiberg
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
24
|
Kumari M, Heeren J, Scheja L. Regulation of immunometabolism in adipose tissue. Semin Immunopathol 2017; 40:189-202. [PMID: 29209828 DOI: 10.1007/s00281-017-0668-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 11/22/2017] [Indexed: 12/14/2022]
|
25
|
Macrophage VLDLR mediates obesity-induced insulin resistance with adipose tissue inflammation. Nat Commun 2017; 8:1087. [PMID: 29057873 PMCID: PMC5651811 DOI: 10.1038/s41467-017-01232-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 08/25/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity is closely associated with increased adipose tissue macrophages (ATMs), which contribute to systemic insulin resistance and altered lipid metabolism by creating a pro-inflammatory environment. Very low-density lipoprotein receptor (VLDLR) is involved in lipoprotein uptake and storage. However, whether lipid uptake via VLDLR in macrophages affects obesity-induced inflammatory responses and insulin resistance is not well understood. Here we show that elevated VLDLR expression in ATMs promotes adipose tissue inflammation and glucose intolerance in obese mice. In macrophages, VLDL treatment upregulates intracellular levels of C16:0 ceramides in a VLDLR-dependent manner, which potentiates pro-inflammatory responses and promotes M1-like macrophage polarization. Adoptive transfer of VLDLR knockout bone marrow to wild-type mice relieves adipose tissue inflammation and improves insulin resistance in diet-induced obese mice. These findings suggest that increased VLDL-VLDLR signaling in ATMs aggravates adipose tissue inflammation and insulin resistance in obesity. VLDLR regulates cellular lipoprotein uptake and storage. Here, the authors show that VLDLR, expressed on adipose tissue macrophages, is upregulated in obesity and promotes adipose tissue inflammation by upregulating ceramide production and facilitating M1-like macrophage polarization.
Collapse
|
26
|
McCurdy S, Baumer Y, Toulmin E, Lee BH, Boisvert WA. Macrophage-Specific Expression of IL-37 in Hyperlipidemic Mice Attenuates Atherosclerosis. THE JOURNAL OF IMMUNOLOGY 2017; 199:3604-3613. [DOI: 10.4049/jimmunol.1601907] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 09/12/2017] [Indexed: 01/05/2023]
|
27
|
Abstract
Cholesterol-rich, apolipoprotein B (apoB)-containing lipoproteins are now widely accepted as the most important causal agents of atherosclerotic cardiovascular disease. Multiple unequivocal and orthogonal lines of evidence all converge on low-density lipoprotein and related particles as being the principal actors in the genesis of atherosclerosis. Here, we review the fundamental role of atherogenic apoB-containing lipoproteins in cardiovascular disease and several other humoral and parietal factors that are required to initiate and maintain arterial degeneration. The biology of foam cells and their interactions with high-density lipoproteins, including cholesterol efflux, are also briefly reviewed.
Collapse
Affiliation(s)
- Michael D Shapiro
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Sergio Fazio
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| |
Collapse
|
28
|
Zhao D, Tong L, Zhang L, Li H, Wan Y, Zhang T. Tanshinone II A stabilizes vulnerable plaques by suppressing RAGE signaling and NF-κB activation in apolipoprotein-E-deficient mice. Mol Med Rep 2016; 14:4983-4990. [PMID: 27840935 PMCID: PMC5355755 DOI: 10.3892/mmr.2016.5916] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 09/05/2016] [Indexed: 11/11/2022] Open
Abstract
Tanshinone II A (TSIIA) is a diterpene quinone extracted from the roots of Salvia miltiorrhiza with anti-inflammatory and anti‑oxidant properties that is used to treat atherosclerosis. In the current study, morphological analyses were conducted to evaluate the effects of TSIIA on atherosclerotic vulnerable plaque stability. Additionally, receptor of advanced glycation end products (RAGE), adhesion molecule, and matrix‑metalloproteinases (MMPs) expression, and nuclear factor-κB (NF‑κB) activation were examined in apolipoprotein E (apoE)‑deficient mice treated with TSIIA. Eight‑week‑old apoE-/- mice were administered TSIIA and fed an atherogenic diet for 8 weeks. TSIIA exhibited no effects on plaque size. Analysis of the vulnerable plaque composition demonstrated decreased numbers of macrophages and smooth muscle cells, and increased collagen content in apoE‑deficient mice treated with TSIIA compared with untreated mice. Western blotting revealed that TSIIA downregulated the expression levels of vascular cellular adhesion molecule-1 (VCAM-1), intercellular adhesion molecule‑1 (ICAM‑1), and MMP‑2, ‑3, and ‑9, suppressed RAGE, and inhibited NF‑κB, JNK and p38 activation. The present study demonstrated that the underlying mechanism of TSIIA stabilization of vulnerable plaques involves interfering with RAGE and NF‑κB activation, and downregulation of downstream inflammatory factors, including ICAM‑1, VCAM‑1, and MMP‑2, ‑3 and ‑9 in apoE-/- mice.
