1
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 PMCID: PMC11833275 DOI: 10.1016/j.preteyeres.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
2
|
Zhang Z, Zhou Y, Lv Q, Gao K, Li Z, Miao Q, Shen L. Gegen Qinlian Decoction Modulates Atherosclerosis and Lipid Metabolism Through Cellular Interplay and Signaling Pathways. Comb Chem High Throughput Screen 2024; 27:2609-2621. [PMID: 38486386 DOI: 10.2174/0113862073285562240305113728] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVE The objective of this study is to investigate Gegen Qinlian decoction (GQD) effects on lipid metabolism and explore its mechanism for preventing and treating atherosclerosis. METHODS An atherosclerotic rat model was established, and after an 8-week high-fat diet, atherosclerosis and non-alcoholic fatty liver disease were assessed. Subsequently, GQD was administered at low and high doses. Histopathological aortic wall changes, hepatic lipid deposition, and blood lipid changes were evaluated. ELISA indicated the influence of TNF-α and IL-13, and Western blotting revealed MerTK, ABCA1, and LXR-α expression. A foam macrophage model was established, and Cell activity was detected by the MTT method. ELISA indicated the influence of PPAR-γ. The expression of ABCA1, ABCA7, ABCG1, GAS6, MerTK, SCARB1, LXR- α and LXR-β mRNA were detected by qPCR. and Western blotting revealed MerTK and LXR-α expression. The impact of drug-containing serum of GQD on efferocytosis-related factors was studied. RESULTS GQD improved atherosclerosis and non-alcoholic fatty liver disease and reduced serum low-density lipoprotein levels in the high-dose group. The high- and low-dose groups showed upregulated ABCA1, MerTK, and LXR-α expression in blood vessels and the liver, respectively. GQD decreased serum TNF-α and increased IL-13 levels. PPAR-γ expression was elevated in the high-, and low-dose groups. In the high-and low-dose groups, ABCA7, GAS6, SCARB1, and LXR-α, ABCA1 and MerTK, and ABCG1 gene expression were upregulated, respectively. Both low- and high-dose serum-containing drugs promoted LXR-β gene expression, and LXR-α protein expression was improved in the high-dose group. CONCLUSION GQD improves rat atherosclerosis and hepatic lipid metabolism by regulating PPAR-γ, LXR-α, LXR-β, ABCA1, ABCA7, and ABCG1 expression and augmenting cellular intercalation through the GAS6/TAM pathway.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Digestive Endocrinology, Beijing Fengtai Hospital of Integrated Traditional and Western Medicine, Beijing, China
| | - Yu Zhou
- Department of Nephrology Endocrinology, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, China
| | - Qin Lv
- Department of Digestive Endocrinology, Beijing Fengtai Hospital of Integrated Traditional and Western Medicine, Beijing, China
| | - Kun Gao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiguo Li
- Department of Digestive Endocrinology, Beijing Fengtai Hospital of Integrated Traditional and Western Medicine, Beijing, China
| | - Qing Miao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Shen
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Puig N, Solé A, Aguilera-Simon A, Griñán R, Rotllan N, Camps-Renom P, Benitez S. Novel Therapeutic Approaches to Prevent Atherothrombotic Ischemic Stroke in Patients with Carotid Atherosclerosis. Int J Mol Sci 2023; 24:14325. [PMID: 37762627 PMCID: PMC10531661 DOI: 10.3390/ijms241814325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/18/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Atherothrombotic stroke represents approximately 20% of all ischemic strokes. It is caused by large-artery atherosclerosis, mostly in the internal carotid artery, and it is associated with a high risk of early recurrence. After an ischemic stroke, tissue plasminogen activator is used in clinical practice, although it is not possible in all patients. In severe clinical situations, such as high carotid stenosis (≥70%), revascularization by carotid endarterectomy or by stent placement is carried out to avoid recurrences. In stroke prevention, the pharmacological recommendations are based on antithrombotic, lipid-lowering, and antihypertensive therapy. Inflammation is a promising target in stroke prevention, particularly in ischemic strokes associated with atherosclerosis. However, the use of anti-inflammatory strategies has been scarcely studied. No clinical trials are clearly successful and most preclinical studies are focused on protection after a stroke. The present review describes novel therapies addressed to counteract inflammation in the prevention of the first-ever or recurrent stroke. The putative clinical use of broad-spectrum and specific anti-inflammatory drugs, such as monoclonal antibodies and microRNAs (miRNAs) as regulators of atherosclerosis, will be outlined. Further studies are necessary to ascertain which patients may benefit from anti-inflammatory agents and how.
Collapse
Affiliation(s)
- Núria Puig
- Cardiovascular Biochemistry, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain; (N.P.); (A.S.)
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Building M, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallés, 08193 Barcelona, Spain; (A.A.-S.); (R.G.)
| | - Arnau Solé
- Cardiovascular Biochemistry, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain; (N.P.); (A.S.)
| | - Ana Aguilera-Simon
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Building M, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallés, 08193 Barcelona, Spain; (A.A.-S.); (R.G.)
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu i Sant Pau, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Raquel Griñán
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Building M, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallés, 08193 Barcelona, Spain; (A.A.-S.); (R.G.)
- Pathofisiology of Lipid-Related Deseases, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain;
| | - Noemi Rotllan
- Pathofisiology of Lipid-Related Deseases, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain;
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pol Camps-Renom
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu i Sant Pau, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Sonia Benitez
- Cardiovascular Biochemistry, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain; (N.P.); (A.S.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
4
|
Abstract
COVID-19 infections decrease total cholesterol, LDL-C, HDL-C, and apolipoprotein A-I, A-II, and B levels while triglyceride levels may be increased or inappropriately normal for the poor nutritional status. The degree of reduction in total cholesterol, LDL-C, HDL-C, and apolipoprotein A-I are predictive of mortality. With recovery lipid/lipoprotein levels return towards pre-infection levels and studies have even suggested an increased risk of dyslipidemia post-COVID-19 infection. The potential mechanisms for these changes in lipid and lipoprotein levels are discussed. Decreased HDL-C and apolipoprotein A-I levels measured many years prior to COVID-19 infections are associated with an increased risk of severe COVID-19 infections while LDL-C, apolipoprotein B, Lp (a), and triglyceride levels were not consistently associated with an increased risk. Finally, data suggest that omega-3-fatty acids and PCSK9 inhibitors may reduce the severity of COVID-19 infections. Thus, COVID-19 infections alter lipid/lipoprotein levels and HDL-C levels may affect the risk of developing COVID-19 infections.
Collapse
|
5
|
Luo L, Guo Y, Chen L, Zhu J, Li C. Crosstalk between cholesterol metabolism and psoriatic inflammation. Front Immunol 2023; 14:1124786. [PMID: 37234169 PMCID: PMC10206135 DOI: 10.3389/fimmu.2023.1124786] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Psoriasis is a chronic autoinflammatory skin disease associated with multiple comorbidities, with a prevalence ranging from 2 to 3% in the general population. Decades of preclinical and clinical studies have revealed that alterations in cholesterol and lipid metabolism are strongly associated with psoriasis. Cytokines (tumor necrosis factor-α (TNF-α), interleukin (IL)-17), which are important in the pathogenesis of psoriasis, have been shown to affect cholesterol and lipid metabolism. Cholesterol metabolites and metabolic enzymes, on the other hand, influence not only the biofunction of keratinocytes (a primary type of cell in the epidermis) in psoriasis, but also the immune response and inflammation. However, the relationship between cholesterol metabolism and psoriasis has not been thoroughly reviewed. This review mainly focuses on cholesterol metabolism disturbances in psoriasis and their crosstalk with psoriatic inflammation.