Collapse
Affiliation(s)
- Dong Zhao
- Department of Geriatric Medicine, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Lufang Tong
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Lixin Zhang
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Hong Li
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Yingxin Wan
- Third Department of Internal Medicine, Changping Chinese and Western Medicine Hospital, Beijing 102208, P.R. China
| | - Tiezhong Zhang
- Department of Geriatric Medicine, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| |
Collapse
|
29
|
Handelsman Y, Shapiro MD. TRIGLYCERIDES, ATHEROSCLEROSIS, AND CARDIOVASCULAR OUTCOME STUDIES: FOCUS ON OMEGA-3 FATTY ACIDS. Endocr Pract 2016; 23:100-112. [PMID: 27819772 DOI: 10.4158/ep161445.ra] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To provide an overview of the roles of triglycerides and triglyceride-lowering agents in atherosclerosis in the context of cardiovascular outcomes studies. METHODS We reviewed the published literature as well as ClinicalTrials.gov entries for ongoing studies. RESULTS Despite improved atherosclerotic cardiovascular disease (ASCVD) outcomes with statin therapy, residual risk remains. Epidemiologic data and recent genetic insights provide compelling evidence that triglycerides are in the causal pathway for the development of atherosclerosis, thereby renewing interest in targeting triglycerides to improve ASCVD outcomes. Fibrates, niacin, and omega-3 fatty acids (OM3FAs) are three classes of triglyceride-lowering drugs. Outcome studies with triglyceride-lowering agents have been inconsistent. With regard to OM3FAs, the JELIS study showed that eicosapentaenoic acid (EPA) significantly reduced major coronary events in statin-treated hypercholesterolemic patients. Regarding other agents, extended-release niacin and fenofibrate are no longer recommended as statin add-on therapy (by some guidelines, though not all) because of the lack of convincing evidence from outcome studies. Notably, subgroup analyses from the outcome studies have generated the hypothesis that triglyceride lowering may provide benefit in statin-treated patients with persistent hypertriglyceridemia. Two ongoing OM3FA outcome studies (REDUCE-IT and STRENGTH) are testing this hypothesis in high-risk, statin-treated patients with triglyceride levels of 200 to 500 mg/dL. CONCLUSION There is consistent evidence that triglycerides are in the causal pathway of atherosclerosis but inconsistent evidence from cardiovascular outcomes studies as to whether triglyceride-lowering agents reduce cardiovascular risk. Ongoing outcomes studies will determine the role of triglyceride lowering in statin-treated patients with high-dose prescription OM3FAs in terms of improved ASCVD outcomes. ABBREVIATIONS AACE = American Association of Clinical Endocrinologists ACCORD = Action to Control Cardiovascular Risk in Diabetes AIM-HIGH = Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglycerides: Impact on Global Health Outcomes apo = apolipoprotein ASCEND = A Study of Cardiovascular Events in Diabetes ASCVD = atherosclerotic cardiovascular disease BIP = Bezafibrate Infarction Prevention CHD = coronary heart disease CI = confidence interval CV = cardiovascular CVD = cardiovascular disease DHA = docosahexaenoic acid DO-IT = Diet and Omega-3 Intervention Trial EPA = eicosapentaenoic acid FIELD = Fenofibrate Intervention and Event Lowering in Diabetes GISSI-HF = GISSI-Heart Failure HDL-C = high-density-lipoprotein cholesterol HPS2-THRIVE = Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events HR = hazard ratio JELIS = Japan Eicosapentaenoic Acid Lipid Intervention Study LDL = low-density lipoprotein LDL-C = low-density-lipoprotein cholesterol MI = myocardial infarction OM3FAs = omega-3 fatty acids VITAL = Vitamin D and Omega-3 Trial.
Collapse
|
30
|
Varbo A, Langsted A, Nordestgaard BG. Commentary: Nonfasting remnant cholesterol simplifies triglyceride-rich lipoproteins for clinical use, and metabolomic phenotyping ignites scientific curiosity. Int J Epidemiol 2016; 45:1379-1385. [PMID: 27581802 DOI: 10.1093/ije/dyw215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2016] [Indexed: 12/12/2022] Open
Affiliation(s)
- Anette Varbo
- Department of Clinical Biochemistry.,Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark and
| | - Anne Langsted
- Department of Clinical Biochemistry.,Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark and
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, .,Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark and.,Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Frederiksberg, Denmark
| |
Collapse
|
31
|
Abstract
The introduction of statins ≈ 30 years ago ushered in the era of lipid lowering as the most effective way to reduce risk of atherosclerotic cardiovascular disease. Nonetheless, residual risk remains high, and statin intolerance is frequently encountered in clinical practice. After a long dry period, the field of therapeutics targeted to lipids and atherosclerosis has entered a renaissance. Moreover, the demonstration of clinical benefits from the addition of ezetimibe to statin therapy in subjects with acute coronary syndromes has renewed the enthusiasm for the cholesterol hypothesis and the hope that additional agents that lower low-density lipoprotein will decrease risk of atherosclerotic cardiovascular disease. Drugs in the orphan disease category are now available for patients with the most extreme hypercholesterolemia. Furthermore, discovery and rapid translation of a novel biological pathway has given rise to a new class of cholesterol-lowering drugs, the proprotein convertase subtilisin kexin-9 inhibitors. Trials of niacin added to statin have failed to demonstrate cardiac benefits, and 3 cholesterol ester transfer protein inhibitors have also failed to reduce atherosclerotic cardiovascular disease risk, despite producing substantial increases in HDL levels. Although the utility of triglyceride-lowering therapies remains uncertain, 2 large clinical trials are testing the influence of omega-3 polyunsaturated fatty acids on atherosclerotic events in hypertriglyceridemia. Novel antisense therapies targeting apolipoprotein C-III (for triglyceride reduction) and apo(a) (for lipoprotein(a) reduction) are showing a promising trajectory. Finally, 2 large clinical trials are formally putting the inflammatory hypothesis of atherosclerosis to the test and may open a new avenue for cardiovascular disease risk reduction.
Collapse
Affiliation(s)
- Michael D Shapiro
- From the Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR
| | - Sergio Fazio
- From the Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR.
| |
Collapse
|
32
|
Chen X, Zhao ZW, Li L, Chen XJ, Xu H, Lou JT, Li LJ, Du LZ, Xie CH. Hypercoagulation and elevation of blood triglycerides are characteristics of Kawasaki disease. Lipids Health Dis 2015; 14:166. [PMID: 26714775 PMCID: PMC4696131 DOI: 10.1186/s12944-015-0167-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/22/2015] [Indexed: 12/17/2022] Open
Abstract
Background Cardiovascular damages poses risks to children with Kawasaki disease (KD). Although hypertriglyceridemia and hypercholesteremia are risk factors of cardiovascular damages, studies on the blood lipid metabolism in KD are still limited. This study aims to analyze the blood lipids and coagulation in KD. Methods Triglyceride (TG) and cholesterol levels in the plasma and serum from 20 children with KD were examined in comparison with 10 healthy children (HC) as well as 10 children with high fever from identified bacterial infections (BT). Using electrospray ionization mass spectrometry, we profiled the lipid species. Blood coagulation was analyzed. Statistics was analyzed by one-way ANOVA using SigmaStat. Results We found that in KD, plasma TG level was significantly increased, but not serum TG. A total of 19 molecular species of TG were identified, and they were all increased in KD and BT patients, and more pronounced in KD. On the other hand, major molecular species of plasma phosphotidylcholine and lyso-phosphotidylcholine were decreased in KD and BT. Pronounced hypercoagulation was found in KD blood. Conclusion Our data indicate hyperlipidemia in KD, especially for TG, which contributes to the hypercoagulation and the potential risk of cardiovascular damages. Evaluation of blood lipid levels in severe KD patients could provide valuable information for treatment and prognosis, thus would be worthy of consideration. Electronic supplementary material The online version of this article (doi:10.1186/s12944-015-0167-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xi Chen
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, No.3333 Bin-Sheng Road, Bin-Jiang Dist, Hangzhou, Zhejiang, 310052, China. .,Key Laboratory for Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Zhen-Wen Zhao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.
| | - Lin Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.
| | - Xue-Jun Chen
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, No.3333 Bin-Sheng Road, Bin-Jiang Dist, Hangzhou, Zhejiang, 310052, China. .,Key Laboratory for Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Hui Xu
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, No.3333 Bin-Sheng Road, Bin-Jiang Dist, Hangzhou, Zhejiang, 310052, China.
| | - Jin-Tu Lou
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, No.3333 Bin-Sheng Road, Bin-Jiang Dist, Hangzhou, Zhejiang, 310052, China. .,Key Laboratory for Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Lin-Jie Li
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, No.3333 Bin-Sheng Road, Bin-Jiang Dist, Hangzhou, Zhejiang, 310052, China.
| | - Li-Zhong Du
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, No.3333 Bin-Sheng Road, Bin-Jiang Dist, Hangzhou, Zhejiang, 310052, China. .,Key Laboratory for Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Chun-Hong Xie
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, No.3333 Bin-Sheng Road, Bin-Jiang Dist, Hangzhou, Zhejiang, 310052, China.
| |
Collapse
|
33
|
Adi N, Perriotte-Olson C, Desouza C, Ramalingam R, Saraswathi V. Hematopoietic cyclooxygenase-2 deficiency increases adipose tissue inflammation and adiposity in obesity. Obesity (Silver Spring) 2015; 23:2037-45. [PMID: 26316178 PMCID: PMC6368065 DOI: 10.1002/oby.21184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 05/18/2015] [Accepted: 05/20/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Adipose tissue (AT) macrophages mediate AT inflammation in obesity, and cyclooxygenase-2 (COX-2) is a major inflammatory gene. It was hypothesized that deletion of hematopoietic COX-2 will inhibit AT inflammation in obesity. METHODS Lethally irradiated wild-type (WT) mice were injected with bone marrow (BM) cells collected from WT or COX-2 knock-out (COX-2-/-) donor mice and fed a high-fat diet for 16 weeks. RESULTS The mice that received BM cells from COX-2-/- mice (BM-COX-2-/-) gained increased body weight, fat mass, and visceral AT (VAT) mass. These mice exhibited reduced inflammatory markers in the VAT stromal vascular cells (SVC). However, the inflammatory markers were increased in adipocyte fraction and/or whole VAT. The activation of ERK1/2 MAPK, a pro-inflammatory signaling pathway, was increased in BM-COX-2-/- mice. The molecular markers of adipogenesis were increased in the VAT or adipocyte fraction. Wnt signaling markers which inhibit adipogenesis, including Wnt3A and DVL3, were reduced, and Wnt5a/b which promotes inflammation was increased in the VAT and/or adipocytes. Finally, an increase in hepatic triglyceride levels in BM-COX-2-/- mice was noted. CONCLUSIONS The data suggest that COX-2 deletion in hematopoietic cells reduces SVC inflammation but increases VAT inflammation and promotes adiposity likely via altered Wnt signaling.
Collapse
Affiliation(s)
- Nikhil Adi
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center
- VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Curtis Perriotte-Olson
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center
- VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Cyrus Desouza
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center
- VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Ramesh Ramalingam
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center
- VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Viswanathan Saraswathi
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center
- VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
- Address correspondence to: Viswanathan Saraswathi, Research Services, VA Nebraska Western Iowa Health Care System, Omaha, NE. Ph: 402-995-3033; Fax: 402-449-0604;
| |
Collapse
|
34
|
Hill AA, Reid Bolus W, Hasty AH. A decade of progress in adipose tissue macrophage biology. Immunol Rev 2015; 262:134-52. [PMID: 25319332 DOI: 10.1111/imr.12216] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
One decade has passed since seminal publications described macrophage infiltration into adipose tissue (AT) as a key contributor to inflammation and obesity-related insulin resistance. Currently, a PubMed search for 'adipose tissue inflammation' reveals over 3500 entries since these original reports. We now know that resident macrophages in lean AT are alternatively activated, M2-like, and play a role in AT homeostasis. In contrast, the macrophages in obese AT are dramatically increased in number and are predominantly classically activated, M1-like, and promote inflammation and insulin resistance. Mediators of AT macrophage (ATM) phenotype include adipokines and fatty acids secreted from adipocytes as well as cytokines secreted from other immune cells in AT. There are several mechanisms that could explain the large increase in ATMs in obesity. These include recruitment-dependent mechanisms such as adipocyte death, chemokine release, and lipolysis of fatty acids. Newer evidence also points to recruitment-independent mechanisms such as impaired apoptosis, increased proliferation, and decreased egress. Although less is known about the homeostatic function of M2-like resident ATMs, recent evidence suggests roles in AT expansion, thermoregulation, antigen presentation, and iron homeostasis. The field of immunometabolism has come a long way in the past decade, and many exciting new discoveries are bound to be made in the coming years that will expand our understanding of how AT stands at the junction of immune and metabolic co-regulation.