Collapse
Affiliation(s)
- Lingling Luo
- Department of Dermatology, Hospital for Skin Disease, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Youming Guo
- Department of Dermatology, Hospital for Skin Disease, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Lihao Chen
- Department of Dermatology, Hospital for Skin Disease, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Jing Zhu
- Department of Dermatology, Hospital for Skin Disease, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Chengrang Li
- Department of Dermatology, Hospital for Skin Disease, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
- Department of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and Sexually Transmitted Infections, Nanjing, Jiangsu, China
| |
Collapse
|
6
|
Ronca A, Pellegrini N, Pagliai G, Dinu M, Manfredini M, Incerti M, Favari E, Sofi F. Effects of a dietary intervention with Mediterranean vs lacto-ovo vegetarian diets on HDL function: Results from the CARDIVEG study. Nutr Metab Cardiovasc Dis 2023; 33:651-658. [PMID: 36642608 DOI: 10.1016/j.numecd.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 09/18/2022] [Accepted: 11/04/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND AIM HDL-cholesterol efflux capacity (CEC) has been shown to be a better cardiovascular (CVD) risk marker than serum HDL concentration. Several foods and nutrients have been shown to improve HDL functions, however no effective dietetic nor pharmacological strategy is available to increase CEC. This study aims to evaluate the possible effect of Mediterranean diet (MD) and lacto-ovo-vegetarian diet (VD) on HDL function in a group of clinically healthy subjects at low-to-moderate CVD risk. METHODS AND RESULTS Thirty apparently healthy subjects with a low-to-moderate cardiovascular risk profile (21 F; mean age: 51.3 ± 9.7 years) were randomly assigned to a 3-month MD or VD diet and then crossed. Participants on VD showed a reduction in total HDL CEC by 8.99% (p < 0.001) as well as a reduction in ABCA1 mediated-CEC by 18.62% (p < 0.001) compared to participants on MD. Regarding CEC mediated by aqueous diffusion, no significant changes were observed after treatment with either diet. Finally, a significant positive association between CEC mediated by the ABCA1 transporter and adiponectin was found (r = 0.462; p = 0.010). CONCLUSION The results of this study suggest that HDL activity in promoting cholesterol efflux and thereby reducing the concentration of pro-atherogenic lipoproteins was more effective in participants undergoing MD than VD. Based on these findings, the MD could be considered a better therapeutic strategy for cardiovascular prevention than VD. CLINICAL TRIAL REGISTRATION URL http://www. CLINICALTRIALS gov. Unique identifier: NCT02641834.
Collapse
Affiliation(s)
- Annalisa Ronca
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Nicoletta Pellegrini
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - Giuditta Pagliai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Monica Dinu
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Matteo Manfredini
- Department of Chemistry, Life Science, And Environmental Sustainability, University of Parma, Parma, Italy
| | - Matteo Incerti
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Elda Favari
- Department of Food and Drug, University of Parma, Parma, Italy.
| | - Francesco Sofi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
7
|
Zhou X, Jin S, Pan J, Lin Q, Yang S, Lu Y, Qiu M, Ambe PC, Basharat Z, Zimmer V, Wang W, Hong W. Relationship between Cholesterol-Related Lipids and Severe Acute Pancreatitis: From Bench to Bedside. J Clin Med 2023; 12:jcm12051729. [PMID: 36902516 PMCID: PMC10003000 DOI: 10.3390/jcm12051729] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/29/2023] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
It is well known that hypercholesterolemia in the body has pro-inflammatory effects through the formation of inflammasomes and augmentation of TLR (Toll-like receptor) signaling, which gives rise to cardiovascular disease and neurodegenerative diseases. However, the interaction between cholesterol-related lipids and acute pancreatitis (AP) has not yet been summarized before. This hinders the consensus on the existence and clinical importance of cholesterol-associated AP. This review focuses on the possible interaction between AP and cholesterol-related lipids, which include total cholesterol, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C) and apolipoprotein (Apo) A1, from the bench to the bedside. With a higher serum level of total cholesterol, LDL-C is associated with the severity of AP, while the persistent inflammation of AP is allied with a decrease in serum levels of cholesterol-related lipids. Therefore, an interaction between cholesterol-related lipids and AP is postulated. Cholesterol-related lipids should be recommended as risk factors and early predictors for measuring the severity of AP. Cholesterol-lowering drugs may play a role in the treatment and prevention of AP with hypercholesterolemia.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Shengchun Jin
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jingyi Pan
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qingyi Lin
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Shaopeng Yang
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yajing Lu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Minhao Qiu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Peter C. Ambe
- Department of General Surgery, Visceral Surgery and Coloproctology, Vinzenz-Pallotti-Hospital Bensberg, Vinzenz-Pallotti-Str. 20–24, 51429 Bensberg, Germany
| | - Zarrin Basharat
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Vincent Zimmer
- Department of Medicine, Marienhausklinik St. Josef Kohlhof, 66539 Neunkirchen, Germany
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany
| | - Wei Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Wandong Hong
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Correspondence: ; Tel./Fax: +86-0577-55579122
| |
Collapse
|
8
|
Kinoo SM, Naidoo P, Singh B, Chuturgoon A, Nagiah S. Human Hepatocyte Nuclear Factors (HNF1 and LXRb) Regulate CYP7A1 in HIV-Infected Black South African Women with Gallstone Disease: A Preliminary Study. Life (Basel) 2023; 13:life13020273. [PMID: 36836631 PMCID: PMC9968087 DOI: 10.3390/life13020273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/20/2023] Open
Abstract
Female sex, high estrogen levels, aging, obesity, and dyslipidemia are some of the risk factors associated with gallstone formation. HIV-infected patients on combination antiretroviral therapy (cART) are more prone to hypercholesterolemia. Bile acid synthesis is initiated by cholesterol 7-alpha hydroxylase (CYP7A1) and regulated by hepatocyte nuclear factors (HNF1α, HNF4α, and LXRb). The aim of this study was to evaluate the expression of HNF1α, HNF4α, LXRb, and miRNAs (HNF4α specific: miR-194-5p and miR-122*_1) that regulate CYP7A1 transcription in HIV-infected Black South African women on cART and presenting with gallstones relative to HIV-negative patients with gallstone disease. Females (n = 96) presenting with gallstone disease were stratified based on HIV status. The gene expression of CYP7A1, HNF1α, HNF4α, LXRb, miR-194-5p, and miR-122*_1 was determined using RT-qPCR. Messenger RNA and miRNA levels were reported as fold change expressed as 2-ΔΔCt (RQ min; RQ max). Fold changes >2 and <0.5 were considered significant. HIV-infected females were older in age (p = 0.0267) and displayed higher low-density lipoprotein cholesterol (LDL-c) (p = 0.0419), CYP7A1 [2.078-fold (RQ min: 1.278; RQ max: 3.381)], LXRb [2.595-fold (RQ min: 2.001; RQ max: 3.000)], and HNF1α [3.428 (RQ min: 1.806; RQ max: 6.507] levels. HNF4α [0.642-fold (RQ min: 0.266; RQ max: 1.55)], miR-194-5p [0.527-fold (RQ min: 0.37; RQ max: 0.752)], and miR-122*_1 [0.595-fold (RQ min: 0.332; RQ max: 1.066)] levels were lower in HIV-infected females. In conclusion, HIV-infected women with gallstone disease displayed higher LDL-c levels and increased bile acid synthesis, which was evidenced by the elevated expression of CYP7A1, HNF1α, and LXRb. This could have been further influenced by cART and aging.
Collapse
Affiliation(s)
- Suman Mewa Kinoo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Science, University of KwaZulu Natal, Glenwood, Durban 4041, South Africa
- Discipline of General Surgery, School of Clinical Medicine, College of Health Science, University of KwaZulu Natal, Umbilo, Durban 4001, South Africa
| | - Pragalathan Naidoo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Science, University of KwaZulu Natal, Glenwood, Durban 4041, South Africa
| | - Bhugwan Singh
- Discipline of General Surgery, School of Clinical Medicine, College of Health Science, University of KwaZulu Natal, Umbilo, Durban 4001, South Africa
| | - Anil Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Science, University of KwaZulu Natal, Glenwood, Durban 4041, South Africa
- Correspondence: (A.C.); (S.N.)
| | - Savania Nagiah
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Science, University of KwaZulu Natal, Glenwood, Durban 4041, South Africa
- Department of Human Biology, Medical School, Faculty of Health Sciences, Nelson Mandela University, Missionvale, Port Elizabeth 6065, South Africa
- Correspondence: (A.C.); (S.N.)
| |
Collapse
|
9
|
Tetramethylpyrazine and Paeoniflorin Synergistically Attenuate Cholesterol Efflux in Macrophage Cells via Enhancing ABCA1 and ABCG1 Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4304790. [PMID: 36387364 PMCID: PMC9653297 DOI: 10.1155/2022/4304790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/28/2022] [Accepted: 10/29/2022] [Indexed: 11/08/2022]
Abstract
The formation of foam cells is a characteristic of the occurrence and development of atherosclerosis. ATP-binding cassette subfamily A1 and G1 (ABCA1 and ABCG1) and scavenger receptor B1 (SR-B1) play critical roles in promoting intracellular cholesterol efflux to high-density lipoprotein (HDL) or apolipoprotein A1 (apoA1). We attempted to test the effect of the tetramethylpyrazine-paeoniflorin pair (TP) on cholesterol outflow in foam cells derived from macrophages. In this study, RAW264.7 macrophages were treated with 80 mg/L oxidized low-density lipoprotein (ox-LDL) for 24 h to obtain foam cells. Then they were intervened with TP (tetramethylpyrazine 40 ug/ml plus paeoniflorin 80 ug/ml) for additional 24 h. The distribution of cholesterol in foam cells was evaluated by oil red O staining. The contents of total cholesterol (TC) and free cholesterol (FC) were assessed with commercial kits. Fluorescent imaging was observed with a fluorescent inverted microscope. The capacity of cholesterol efflux was measured with a fluorescent plate reader, and the transcript and protein levels of ABCA1, ABCG1, and SR-B1 were detected by Western blot and quantitative polymerase chain reactions (Q-PCRs). Cytokines in the medium were detected by ELISA and adjusted by total cellular proteins. The results showed that TP decreased ox-LDL-induced cholesterol deposition and foam cell formation by promoting cholesterol efflux to apoA1, which was related to the upregulation of ABCA1 and ABCG1. Moreover, TP decreased the secretion of ox-LDL-induced tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), and monocyte chemotactic protein-1 (MCP-1), an important profoam cell cytokine in atherosclerosis.
Collapse
|
10
|
González F, Considine RV, Abdelhadi OA, Xue J, Acton AJ. Saturated fat ingestion stimulates proatherogenic inflammation in polycystic ovary syndrome. Am J Physiol Endocrinol Metab 2021; 321:E689-E701. [PMID: 34632798 PMCID: PMC8782660 DOI: 10.1152/ajpendo.00213.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Inflammation and dyslipidemia are often present in polycystic ovary syndrome (PCOS). We determined the effect of saturated fat ingestion on circulating heat shock protein-70 (HSP-70) and mononuclear cell (MNC) toll-like receptor-2 (TLR2) gene expression, activator protein-1 (AP-1) activation, and matrix matalloproteinase-2 (MMP-2) protein in women with PCOS. Twenty reproductive-age women with PCOS (10 lean, 10 with obesity) and 20 ovulatory controls (10 lean, 10 with obesity) participated in the study. HSP-70 was measured in serum and TLR2 mRNA and protein, AP-1 activation, and MMP-2 protein were quantified in MNC from blood drawn while fasting and 2, 3, and 5 h after saturated fat ingestion. Insulin sensitivity was derived from an oral glucose tolerance test (ISOGTT). Androgen secretion was assessed from blood drawn while fasting and 24, 48, and 72 h after human chorionic gonadotropin (HCG) administration. In response to saturated fat ingestion, serum HSP-70, TLR2 gene expression, activated AP-1, and MMP-2 protein were greater in lean women with PCOS compared with lean controls and in women with PCOS and obesity compared with controls with obesity. Both PCOS groups exhibited lower ISOGTT and greater HCG-stimulated androgen secretion compared with control subjects of their respective weight classes. Lipid-stimulated proatherogenic inflammation marker responses were negatively correlated with ISOGTT and positively correlated with abdominal adiposity and HCG-stimulated androgen secretion. In PCOS, saturated fat ingestion stimulates proatherogenic inflammation independent of obesity. This effect is greater when PCOS is combined with obesity compared with obesity alone. Abdominal adiposity and hyperandrogenism may perpetuate proatherogenic inflammation.NEW & NOTEWORTHY This paper demonstrates that in polycystic ovary syndrome (PCOS), ingestion of saturated fat triggers a molecular pathway of inflammation known to drive atherogenesis. This effect is independent of obesity as it occurs in lean women with PCOS and not in lean ovulatory control subjects. Furthermore, the combined effects of PCOS and obesity are greater compared with obesity alone.
Collapse
Affiliation(s)
- Frank González
- Department of Obstetrics and Gynecology, University of Illinois Chicago College of Medicine, Chicago, Illinois
| | - Robert V Considine
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ola A Abdelhadi
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jiaping Xue
- Department of Obstetrics and Gynecology, University of Illinois Chicago College of Medicine, Chicago, Illinois
| | - Anthony J Acton
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
11
|
Miranda-Bautista J, Rodríguez-Feo JA, Puerto M, López-Cauce B, Lara JM, González-Novo R, Martín-Hernández D, Ferreiro-Iglesias R, Bañares R, Menchén L. Liver X Receptor Exerts Anti-Inflammatory Effects in Colonic Epithelial Cells via ABCA1 and Its Expression Is Decreased in Human and Experimental Inflammatory Bowel Disease. Inflamm Bowel Dis 2021; 27:1661-1673. [PMID: 33609028 DOI: 10.1093/ibd/izab034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Liver X receptor (LXR) exerts anti-inflammatory effects in macrophages. The aim of this study was to explore the expression and function of LXR in the colonic epithelium under inflammatory conditions. METHODS The expression of LXR was explored by Western blot and immunohistochemistry in colonic biopsies from patients diagnosed with inflammatory bowel disease (IBD) and control patients. In addition, LXR and its target gene expression were analyzed in the colon from interleukin (IL)-10-deficient (IL-10-/-) and wild-type mice. Caco-2 cells were pretreated with the synthetic LXR agonist GW3965 and further challenged with IL-1β, the expression of IL-8 and chemokine (C-C motif) ligand (CCL)-28 chemokines, the activation of mitogen-activated protein (MAP) kinases, and the nuclear translocation of the p65 subunit of nuclear factor kappa B was evaluated. Glibenclamide was used as an ABCA1 antagonist. RESULTS We found that LXR expression was downregulated in colonic samples from patients with IBD and IL-10-/- mice. The nuclear positivity of LXR inversely correlated with ulcerative colitis histologic activity. Colonic IL-1β mRNA levels negatively correlated with both LXRα and LXRβ in the colon of IL-10-/- mice, where a decreased mRNA expression of the LXR target genes ABCA1 and FAS was shown. In addition, IL-1β decreased the expression of the LXR target gene ABCA1 in cultured intestinal epithelial cells. The synthetic LXR agonist GW3965 led to a decreased nuclear positivity of the p65 subunit of nuclear factor kappa B, a phosphorylation ratio of the p44-42 MAP kinase, and the expression of CCL-28 and IL-8 in IL-1β-stimulated Caco-2 cells. The pharmacological inhibition of ABCA1 increased the phosphorylation of p44-42 after GW3965 treatment and IL-1β stimulation. CONCLUSIONS The LXR-ABCA1 pathway exerts anti-inflammatory effects in intestinal epithelial cells and is impaired in the colonic mucosa of patients with IBD and IL-10-/- mice.