Collapse
Affiliation(s)
- Andrea A Hill
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | |
Collapse
|
35
|
White KT, Moorthy MV, Akinkuolie AO, Demler O, Ridker PM, Cook NR, Mora S. Identifying an Optimal Cutpoint for the Diagnosis of Hypertriglyceridemia in the Nonfasting State. Clin Chem 2015; 61:1156-63. [PMID: 26071491 DOI: 10.1373/clinchem.2015.241752] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/26/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Nonfasting triglycerides are similar or superior to fasting triglycerides at predicting cardiovascular events. However, diagnostic cutpoints are based on fasting triglycerides. We examined the optimal cutpoint for increased nonfasting triglycerides. METHODS We obtained baseline nonfasting (<8 h since last meal) samples from 6391 participants in the Women's Health Study who were followed prospectively for ≤17 years. The optimal diagnostic threshold for nonfasting triglycerides, determined by logistic regression models by use of c-statistics and the Youden index (sum of sensitivity and specificity minus 1), was used to calculate hazard ratios (HRs) for incident cardiovascular events. Performance was compared to thresholds recommended by the American Heart Association (AHA) and European guidelines. RESULTS The optimal threshold was 175 mg/dL (1.98 mmol/L), with a c-statistic of 0.656, statistically better than the AHA cutpoint of 200 mg/dL (c-statistic 0.628). For nonfasting triglycerides above and below 175 mg/dL, after adjusting for age, hypertension, smoking, hormone use, and menopausal status, the HR for cardiovascular events was 1.88 (95% CI 1.52-2.33, P < 0.001), and for triglycerides measured at 0-4 and 4-8 h since the last meal, 2.05 (1.54- 2.74) and 1.68 (1.21-2.32), respectively. We validated performance of this optimal cutpoint by use of 10-fold cross-validation and bootstrapping of multivariable models that included standard risk factors plus total and HDL cholesterol, diabetes, body mass index, and C-reactive protein. CONCLUSIONS In this study of middle-aged and older apparently healthy women, we identified a diagnostic threshold for nonfasting hypertriglyceridemia of 175 mg/dL (1.98 mmol/L), with the potential to more accurately identify cases than the currently recommended AHA cutpoint.
Collapse
Affiliation(s)
- Khendi T White
- Division of Preventive Medicine, Division of Internal Medicine, and
| | | | | | | | - Paul M Ridker
- Division of Preventive Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | - Nancy R Cook
- Division of Preventive Medicine, Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | - Samia Mora
- Division of Preventive Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA;
| |
Collapse
|
36
|
Rojas JM, Bruinstroop E, Printz RL, Alijagic-Boers A, Foppen E, Turney MK, George L, Beck-Sickinger AG, Kalsbeek A, Niswender KD. Central nervous system neuropeptide Y regulates mediators of hepatic phospholipid remodeling and very low-density lipoprotein triglyceride secretion via sympathetic innervation. Mol Metab 2015; 4:210-21. [PMID: 25737956 PMCID: PMC4338317 DOI: 10.1016/j.molmet.2015.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 12/29/2014] [Accepted: 01/09/2015] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Elevated very low-density lipoprotein (VLDL)-triglyceride (TG) secretion from the liver contributes to an atherogenic dyslipidemia that is associated with obesity, diabetes and the metabolic syndrome. Numerous models of obesity and diabetes are characterized by increased central nervous system (CNS) neuropeptide Y (NPY); in fact, a single intracerebroventricular (icv) administration of NPY in lean fasted rats elevates hepatic VLDL-TG secretion and does so, in large part, via signaling through the CNS NPY Y1 receptor. Thus, our overarching hypothesis is that elevated CNS NPY action contributes to dyslipidemia by activating central circuits that modulate liver lipid metabolism. METHODS Chow-fed Zucker fatty (ZF) rats were pair-fed by matching their caloric intake to that of lean controls and effects on body weight, plasma TG, and liver content of TG and phospholipid (PL) were compared to ad-libitum (ad-lib) fed ZF rats. Additionally, lean 4-h fasted rats with intact or disrupted hepatic sympathetic innervation were treated with icv NPY or NPY Y1 receptor agonist to identify novel hepatic mechanisms by which NPY promotes VLDL particle maturation and secretion. RESULTS Manipulation of plasma TG levels in obese ZF rats, through pair-feeding had no effect on liver TG content; however, hepatic PL content was substantially reduced and was tightly correlated with plasma TG levels. Treatment with icv NPY or a selective NPY Y1 receptor agonist in lean fasted rats robustly activated key hepatic regulatory proteins, stearoyl-CoA desaturase-1 (SCD-1), ADP-ribosylation factor-1 (ARF-1), and lipin-1, known to be involved in remodeling liver PL into TG for VLDL maturation and secretion. Lastly, we show that the effects of CNS NPY on key liporegulatory proteins are attenuated by hepatic sympathetic denervation. CONCLUSIONS These data support a model in which CNS NPY modulates mediators of hepatic PL remodeling and VLDL maturation to stimulate VLDL-TG secretion that is dependent on the Y1 receptor and sympathetic signaling to the liver.