Collapse
Affiliation(s)
- José Miranda-Bautista
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón-Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Juan A Rodríguez-Feo
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón-Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Marta Puerto
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón-Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Centro de Investigación en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - Beatriz López-Cauce
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón-Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - José M Lara
- Servicio de Anatomía Patológica, Hospital General Universitario Gregorio Marañón-Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Raquel González-Novo
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón-Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - David Martín-Hernández
- Servicio de Psiquiatría del Niño y del Adolescente, Hospital General Universitario Gregorio Marañón-Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Rafael Bañares
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón-Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Centro de Investigación en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain.,Departamento de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis Menchén
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón-Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Centro de Investigación en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain.,Departamento de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
12
|
Abstract
Mesenchymal stem cells have the ability to differentiate into several cell types when exposed to determined substances, including oxysterols. Oxysterols are cholesterol products derived from its auto-oxidation by reactive species or from enzymatic action. They are present in the body in low quantities under physiological conditions and exhibit several physiological and pharmacological actions according to both the types of oxysterol and tissue. Some of them are cytotoxic while others have been shown to promote cell differentiation through the action on several different receptors, such as nuclear LXR receptors and Smoothened receptor ligands. Here, we review the main pathways by which oxysterols have been associated with cell differentiation and death of mesenchymal stem cells.
Collapse
|
13
|
Zhang H, Bai Z, Zhu L, Liang Y, Fan X, Li J, Wen H, Shi T, Zhao Q, Wang Z. Hydrogen sulfide donors: Therapeutic potential in anti-atherosclerosis. Eur J Med Chem 2020; 205:112665. [DOI: 10.1016/j.ejmech.2020.112665] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/09/2020] [Accepted: 07/12/2020] [Indexed: 12/15/2022]
|
14
|
Anand PK. Lipids, inflammasomes, metabolism, and disease. Immunol Rev 2020; 297:108-122. [PMID: 32562313 DOI: 10.1111/imr.12891] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/19/2020] [Accepted: 05/27/2020] [Indexed: 12/19/2022]
Abstract
Inflammasomes are multi-protein complexes that regulate the cleavage of cysteine protease caspase-1, secretion of inflammatory cytokines, and induction of inflammatory cell death, pyroptosis. Several members of the nod-like receptor family assemble inflammasome in response to specific ligands. An exception to this is the NLRP3 inflammasome which is activated by structurally diverse entities. Recent studies have suggested that NLRP3 might be a sensor of cellular homeostasis, and any perturbation in distinct metabolic pathways results in the activation of this inflammasome. Lipid metabolism is exceedingly important in maintaining cellular homeostasis, and it is recognized that cells and tissues undergo extensive lipid remodeling during activation and disease. Some lipids are involved in instigating chronic inflammatory diseases, and new studies have highlighted critical upstream roles for lipids, particularly cholesterol, in regulating inflammasome activation implying key functions for inflammasomes in diseases with defective lipid metabolism. The focus of this review is to highlight how lipids regulate inflammasome activation and how this leads to the progression of inflammatory diseases. The key roles of cholesterol metabolism in the activation of inflammasomes have been comprehensively discussed. Besides, the roles of oxysterols, fatty acids, phospholipids, and lipid second messengers are also summarized in the context of inflammasomes. The overriding theme is that lipid metabolism has numerous but complex functions in inflammasome activation. A detailed understanding of this area will help us develop therapeutic interventions for diseases where dysregulated lipid metabolism is the underlying cause.
Collapse
Affiliation(s)
- Paras K Anand
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
15
|
Castaño D, Rattanasopa C, Monteiro-Cardoso VF, Corlianò M, Liu Y, Zhong S, Rusu M, Liehn EA, Singaraja RR. Lipid efflux mechanisms, relation to disease and potential therapeutic aspects. Adv Drug Deliv Rev 2020; 159:54-93. [PMID: 32423566 DOI: 10.1016/j.addr.2020.04.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
Lipids are hydrophobic and amphiphilic molecules involved in diverse functions such as membrane structure, energy metabolism, immunity, and signaling. However, altered intra-cellular lipid levels or composition can lead to metabolic and inflammatory dysfunction, as well as lipotoxicity. Thus, intra-cellular lipid homeostasis is tightly regulated by multiple mechanisms. Since most peripheral cells do not catabolize cholesterol, efflux (extra-cellular transport) of cholesterol is vital for lipid homeostasis. Defective efflux contributes to atherosclerotic plaque development, impaired β-cell insulin secretion, and neuropathology. Of these, defective lipid efflux in macrophages in the arterial walls leading to foam cell and atherosclerotic plaque formation has been the most well studied, likely because a leading global cause of death is cardiovascular disease. Circulating high density lipoprotein particles play critical roles as acceptors of effluxed cellular lipids, suggesting their importance in disease etiology. We review here mechanisms and pathways that modulate lipid efflux, the role of lipid efflux in disease etiology, and therapeutic options aimed at modulating this critical process.
Collapse
|
16
|
Luo J, Wang X, Jiang X, Liu C, Li Y, Han X, Zuo X, Li Y, Li N, Xu Y, Si S. Rutaecarpine derivative R3 attenuates atherosclerosis via inhibiting NLRP3 inflammasome-related inflammation and modulating cholesterol transport. FASEB J 2019; 34:1398-1411. [PMID: 31914630 DOI: 10.1096/fj.201900903rrr] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 10/31/2019] [Accepted: 11/12/2019] [Indexed: 12/17/2022]
Abstract
Atherosclerosis is a chronic disease characterized by lipid deposition and inflammatory response. NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome-facilitated inflammatory responses are crucial in the pathogenesis of atherosclerosis, and thus new therapeutic approaches are emerging that target NLRP3 and inflammation. Here, we explored the anti-atherosclerotic effect and mechanisms of a new rutaecarpine derivative, 5-deoxy-rutaecarpine (R3) in vitro and in vivo. R3 treatment attenuated atherosclerosis development and increased plaque stability in Apoe-/- mice fed a high-fat diet, and decreased levels of inflammatory mediators, such as interleukin-1β, in the serum of Apoe-/- mice and in oxidized low-density lipoprotein (ox-LDL)-stimulated murine macrophages. R3 treatment inhibited NLRP3 inflammasome activation in the livers of Apoe-/- mice and ox-LDL-stimulated murine macrophages by inhibiting NF-κB and MAPK pathways. Additionally, R3 significantly decreased total cholesterol in the serum and livers of Apoe-/- mice and promoted cholesterol efflux in murine macrophages through upregulating protein expression of ATP-binding cassette subfamily A member 1 and scavenger receptor class B type I/human CD36 and lysosomal integral membrane protein-II analogous-1. Our results demonstrated that R3 prevented atherosclerotic progression via attenuating NLRP3 inflammasome-related inflammation and modulating cholesterol transport.
Collapse
Affiliation(s)
- Jinque Luo
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinhai Jiang
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Liu
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongzhen Li
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaowan Han
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuan Zuo
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yining Li
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ni Li
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanni Xu
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuyi Si
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
[Liver X receptors define the immune response during general adaptation syndrome (GAS)]. C R Biol 2019; 342:136-141. [PMID: 31501012 DOI: 10.1016/j.crvi.2019.08.001] [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: 07/05/2019] [Revised: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 11/23/2022]
Abstract
The impact of stressful conditions on immunity seems mixed and at times counterbalanced. Such inconsistencies can often be attributed to the fact that the notion of stress has a very wide meaning and covers a large number of different situations. Research on liver X receptors using both natural and synthetic ligands may help to solve this conflict. When an infectious agent is present in a stressed body, LXR activation is likely to be a key element in the regulation of POMC, IFN-γ, and IL-18; moreover, it is a unique anti-inflammatory mode of action. They concurrently stimulate a non-specific immune reaction as they suppress inflammatory and autoimmune processes.