Collapse
Key Words
- AGPAT, 1-acyl-glycerol-3-phosphate acyltransferase
- ARF-1, ADP-ribosylation factor-1
- ApoB, apolipoprotein B
- CNS, central nervous system
- Cyto, cytoplasmic
- DAG, diacylglycerol
- DGAT, diacylglycerol acyltransferase
- ER, endoplasmic reticulum
- FFA(s), free fatty acid(s)
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- HDAC-1, histone deacetylase-1
- Lipin-1
- NE, norepinephrine
- NPY Y1 receptor
- NPY, neuropeptide Y
- Nuc, nuclear
- PA, phosphatidic acid
- PAP-1, phosphatidic acid phosphatase-1
- PF, pair-fed
- PL, phospholipid
- PLD, phospholipase D
- POMC, proopiomelanocortin
- Phospholipid
- RPL13A, ribosomal protein L13a
- RT-PCR, real-time PCR
- SCD-1, stearoyl-CoA desaturase-1
- SNS, sympathetic nervous system
- Sham, sham-denervation
- Sx, sympathetic denervation
- Sympathetic denervation
- TG, triglyceride
- Triglyceride
- VLDL
- VLDL, very low-density lipoprotein
- Veh, vehicle
- ZF, Zucker fatty
- ad-lib, ad-libitum
- icv, intracerebroventricular
Collapse
Affiliation(s)
- Jennifer M. Rojas
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Eveline Bruinstroop
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Richard L. Printz
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Aldijana Alijagic-Boers
- Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Science, Amsterdam, The Netherlands
| | - Ewout Foppen
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Maxine K. Turney
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Leena George
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Annette G. Beck-Sickinger
- Institute of Biochemistry, Faculty of Bioscience, Pharmacy and Psychology, Leipzig University, Leipzig, Germany
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Science, Amsterdam, The Netherlands
| | - Kevin D. Niswender
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
37
|
Shrestha R, Hui SP, Miura Y, Yagi A, Takahashi Y, Takeda S, Fuda H, Chiba H. Identification of molecular species of oxidized triglyceride in plasma and its distribution in lipoproteins. ACTA ACUST UNITED AC 2015; 53:1859-69. [DOI: 10.1515/cclm-2014-1088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/11/2015] [Indexed: 02/01/2023]
Abstract
AbstractThe role of triglycerides carried in the triglyceride-rich lipoproteins (TRL) in the progression of atherosclerosis is uncertain. Identification of oxidized triglycerides and its possible association with atherosclerosis were largely ignored. Here we applied mass spectrometric approach to detect and identify triglyceride hydroperoxides (TGOOH) in human plasma and lipoproteins.EDTA plasma was collected from healthy human volunteers (n=9) after 14–16 h of fasting. Very low-density lipoprotein (VLDL)We identified 11 molecular species of TGOOH in either plasma or VLDL and IDL, of which TGOOH-18:1/18:2/16:0, TGOOH-18:1/18:1/16:0, TGOOH-16:0/18:2/16:0, TGOOH-18:1/18:1/18:1, and TGOOH-16:0/20:4/16:0 were most dominant. These TGOOH molecules are carried by TRL but not by LDL and HDL. Mean concentration of TGOOH in plasma, VLDL and IDL were, respectively, 56.1±25.6, 349.8±253.6 and 512.5±173.2 μmol/mol of triglycerides.This is the first report to identify several molecular species of oxidized triglycerides in TRL. Presence of oxidized triglyceride may contribute to the atherogenicity of TRL. Further work is needed to elucidate the association of the oxidized triglyceride in atherosclerosis.
Collapse
|
38
|
Escobar DA, Botero-Quintero AM, Kautza BC, Luciano J, Loughran P, Darwiche S, Rosengart MR, Zuckerbraun BS, Gomez H. Adenosine monophosphate-activated protein kinase activation protects against sepsis-induced organ injury and inflammation. J Surg Res 2014; 194:262-72. [PMID: 25456115 DOI: 10.1016/j.jss.2014.10.009] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 09/18/2014] [Accepted: 10/03/2014] [Indexed: 01/20/2023]
Abstract
BACKGROUND Mortality in sepsis is most often attributed to the development of multiple organ failure. In sepsis, inflammation-mediated endothelial activation, defined as a proinflammatory and procoagulant state of the endothelial cells, has been associated with severity of disease. Thus, the objective of this study was to test the hypothesis that adenosine monophosphate-activated protein kinase (AMPK) activation limits inflammation and endothelium activation to protect against organ injury in sepsis. 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), which is an adenosine monophosphate analog, has been used to upregulate activity of AMPK. Compound C is a cell-permeable pyrrazolopyrimidine compound that inhibits AMPK activity. METHODS Wild-type mice underwent cecal ligation and puncture (CLP) or sham surgery. Mice were randomized to vehicle, AICAR, or compound C. Mouse kidney endothelial cells were used for in vitro experiments. Renal and liver function were determined by serum cystatin C, blood urea nitrogen (BUN), creatinine, and alanine aminotransferase. Serum cytokines were measured by enzyme-linked immunosorbent assay. Microvascular injury was determined using Evans blue dye and electron microscopy. Immunohistochemistry was used to measure protein levels of phospho-AMPK (p-AMPK), microtubule-associated protein 1A/1B-light chain 3 (LC3), and intracellular adhesion molecule. LC3 levels were used as a measure of autophagosome formation. RESULTS AICAR decreased liver and kidney injury induced by CLP and minimized cytokine elevation in vivo and in vitro. CLP increased renal and hepatic phosphorylation of AMPK and autophagic signaling as determined by LC3. Inhibition of AMPK with compound C prevented CLP-induced autophagy and exacerbated tissue injury. Additionally, CLP led to endothelial injury as determined by electron microscopy and Evans blue dye extravasation, and AICAR limited this injury. Furthermore, AICAR limited CLP and lipopolysaccharide (LPS)-induced upregulation of intracellular adhesion molecule in vivo and in vitro and decreased LPS-induced neutrophil adhesion in vitro. CONCLUSIONS In this model, activation of AMPK was protective, and AICAR minimized organ injury by decreasing inflammatory cytokines and endothelial activation. These data suggest that AMPK signaling influences sepsis or LPS-induced endothelial activation and organ injury.