Collapse
|
18
|
Yang L, Li T, Zhao S, Zhang S. Niacin regulates apolipoprotein M expression via liver X receptor‑α. Mol Med Rep 2019; 20:3285-3291. [PMID: 31432166 PMCID: PMC6755166 DOI: 10.3892/mmr.2019.10557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/15/2019] [Indexed: 12/29/2022] Open
Abstract
Niacin is currently the most effective drug that increases HDL-C levels. Apolipoprotein M (ApoM) in humans is mainly found in plasma high-density lipoprotein (HDL). Little is known about the role played by niacin in ApoM expression. In this study, the effects of niacin on ApoM expression were assessed as well as the associated mechanism. Human liver cancer cell line HepG2 was treated with niacin alone or with liver X receptor-α (LXRα) inhibitor at multiple concentrations. The mRNA and protein expression of ApoM were assessed by qRT-PCR and western blotting. Specific LXRα shRNA was transfected into HepG2 cells to further evaluate the regulatory effects of LXRα on ApoM. An in vivo model was also established to investigate the LXRα inhibitor on the mouse ApoM levels. The comparisons among groups were evaluated using one-way ANOVA and Student-Newman-Keuls test. It was revealed that in HepG2 cells, niacin dose-dependently increased ApoM gene and protein expression levels. Niacin-induced upregulation of ApoM was attenuated by an LXRα inhibitor or LXRα shRNA, indicating that LXRα mediated this effect. Moreover, niacin treatment resulted in increased LXRα mRNA levels, in vivo and in vitro; niacin treatment resulted in increased ApoM gene and protein expression levels in mice. In conclusion, niacin upregulates ApoM expression by increasing LXRα expression in vivo and in vitro.
Collapse
Affiliation(s)
- Liu Yang
- International Medical Center, Geriatric Department, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Tie Li
- Department of Cardiovascular Medicine, Changsha Central Hospital, Changsha, Hunan 410001, P.R. China
| | - Shuiping Zhao
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Saidan Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
19
|
Rahmati-Ahmadabad S, Broom DR, Ghanbari-Niaki A, Shirvani H. Effects of exercise on reverse cholesterol transport: A systemized narrative review of animal studies. Life Sci 2019; 224:139-148. [PMID: 30922848 DOI: 10.1016/j.lfs.2019.03.058] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/22/2019] [Accepted: 03/23/2019] [Indexed: 12/20/2022]
Abstract
AIMS Reverse Cholesterol Transport (RCTr) is the mechanism by which excess cholesterol from peripheral tissues is transported to the liver for hepatobiliary excretion, thereby inhibiting foam cell formation and the development of atherosclerosis. Exercise affects RCTr, by influencing high-density lipoprotein cholesterol (HDL) through remodeling and by promoting hepatobiliary sterol excretion. The objectives of this systematized review of animal studies is to summarize the literature and provide an overview of the effects of chronic exercise (at least two weeks) on apolipoproteins (Apo A-I, Apo-E), Paraoxonase-1 (PON1), ATP-binding cassette transporters (ABCA1, ABCG1, ABCG4, ABCG5, ABCG8), scavenger receptor class B type I (SR-BI), cholesteryl ester transfer protein (CETP), low-density lipoprotein receptor (LDLr) and cholesterol 7 alpha-hydroxylase (CYP7A1) and Niemann-Pick C1-like 1 (NPC1L1). MATERIALS AND METHODS Three electronic databases (PubMed, Science Direct and Google Scholar) were searched for eligible studies conducted from the earliest available date to August 2018. KEY FINDINGS Most of studies investigate the effects of low to moderate intensity aerobic training on RCTr elements. The majority were on exercised rats undertaking moderate intensity aerobic training. SIGNIFICANCE This review highlights that moderate intensity and longer-term training has a greater effect on RCTr elements than low intensity training. There a few studies examining high intensity training which warrants further investigation.
Collapse
Affiliation(s)
| | - David Robert Broom
- Academy of Sport and Physical Activity, Faculty of Health and Wellbeing, Sheffield Hallam University, Sheffield, UK
| | - Abbass Ghanbari-Niaki
- Exercise Biochemistry Division, Faculty of Sport Sciences, University of Mazandaran, Babolsar, Mazandaran, Iran
| | - Hossein Shirvani
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
He X, Chen X, Wang L, Wang W, Liang Q, Yi L, Wang Y, Gao Q. Metformin ameliorates Ox-LDL-induced foam cell formation in raw264.7 cells by promoting ABCG-1 mediated cholesterol efflux. Life Sci 2018; 216:67-74. [PMID: 30218721 DOI: 10.1016/j.lfs.2018.09.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/29/2018] [Accepted: 09/12/2018] [Indexed: 01/22/2023]
Abstract
AIMS The accumulation of lipids in macrophages contributes to the development of atherosclerosis. Cholesterol efflux of lipid-loaded macrophages mediated by ATP binding cassette (ABC) cholesterol transporters, on the other hand, has been shown to attenuate atherosclerosis progression in patients with unknown mechanism. We therefore sought to test the effect of metformin that reduced cardiovascular risk in diabetic patients independent of its hypoglycemia effect on cholesterol transport in murine raw264.7 macrophages. MATERIALS AND METHODS Mouse raw264.7 macrophages were loaded with Ox-LDL (50 μg/ml) for 24 h before incubated with metformin (15 μM) for 24 h. Foam cell formation was assessed by Oil red staining and BIODIPY fluorescent staining as well as cholesterol-ester quantification by commercial kit. Cholesterol uptake and expression of scavenger receptors were detected by flow-cytometry. Cholesterol efflux capacity was measured by fluorescent plate-reader and ABC transporters were detected by Western Blots. Cytokines were detected by ELISA in supernatants and normalized by cellular lysates. KEY FINDINGS Our results showed that metformin decreased oxidized low-density lipoprotein (Ox-LDL)-induced cholesterol accumulation and foam cell formation by increasing cholesterol efflux to HDL, which was associated with an upregulation of ABC transporter ABCG-1. Moreover, metformin increased Ox-LDL-impaired IL-10 secretion, an important anti-foam cell cytokine in atherosclerosis. SIGNIFICANCE Our data highlighted the therapeutic potential of targeting macrophage cholesterol efflux with new or existing drugs for the possible reduction of foam cell formation in the prevention and treatment of diabetes-accelerated atherosclerosis.
Collapse
Affiliation(s)
- Xuan He
- Medical School of Nanjing University, Nanjing 210093, China
| | - Xiufang Chen
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Lei Wang
- Medical School of Nanjing University, Nanjing 210093, China
| | - Wenqing Wang
- Medical School of Nanjing University, Nanjing 210093, China
| | - Qiao Liang
- Medical School of Nanjing University, Nanjing 210093, China
| | - Long Yi
- Medical School of Nanjing University, Nanjing 210093, China
| | - Yong Wang
- Medical School of Nanjing University, Nanjing 210093, China
| | - Qian Gao
- Medical School of Nanjing University, Nanjing 210093, China.
| |
Collapse
|
21
|
Guerrini V, Prideaux B, Blanc L, Bruiners N, Arrigucci R, Singh S, Ho-Liang HP, Salamon H, Chen PY, Lakehal K, Subbian S, O’Brien P, Via LE, Barry CE, Dartois V, Gennaro ML. Storage lipid studies in tuberculosis reveal that foam cell biogenesis is disease-specific. PLoS Pathog 2018; 14:e1007223. [PMID: 30161232 PMCID: PMC6117085 DOI: 10.1371/journal.ppat.1007223] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/16/2018] [Indexed: 12/16/2022] Open
Abstract
Foam cells are lipid-laden macrophages that contribute to the inflammation and tissue damage associated with many chronic inflammatory disorders. Although foam cell biogenesis has been extensively studied in atherosclerosis, how these cells form during a chronic infectious disease such as tuberculosis is unknown. Here we report that, unlike the cholesterol-laden cells of atherosclerosis, foam cells in tuberculous lung lesions accumulate triglycerides. Consequently, the biogenesis of foam cells varies with the underlying disease. In vitro mechanistic studies showed that triglyceride accumulation in human macrophages infected with Mycobacterium tuberculosis is mediated by TNF receptor signaling through downstream activation of the caspase cascade and the mammalian target of rapamycin complex 1 (mTORC1). These features are distinct from the known biogenesis of atherogenic foam cells and establish a new paradigm for non-atherogenic foam cell formation. Moreover, they reveal novel targets for disease-specific pharmacological interventions against maladaptive macrophage responses.
Collapse
Affiliation(s)
- Valentina Guerrini
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Brendan Prideaux
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Landry Blanc
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Natalie Bruiners
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Riccardo Arrigucci
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Sukhwinder Singh
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Hsin Pin Ho-Liang
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Hugh Salamon
- Knowledge Synthesis, Berkeley, CA, United States of America
| | - Pei-Yu Chen
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Karim Lakehal
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Paul O’Brien
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Laura E. Via
- Tuberculosis Research Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Clifton E. Barry
- Tuberculosis Research Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Véronique Dartois
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | - Maria Laura Gennaro
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| |
Collapse
|
22
|
Genoula M, Marín Franco JL, Dupont M, Kviatcovsky D, Milillo A, Schierloh P, Moraña EJ, Poggi S, Palmero D, Mata-Espinosa D, González-Domínguez E, León Contreras JC, Barrionuevo P, Rearte B, Córdoba Moreno MO, Fontanals A, Crotta Asis A, Gago G, Cougoule C, Neyrolles O, Maridonneau-Parini I, Sánchez-Torres C, Hernández-Pando R, Vérollet C, Lugo-Villarino G, Sasiain MDC, Balboa L. Formation of Foamy Macrophages by Tuberculous Pleural Effusions Is Triggered by the Interleukin-10/Signal Transducer and Activator of Transcription 3 Axis through ACAT Upregulation. Front Immunol 2018; 9:459. [PMID: 29593722 PMCID: PMC5854656 DOI: 10.3389/fimmu.2018.00459] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/20/2018] [Indexed: 12/18/2022] Open
Abstract
The ability of Mycobacterium tuberculosis (Mtb) to persist in its human host relies on numerous immune evasion strategies, such as the deregulation of the lipid metabolism leading to the formation of foamy macrophages (FM). Yet, the specific host factors leading to the foamy phenotype of Mtb-infected macrophages remain unknown. Herein, we aimed to address whether host cytokines contribute to FM formation in the context of Mtb infection. Our approach is based on the use of an acellular fraction of tuberculous pleural effusions (TB-PE) as a physiological source of local factors released during Mtb infection. We found that TB-PE induced FM differentiation as observed by the increase in lipid bodies, intracellular cholesterol, and expression of the scavenger receptor CD36, as well as the enzyme acyl CoA:cholesterol acyl transferase (ACAT). Importantly, interleukin-10 (IL-10) depletion from TB-PE prevented the augmentation of all these parameters. Moreover, we observed a positive correlation between the levels of IL-10 and the number of lipid-laden CD14+ cells among the pleural cells in TB patients, demonstrating that FM differentiation occurs within the pleural environment. Downstream of IL-10 signaling, we noticed that the transcription factor signal transducer and activator of transcription 3 was activated by TB-PE, and its chemical inhibition prevented the accumulation of lipid bodies and ACAT expression in macrophages. In terms of the host immune response, TB-PE-treated macrophages displayed immunosuppressive properties and bore higher bacillary loads. Finally, we confirmed our results using bone marrow-derived macrophage from IL-10-/- mice demonstrating that IL-10 deficiency partially prevented foamy phenotype induction after Mtb lipids exposure. In conclusion, our results evidence a role of IL-10 in promoting the differentiation of FM in the context of Mtb infection, contributing to our understanding of how alterations of the host metabolic factors may favor pathogen persistence.
Collapse
Affiliation(s)
- Melanie Genoula
- Laboratorio de Inmunología de Enfermedades Respiratorias, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina
| | - José Luis Marín Franco
- Laboratorio de Inmunología de Enfermedades Respiratorias, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina
| | - Maeva Dupont
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina.,Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Denise Kviatcovsky
- Laboratorio de Inmunología de Enfermedades Respiratorias, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina
| | - Ayelén Milillo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Pablo Schierloh
- Laboratorio de Inmunología de Enfermedades Respiratorias, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina
| | - Eduardo Jose Moraña
- Instituto Prof. Dr. Raúl Vaccarezza, Hospital de Infecciosas Dr. F. J. Muñiz, Buenos Aires, Argentina
| | - Susana Poggi
- Instituto Prof. Dr. Raúl Vaccarezza, Hospital de Infecciosas Dr. F. J. Muñiz, Buenos Aires, Argentina
| | - Domingo Palmero
- Instituto Prof. Dr. Raúl Vaccarezza, Hospital de Infecciosas Dr. F. J. Muñiz, Buenos Aires, Argentina
| | - Dulce Mata-Espinosa
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Erika González-Domínguez
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Juan Carlos León Contreras
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Paula Barrionuevo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Bárbara Rearte
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Marlina Olyissa Córdoba Moreno
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | | | - Agostina Crotta Asis
- Laboratory of Physiology and Genetics of Actinomycetes, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Gabriela Gago
- Laboratory of Physiology and Genetics of Actinomycetes, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Céline Cougoule
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina.,Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina.,Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Isabelle Maridonneau-Parini
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina.,Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Carmen Sánchez-Torres
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Christel Vérollet
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina.,Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Geanncarlo Lugo-Villarino
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina.,Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - María Del Carmen Sasiain
- Laboratorio de Inmunología de Enfermedades Respiratorias, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina
| | - Luciana Balboa
- Laboratorio de Inmunología de Enfermedades Respiratorias, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Toulouse, France.,International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina
| |
Collapse
|
23
|
Sanmarco LM, Eberhardt N, Ponce NE, Cano RC, Bonacci G, Aoki MP. New Insights into the Immunobiology of Mononuclear Phagocytic Cells and Their Relevance to the Pathogenesis of Cardiovascular Diseases. Front Immunol 2018; 8:1921. [PMID: 29375564 PMCID: PMC5767236 DOI: 10.3389/fimmu.2017.01921] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/14/2017] [Indexed: 12/18/2022] Open
Abstract
Macrophages are the primary immune cells that reside within the myocardium, suggesting that these mononuclear phagocytes are essential in the orchestration of cardiac immunity and homeostasis. Independent of the nature of the injury, the heart triggers leukocyte activation and recruitment. However, inflammation is harmful to this vital terminally differentiated organ with extremely poor regenerative capacity. As such, cardiac tissue has evolved particular strategies to increase the stress tolerance and minimize the impact of inflammation. In this sense, growing evidences show that mononuclear phagocytic cells are particularly dynamic during cardiac inflammation or infection and would actively participate in tissue repair and functional recovery. They respond to soluble mediators such as metabolites or cytokines, which play central roles in the timing of the intrinsic cardiac stress response. During myocardial infarction two distinct phases of monocyte influx have been identified. Upon infarction, the heart modulates its chemokine expression profile that sequentially and actively recruits inflammatory monocytes, first, and healing monocytes, later. In the same way, a sudden switch from inflammatory macrophages (with microbicidal effectors) toward anti-inflammatory macrophages occurs within the myocardium very shortly after infection with Trypanosoma cruzi, the causal agent of Chagas cardiomyopathy. While in sterile injury, healing response is necessary to stop tissue damage; during an intracellular infection, the anti-inflammatory milieu in infected hearts would promote microbial persistence. The balance of mononuclear phagocytic cells seems to be also dynamic in atherosclerosis influencing plaque initiation and fate. This review summarizes the participation of mononuclear phagocyte system in cardiovascular diseases, keeping in mind that the immune system evolved to promote the reestablishment of tissue homeostasis following infection/injury, and that the effects of different mediators could modulate the magnitude and quality of the immune response. The knowledge of the effects triggered by diverse mediators would serve to identify new therapeutic targets in different cardiovascular pathologies.