Collapse
Affiliation(s)
- Daniel A Escobar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Benjamin C Kautza
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jason Luciano
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sophie Darwiche
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew R Rosengart
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brian S Zuckerbraun
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Surgery, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hernando Gomez
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
39
|
Esquivel AL, Pérez-Ramos J, Cisneros J, Herrera I, Rivera-Rosales R, Montaño M, Ramos C. The effect of obesity and tobacco smoke exposure on inflammatory mediators and matrix metalloproteinases in rat model. Toxicol Mech Methods 2014; 24:633-43. [PMID: 25141943 DOI: 10.3109/15376516.2014.956911] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Obesity is characterized by hypertrophy of adipose tissue and chronic obstructive pulmonary disease (COPD) by lung damage; both diseases are associated with systemic low-grade inflammation. There are no animal models combining obesity and COPD; therefore, these diseases were induced simultaneously in rats to analyze their effects on the expression of inflammatory mediators and enzymes involved in lung tissue remodeling. Obesity was induced with sucrose (30%) for 4 months concomitant with tobacco smoke exposure (20 cigarettes/day, 5 days/wk) for the last 2 months. Were evaluated: body weight, abdominal fat, dyslipidemia, glucose tolerance test (GTT), histology, inflammatory mediators with qPCR and enzyme-linked immunosorbent assay, matrix metalloproteinases (MMP-2), MMP-9, MMP-12, TIMP-1 and TIMP-2 through qRT-PCR, and MMP-2 and MMP-9 by gelatin zymography. The rats on a sucrose diet exhibited increased body weight, abdominal fat, triglycerides, GTT, and plasma levels of insulin, adiponectin, leptin, resistin, IL-6, IL-1β, tumor necrosis factor-α (TNF-α) and IFN-γ, upregulated lung IL-6, IL-1β, TNF-α and IFN-γ, showing hyperplastic bronchial and alveolar epithelium. The animals exposed to sucrose and tobacco smoke exhibited decreased body weight, abdominal fat and plasma levels of leptin, resistin, IL-1β and IFN-γ, reducing inflammation but showing emphysematous lesions. Expression of gelatinases and MMP-12 augmented in the rats exposed to tobacco smoke alone or combined with sucrose. Zymography showed prominent gelatinases activity in all the experimental groups. These results suggest that simultaneous exposure to sucrose and tobacco smoke decreases inflammation but results in emphysematous lesions similar to those observed with tobacco smoke exposure, suggesting that obesity does not confer any protective effect against lung damage.
Collapse
Affiliation(s)
- Ana Laura Esquivel
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Xochimilco-Iztapalapa-Cuajimalpa , Mexico, DF , Mexico
| | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
After the introduction of statins, clinical emphasis first focussed on LDL cholesterol-lowering, then on the potential for raising HDL cholesterol, with less focus on lowering triglycerides. However, the understanding from genetic studies and negative results from randomised trials that low HDL cholesterol might not cause cardiovascular disease as originally thought has now generated renewed interest in raised concentrations of triglycerides. This renewed interest has also been driven by epidemiological and genetic evidence supporting raised triglycerides, remnant cholesterol, or triglyceride-rich lipoproteins as an additional cause of cardiovascular disease and all-cause mortality. Triglycerides can be measured in the non-fasting or fasting states, with concentrations of 2-10 mmol/L conferring increased risk of cardiovascular disease, and concentrations greater than 10 mmol/L conferring increased risk of acute pancreatitis and possibly cardiovascular disease. Although randomised trials showing cardiovascular benefit of triglyceride reduction are scarce, new triglyceride-lowering drugs are being developed, and large-scale trials have been initiated that will hopefully provide conclusive evidence as to whether lowering triglycerides reduces the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Børge G Nordestgaard
- Department of Clinical Biochemistry and The Copenhagen General Population Study, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Anette Varbo
- Department of Clinical Biochemistry and The Copenhagen General Population Study, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
41
|
Murali G, Desouza CV, Clevenger ME, Ramalingam R, Saraswathi V. Differential effects of eicosapentaenoic acid and docosahexaenoic acid in promoting the differentiation of 3T3-L1 preadipocytes. Prostaglandins Leukot Essent Fatty Acids 2014; 90:13-21. [PMID: 24332315 DOI: 10.1016/j.plefa.2013.10.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/01/2013] [Accepted: 10/30/2013] [Indexed: 11/24/2022]
Abstract
The objective of this study was to determine the effects of enrichment with n-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), on the differentiation of 3T3-L1 preadipocytes. Enrichment with DHA but not EPA significantly increased the differentiation markers compared to control differentiated cells. DHA compared to EPA treatment led to a greater increase in adiponectin secretion and, conditioned media collected from DHA treated cells inhibited monocyte migration. Moreover, DHA treatment resulted in inhibition of pro-inflammatory signaling pathways. DHA treated cells predominantly accumulated DHA in phospholipids whereas EPA treatment led to accumulation of both EPA and its elongation product docosapentaenoic acid (DPA), an n-3 fatty acid. Of note, adding DPA to DHA inhibited DHA-induced differentiation. The differential effects of EPA and DHA on preadipocyte differentiation may be due, in part, to differences in their intracellular modification which could impact the type of n-3 fatty acids incorporated into the cells.
Collapse
Affiliation(s)
- Ganesan Murali
- Departments of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, Omaha, NE, United States; Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States; Research Services, VA Nebraska Western Iowa Health Care System, Omaha, NE, United States
| | - Cyrus V Desouza
- Research Services, VA Nebraska Western Iowa Health Care System, Omaha, NE, United States; Departments of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, Omaha, NE, United States
| | - Michelle E Clevenger
- Departments of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, Omaha, NE, United States; Research Services, VA Nebraska Western Iowa Health Care System, Omaha, NE, United States
| | - Ramesh Ramalingam
- Departments of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, Omaha, NE, United States; Research Services, VA Nebraska Western Iowa Health Care System, Omaha, NE, United States
| | - Viswanathan Saraswathi
- Departments of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, Omaha, NE, United States; Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States; Research Services, VA Nebraska Western Iowa Health Care System, Omaha, NE, United States.