Collapse
Affiliation(s)
- Liliana Maria Sanmarco
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| | - Natalia Eberhardt
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| | - Nicolás Eric Ponce
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Laboratorio de Neuropatología Experimental, Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Roxana Carolina Cano
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Universidad Católica de Córdoba, Unidad Asociada Área Ciencias Agrarias, Ingeniería, Ciencias Biológicas y de la Salud, Facultad de Ciencias Químicas, Córdoba, Argentina
| | - Gustavo Bonacci
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| | - Maria Pilar Aoki
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| |
Collapse
|
24
|
Herbert KE, Erridge C. Regulation of low-density lipoprotein cholesterol by intestinal inflammation and the acute phase response. Cardiovasc Res 2017; 114:226-232. [DOI: 10.1093/cvr/cvx237] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
AbstractSystemic inflammation, induced by disease or experimental intervention, is well established to result in elevated levels of circulating triglycerides, and reduced levels of high-density lipoprotein-cholesterol (HDL-C), in most mammalian species. However, the relationship between inflammation and low-density lipoprotein-cholesterol (LDL-C) concentrations is less clear. Most reports indicate that systemic inflammation, as observed during sepsis or following high dose experimental endotoxaemia, lowers total, and LDL-C in man. However, isolated reports have suggested that certain inflammatory conditions are associated with increased LDL-C. In this review, we summarize the emerging evidence that low-grade inflammation specifically of intestinal origin may be associated with increased serum LDL-C levels. Preliminary insights into potential mechanisms that may mediate these effects, including those connecting inflammation to trans-intestinal cholesterol efflux (TICE), are considered. We conclude that this evidence supports the potential downregulation of major mediators of TICE by inflammatory mediators in vitro and during intestinal inflammation in vivo. The TICE-inflammation axis therefore merits further study in terms of its potential to regulate serum LDL-C, and as a readily druggable target for hypercholesterolaemia.
Collapse
Affiliation(s)
- Karl E Herbert
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Groby Road, Leicester, Leicestershire, LE3 9QP, UK
| | - Clett Erridge
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Groby Road, Leicester, Leicestershire, LE3 9QP, UK
- Department of Biomedical and Forensic Sciences, Anglia Ruskin University, East Road, Cambridge, Cambridgeshire, CB1 1PT, UK
| |
Collapse
|
25
|
Luo T, Hu J, Xi D, Xiong H, He W, Liu J, Li M, Lu H, Zhao J, Lai W, Guo Z. Lck Inhibits Heat Shock Protein 65-Mediated Reverse Cholesterol Transport in T Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:3861-3870. [PMID: 27742830 DOI: 10.4049/jimmunol.1502710] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 09/20/2016] [Indexed: 02/05/2023]
Abstract
Previously, we reported that heat shock protein (HSP)65 impairs the effects of high-density lipoprotein on macrophages. We also showed that immune response activation adversely affects reverse cholesterol transport (RCT). In this study, we investigated the effects of the Src family kinase lymphocyte-specific protein tyrosine kinase (Lck) and elucidated the mechanism underlying HSP65-regulated cholesterol efflux in T cells. We evaluated cell proliferation, Lck expression, and inflammatory cytokine production in Jurkat cells and CD4+ T cells. HSP65-mediated inhibition of RCT was assessed by evaluating ABCA1, ABCG1, SR-BI, PPAR-γ, and liver X receptor-α expression. A dose-dependent relationship was found between the levels of these proteins and the suppression of cholesterol efflux. Stimulation of Lck-silenced T cells with ionomycin resulted in a decrease in intracellular calcium levels. Treatment of Jurkat cells with PP2, an inhibitor of Src family kinase, inhibited calcium-induced, but not PMA-induced, ERK phosphorylation. NF-κB activation in response to PMA was minimally inhibited in cells stimulated with PP2. HSP65 failed to trigger downstream ERK or JNK phosphorylation or to activate NF-κB or protein kinase C-γ in Lck-silenced cells. Additionally, elevation of intracellular calcium was also impaired. However, HSP65 significantly enhanced cholesterol efflux and decreased cellular cholesterol content by inducing the expression of cholesterol transport proteins in Lck-silenced cells. The treatment of Jurkat cells with PP2 also inhibited cell proliferation and promoted RCT. In conclusion, Lck is a key molecule in the TCR signaling cascade that inhibits cholesterol efflux and upregulates intracellular cholesterol ester content in T cells. Our results demonstrate that the immune response plays a previously unrecognized role in RCT.
Collapse
Affiliation(s)
- Tiantian Luo
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jing Hu
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Dan Xi
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Haowei Xiong
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Wenshuai He
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jichen Liu
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Menghao Li
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Hao Lu
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jinzhen Zhao
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Wenyan Lai
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Zhigang Guo
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Studies have shown that chronic inflammatory disorders, such as rheumatoid arthritis, systemic lupus erythematosus, and psoriasis are associated with an increased risk of atherosclerotic cardiovascular disease. The mechanism by which inflammation increases cardiovascular disease is likely multifactorial but changes in HDL structure and function that occur during inflammation could play a role. RECENT FINDINGS HDL levels decrease with inflammation and there are marked changes in HDL-associated proteins. Serum amyloid A markedly increases whereas apolipoprotein A-I, lecithin:cholesterol acyltransferase, cholesterol ester transfer protein, paraoxonase 1, and apolipoprotein M decrease. The exact mechanism by which inflammation decreases HDL levels is not defined but decreases in apolipoprotein A-I production, increases in serum amyloid A, increases in endothelial lipase and secretory phospholipase A2 activity, and decreases in lecithin:cholesterol acyltransferase activity could all contribute. The changes in HDL induced by inflammation reduce the ability of HDL to participate in reverse cholesterol transport and protect LDL from oxidation. SUMMARY During inflammation multiple changes in HDL structure occur leading to alterations in HDL function. In the short term, these changes may be beneficial resulting in an increase in cholesterol in peripheral cells to improve host defense and repair but over the long term these changes may increase the risk of atherosclerosis.
Collapse
Affiliation(s)
- Kenneth R Feingold
- Metabolism Section, Department of Veterans Affairs Medical Center, University of California San Francisco, San Francisco, California, USA
| | | |
Collapse
|
27
|
Berbee JF, Havekes LM, Rensen PC. Apolipoproteins modulate the inflammatory response to lipopolysaccharide. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519050110020501] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An increasing body of evidence demonstrates a close interplay between lipoprotein metabolism and sepsis. Sepsis results in an increase of plasma triglycerides within VLDL as a consequence of an enhanced hepatic VLDL production and/or inhibited peripheral and hepatic VLDL clearance. In contrast, sepsis decreases plasma cholesterol within LDL and mainly HDL. The decrease in HDL is accompanied by a loss of mainly apoAI-containing particles, an almost total loss of apoCI, and an increase in apoE-containing HDL, as related to the effect of LPS on a wide range of apolipoproteins, plasma enzymes, lipid transfer factors, and receptors that are involved in HDL metabolism. Reciprocally, all lipoprotein classes have been shown to bind LPS and to attenuate the biological response to LPS in vitro and in rodents. Moreover, triglyceride-rich lipoproteins protect rodents against death from LPS and bacterial sepsis. Accumulating evidence indicates that apolipoproteins such as apoE and apoAI, and not the lipid moieties of the particles, may be responsible for these protective effects of lipoproteins. Therefore, to increase our understanding of the complex interaction between lipoprotein metabolism and sepsis, further studies that address the specific roles of apolipoproteins in sepsis are warranted.