| |
Collapse
|
42
|
Nguyen A, Tao H, Metrione M, Hajri T. Very low density lipoprotein receptor (VLDLR) expression is a determinant factor in adipose tissue inflammation and adipocyte-macrophage interaction. J Biol Chem 2013; 289:1688-703. [PMID: 24293365 DOI: 10.1074/jbc.m113.515320] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Obesity is associated with adipose tissue remodeling, characterized by adipocyte hypertrophy and macrophage infiltration. Previously, we have shown that very low density lipoprotein receptor (VLDLR) is virtually absent in preadipocytes but is strongly induced during adipogenesis and actively participates in adipocyte hypertrophy. In this study, we investigated the role of VLDLR in adipose tissue inflammation and adipocyte-macrophage interactions in wild type and VLDLR-deficient mice fed a high fat diet. The results show that VLDLR deficiency reduced high fat diet-induced inflammation and endoplasmic reticulum (ER) stress in adipose tissue in conjunction with reduced macrophage infiltration, especially those expressing pro-inflammatory markers. In adipocyte culture, VLDLR deficiency prevented adipocyte hypertrophy and strongly reduced VLDL-induced ER stress and inflammation. Likewise, cultures of primary peritoneal macrophages show that VLDLR deficiency reduced lipid accumulation and inflammation but did not alter chemotactic response of macrophages to adipocyte signals. Moreover, VLDLR deficiency tempered the synergistic inflammatory interactions between adipocytes and macrophages in a co-culture system. Collectively, these results show that VLDLR contributes to adipose tissue inflammation and mediates VLDL-induced lipid accumulation and induction of inflammation and ER stress in adipocytes and macrophages.
Collapse
Affiliation(s)
- Andrew Nguyen
- From the Department of Surgery, Hackensack University Medical Center, Hackensack, New Jersey 07601 and
| | | | | | | |
Collapse
|
43
|
Postprandial lipoproteins and the molecular regulation of vascular homeostasis. Prog Lipid Res 2013; 52:446-64. [DOI: 10.1016/j.plipres.2013.06.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 06/06/2013] [Accepted: 06/06/2013] [Indexed: 12/17/2022]
|
44
|
Napolitano M, Botham KM, Bravo E. Postprandial human triglyceride-rich lipoproteins increase chemoattractant protein secretion in human macrophages. Cytokine 2013; 63:18-26. [DOI: 10.1016/j.cyto.2013.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2012] [Revised: 04/19/2013] [Accepted: 04/21/2013] [Indexed: 01/17/2023]
|
45
|
Georgiadi A, Wang Y, Stienstra R, Tjeerdema N, Janssen A, Stalenhoef A, van der Vliet JA, de Roos A, Tamsma JT, Smit JW, Tan NS, Müller M, Rensen PC, Kersten S. Overexpression of Angiopoietin-Like Protein 4 Protects Against Atherosclerosis Development. Arterioscler Thromb Vasc Biol 2013; 33:1529-37. [DOI: 10.1161/atvbaha.113.301698] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Anastasia Georgiadi
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Yanan Wang
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Rinke Stienstra
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Nathanja Tjeerdema
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Aafke Janssen
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Anton Stalenhoef
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - J. Adam van der Vliet
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Albert de Roos
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Jouke T. Tamsma
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Johannes W.A. Smit
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Nguan Soon Tan
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Michael Müller
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Patrick C.N. Rensen
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| | - Sander Kersten
- From the Nutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands (A.G., R.S., A.J., M.M., S.K.); Department of Endocrinology and Metabolic Diseases and Einthoven Laboratory for Experimental Vascular Medicine (Y.W., N.T., J.T.T., J.W.A.S., P.C.N.R.), and Department of Radiology (A.d.R.), Leiden University Medical Center, Leiden, The Netherlands; Department of Medicine (R.S., A.S., J.W.A.S.), and Department of Surgery (J.A.v.d.V.), Radboud University Nijmegen
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW Therapeutic strategies to alleviate the growing epidemic of insulin-resistant syndromes (obesity and type 2 diabetes) as well as the conferred cardiovascular disease risk remain sparse. The peroxisome proliferator-activated receptor δ (PPARδ) has emerged as a versatile regulator of lipid homeostasis and inflammatory signaling, making it an attractive therapeutic target for the treatment and prevention of type 2 diabetes and atherosclerosis. RECENT FINDINGS PPARδ activation regulates lipid homeostasis and inflammatory signaling in a variety of cell types, conferring protection from metabolic disease and atherosclerosis. Specifically, PPARδ activation in the liver stimulates glucose utilization and inhibits gluconeogenesis, which improves insulin resistance and hyperglycemia. In macrophages, PPARδ-specific activation with synthetic agonists inhibits VLDL-induced triglyceride accumulation and inflammation. In mice, PPARδ agonists halt the progression of atherosclerosis and stabilize existing lesions by promoting an anti-inflammatory milieu within the diseased macrovasculature. In humans, PPARδ activation improves insulin sensitivity and reduces atherogenic dyslipidemia via a mechanism complementary to statin monotherapy. SUMMARY Recent advances in the understanding of PPARδ reveal that activation of this receptor represents a multifaceted therapeutic strategy for the prevention and treatment of insulin-resistant syndromes and atherosclerosis.
Collapse
Affiliation(s)
- Lazar A Bojic
- Department of Biochemistry, Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
| | | |
Collapse
|
47
|
Johnson AR, Milner JJ, Makowski L. The inflammation highway: metabolism accelerates inflammatory traffic in obesity. Immunol Rev 2013; 249:218-38. [PMID: 22889225 DOI: 10.1111/j.1600-065x.2012.01151.x] [Citation(s) in RCA: 434] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As humans evolved, perhaps the two strongest selection determinants of survival were a robust immune response able to clear bacterial, viral, and parasitic infection and an ability to efficiently store nutrients to survive times when food sources were scarce. These traits are not mutually exclusive. It is now apparent that critical proteins necessary for regulating energy metabolism, such as peroxisome proliferator-activated receptors, Toll-like receptors, and fatty acid-binding proteins, also act as links between nutrient metabolism and inflammatory pathway activation in immune cells. Obesity in humans is a symptom of energy imbalance: the scale has been tipped such that energy intake exceeds energy output and may be a result, in part, of evolutionary selection toward a phenotype characterized by efficient energy storage. As discussed in this review, obesity is a state of low-grade, chronic inflammation that promotes the development of insulin resistance and diabetes. Ironically, the formation of systemic and/or local, tissue-specific insulin resistance upon inflammatory cell activation may actually be a protective mechanism that co-evolved to repartition energy sources within the body during times of stress during infection. However, the point has been reached where a once beneficial adaptive trait has become detrimental to the health of the individual and an immense public health and economic burden. This article reviews the complex relationship between obesity, insulin resistance/diabetes, and inflammation, and although the liver, brain, pancreas, muscle, and other tissues are relevant, we focus specifically on how the obese adipose microenvironment can promote immune cell influx and sustain damaging inflammation that can lead to the onset of insulin resistance and diabetes. Finally, we address how substrate metabolism may regulate the immune response and discuss how fuel uptake and metabolism may be a targetable approach to limit or abrogate obesity-induced inflammation.
Collapse
Affiliation(s)
- Amy R Johnson
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
48
|
Takahashi M, Yagyu H, Tazoe F, Nagashima S, Ohshiro T, Okada K, Osuga JI, Goldberg IJ, Ishibashi S. Macrophage lipoprotein lipase modulates the development of atherosclerosis but not adiposity. J Lipid Res 2013; 54:1124-34. [PMID: 23378601 DOI: 10.1194/jlr.m035568] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The role of macrophage lipoprotein lipase (LpL) in the development of atherosclerosis and adiposity was examined in macrophage LpL knockout (MLpLKO) mice. MLpLKO mice were generated using cre-loxP gene targeting. Loss of LpL in macrophages did not alter plasma LpL activity or lipoprotein levels. Incubation of apolipoprotein E (ApoE)-deficient β-VLDL with peritoneal macrophages from ApoE knockout mice lacking macrophage LpL (MLpLKO/ApoEKO) led to less cholesteryl ester formation than that found with ApoEKO macrophages. MLpLKO/ApoEKO macrophages had reduced intracellular triglyceride levels, with decreased CD36 and carnitine palmitoyltransferase-1 mRNA levels compared with ApoEKO macrophages, when incubated with VLDL. Although both MLpLKO/ApoEKO and ApoEKO mice developed comparable hypercholesterolemia in response to feeding with a Western-type diet for 12 weeks, atherosclerosis was less in MLpLKO/ApoEKO mice. Epididymal fat mass and gene expression levels associated with inflammation did not differ between the two groups. In conclusion, macrophage LpL plays an important role in the development of atherosclerosis but not adiposity.
Collapse
Affiliation(s)
- Manabu Takahashi
- Department of Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
de Jager SCA, Bot I, Kraaijeveld AO, Korporaal SJA, Bot M, van Santbrink PJ, van Berkel TJC, Kuiper J, Biessen EAL. Leukocyte-specific CCL3 deficiency inhibits atherosclerotic lesion development by affecting neutrophil accumulation. Arterioscler Thromb Vasc Biol 2013; 33:e75-83. [PMID: 23288165 DOI: 10.1161/atvbaha.112.300857] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Despite common disbelief that neutrophils are involved in atherosclerosis, evidence is accumulating for a causal role of neutrophils in atherosclerosis. CC chemokine ligand (CCL)3 is an inflammatory chemokine and its expression is significantly increased during atherosclerotic lesion formation in mice. It has recently been shown that under conditions of inflammation neutrophils can migrate along a CCL3 gradient. In this study, we aimed to elucidate the role of leukocyte-derived CCL3 in atherogenesis. METHODS AND RESULTS Irradiated low density lipoprotein receptor(-/-) mice, reconstituted with CCL3(-/-) or littermate bone marrow showed markedly reduced CCL3 response to lipopolysaccharide treatment, establishing the critical relevance of leukocytes as source of CCL3. Hematopoietic deficiency of CCL3 significantly reduced aortic sinus lesion formation by 31% after 12 weeks of western-type diet. Interestingly, whereas plaque macrophage, collagen, and vascular smooth muscle cell content were unchanged, neutrophil adhesion to and presence in plaques was significantly attenuated in CCL3(-/-) chimeras. These mice had reduced circulating neutrophil numbers, which could be ascribed to an increased neutrophil turnover and CCL3(-/-) neutrophils were shown to be less responsive toward the neutrophil chemoattractant CXC chemokine ligand 1. CONCLUSIONS Our data indicate that under conditions of acute inflammation leukocyte-derived CCL3 can induce neutrophil chemotaxis toward the atherosclerotic plaque, thereby accelerating lesion formation.
Collapse
Affiliation(s)
- Saskia C A de Jager
- Division of Biopharmaceutics, Leiden Academic Center for Drug Research, Gorlaeus Laboratories, Leiden University, Leiden, the Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Postprandial apoE isoform and conformational changes associated with VLDL lipolysis products modulate monocyte inflammation. PLoS One 2012; 7:e50513. [PMID: 23209766 PMCID: PMC3509065 DOI: 10.1371/journal.pone.0050513] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 10/22/2012] [Indexed: 11/19/2022] Open
Abstract
Objective Postprandial hyperlipemia, characterized by increased circulating very low-density lipoproteins (VLDL) and circulating lipopolysaccharide (LPS), has been proposed as a mechanism of vascular injury. Our goal was to examine the interactions between postprandial lipoproteins, LPS, and apoE3 and apoE4 on monocyte activation. Methods and Results We showed that apoE3 complexed to phospholipid vesicles attenuates LPS-induced THP-1 monocyte cytokine expression, while apoE4 increases expression. ELISA revealed that apoE3 binds to LPS with higher affinity than apoE4. Electron paramagnetic resonance (EPR) spectroscopy of site-directed spin labels placed on specific amino acids of apoE3 showed that LPS interferes with conformational changes normally associated with lipid binding. Specifically, compared to apoE4, apoE bearing the E3-like R112→Ser mutation displays increased self association when exposed to LPS, consistent with a stronger apoE3-LPS interaction. Additionally, lipolysis of fasting VLDL from normal human donors attenuated LPS-induced TNFα secretion from monocytes to a greater extent than postprandial VLDL, an effect partially reversed by blocking apoE. This effect was reproduced using fasting VLDL lipolysis products from e3/e3 donors, but not from e4/e4 subjects, suggesting that apoE3 on fasting VLDL prevents LPS-induced inflammation more readily than apoE4. Conclusion Postprandial apoE isoform and conformational changes associated with VLDL dramatically modulate vascular inflammation.
Collapse
|