Collapse
Affiliation(s)
- Jimmy F.P. Berbee
- TNO-Quality of Life, Department of Biomedical Research, Gaubius Laboratory, Leiden, The Netherlands, Department of General Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Louis M. Havekes
- TNO-Quality of Life, Department of Biomedical Research, Gaubius Laboratory, Leiden, The Netherlands, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C.N. Rensen
- TNO-Quality of Life, Department of Biomedical Research, Gaubius Laboratory, Leiden, The Netherlands, Department of General Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands,
| |
Collapse
|
28
|
Liu H, Pathak P, Boehme S, Chiang JL. Cholesterol 7α-hydroxylase protects the liver from inflammation and fibrosis by maintaining cholesterol homeostasis. J Lipid Res 2016; 57:1831-1844. [PMID: 27534992 DOI: 10.1194/jlr.m069807] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Indexed: 12/30/2022] Open
Abstract
Cholesterol 7α-hydroxylase (CYP7A1) plays a critical role in control of bile acid and cholesterol homeostasis. Bile acids activate farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5) to regulate lipid, glucose, and energy metabolism. However, the role of bile acids in hepatic inflammation and fibrosis remains unclear. In this study, we showed that adenovirus-mediated overexpression of Cyp7a1 ameliorated lipopolysaccharide (LPS)-induced inflammatory cell infiltration and pro-inflammatory cytokine production in WT and TGR5-deficient (Tgr5-/-) mice, but not in FXR-deficient (Fxr-/-) mice, suggesting that bile acid signaling through FXR protects against hepatic inflammation. Nuclear factor κ light-chain enhancer of activated B cells (NF-κB)-luciferase reporter assay showed that FXR agonists significantly inhibited TNF-α-induced NF-κB activity. Furthermore, chromatin immunoprecipitation and mammalian two-hybrid assays showed that ligand-activated FXR interacted with NF-κB and blocked recruitment of steroid receptor coactivator-1 to cytokine promoter and resulted in inhibition of NF-κB activity. Methionine/choline-deficient (MCD) diet increased hepatic inflammation, free cholesterol, oxidative stress, apoptosis, and fibrosis in CYP7A1-deficient (Cyp7a1-/-) mice compared with WT mice. Remarkably, adenovirus-mediated overexpression of Cyp7a1 effectively reduced hepatic free cholesterol and oxidative stress and reversed hepatic inflammation and fibrosis in MCD diet-fed Cyp7a1-/- mice. Current studies suggest that increased Cyp7a1 expression and bile acid synthesis ameliorate hepatic inflammation through activation of FXR, whereas reduced bile acid synthesis aggravates MCD diet-induced hepatic inflammation and fibrosis. Maintaining bile acid and cholesterol homeostasis is important for protecting against liver injury and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Hailiang Liu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Preeti Pathak
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Shannon Boehme
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - JohnY L Chiang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272.
| |
Collapse
|
29
|
Wehmeier KR, Kurban W, Chandrasekharan C, Onstead-Haas L, Mooradian AD, Haas MJ. Inhibition of ABCA1 Protein Expression and Cholesterol Efflux by TNF α in MLO-Y4 Osteocytes. Calcif Tissue Int 2016; 98:586-95. [PMID: 26759003 DOI: 10.1007/s00223-015-0106-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/28/2015] [Indexed: 01/12/2023]
Abstract
Hip fracture and myocardial infarction cause significant morbidity and mortality. In vivo studies raising serum cholesterol levels as well as pro-inflammatory cytokines such as TNF α manifest bone loss and atherosclerotic vascular disease, suggesting that abnormalities of cholesterol transport may contribute to osteoporosis. We used the mouse osteocyte cell line (MLO-Y4) to investigate the effects of TNF α on the expression of cholesterol acceptor proteins such as apolipoprotein A-I (apo A-I) and apolipoprotein E (apo E), as well as on the cholesterol transporters ATP-binding cassette-1 (ABCA1), scavenger receptor class B type 1 (SRB1), and cluster of differentiation 36 (CD36). MLO-Y4 cells do not express apo A-I or apo E; however, they do express all three cholesterol transporters (ABCA1, SRB1, and CD36). Treatment of MLO-Y4 cells with TNF α had no effect on SRB1, CD36, and osteocalcin levels; however, TNF α reduced ABCA1 protein levels in a dose-dependent manner and cholesterol efflux to apo A-I. Interestingly, TNF α treatment increased ABCA1 promoter activity and ABCA1 mRNA levels, and increased liver X receptor α protein expression, but had no effect on retinoid X receptor α and retinoic acid receptor α levels. Pharmacological inhibition of p38 mitogen-activated protein (MAP) kinase, but not c-jun-N-terminal kinase 1 or mitogen-activated protein kinase (MEK), restored ABCA1 protein levels in TNF α-treated cells. These results suggest that pro-inflammatory cytokines regulate cholesterol metabolism in osteocytes in part by suppressing ABCA1 levels post-translationally in a p38 MAP kinase-dependent manner.
Collapse
Affiliation(s)
- Kent R Wehmeier
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida College of Medicine, 653-1 West 8th Street, L14, Jacksonville, FL, 32209, USA
| | - William Kurban
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida College of Medicine, 653-1 West 8th Street, L14, Jacksonville, FL, 32209, USA
| | - Chandrikha Chandrasekharan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida College of Medicine, 653-1 West 8th Street, L14, Jacksonville, FL, 32209, USA
| | - Luisa Onstead-Haas
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida College of Medicine, 653-1 West 8th Street, L14, Jacksonville, FL, 32209, USA
| | - Arshag D Mooradian
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida College of Medicine, 653-1 West 8th Street, L14, Jacksonville, FL, 32209, USA
| | - Michael J Haas
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida College of Medicine, 653-1 West 8th Street, L14, Jacksonville, FL, 32209, USA.
| |
Collapse
|
30
|
Ding H, Li Y, Feng Y, Chen J, Zhong X, Wang N, Wang W, Zhang P, Wang L. LXR agonist T0901317 upregulates thrombomodulin expression in glomerular endothelial cells by inhibition of nuclear factor‑κB. Mol Med Rep 2016; 13:4888-96. [PMID: 27082844 DOI: 10.3892/mmr.2016.5138] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 01/25/2016] [Indexed: 11/06/2022] Open
Abstract
Dysfunction of glomerular endothelial cells (GECs) induces a variety of symptoms, including proteinuria, inflammation, vascular diseases, fibrosis and thrombosis. Thrombomodulin (TM) acts as a vasoprotective molecule on the surface of the vascular endothelial cells to maintain the homeostasis of the endothelial microenvironment by suppressing cellular proliferation, adhesion and inflammatory responses. Liver X receptor (LXR), a nuclear receptor (NR) and a bile acid‑activated transcription factor, regulates metabolism and cholesterol transport, vascular tension and inflammation. Previous studies indicated that TM expression is upregulated by various NRs; however, it is unclear whether pharmacological modulation of LXR may affect TM expression and GEC function. The current study revealed that LXR activation by its agonist, T0901317, upregulates the expression and activity of TM. This effect was mediated specifically through LXR‑α, and not through LXR‑β. Additionally, T0901317 treatment inhibited nuclear factor‑κB (NF‑κB) signaling and the secretion of high glucose‑induced proinflammatory mediators, including tumor necrosis factor‑α and interleukin‑1β in GECs. Co‑immunoprecipitation experiments determined that treatment with T0901317 enhances the interaction between LXR‑α and the transcriptional coactivator, p300, in GEC extracts. The present findings suggest that NF‑κB may be a negative regulator of TM expression, and its removal may contribute to TM gene expression, particularly when in competition with the T0901317‑enhanced formation of the LXR/p300 complex. Therefore, LXR may be a novel molecular target for manipulating TM in GECs, which may advance the treatment of endothelial cell‑associated diseases.
Collapse
Affiliation(s)
- Hanlu Ding
- Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences, Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Yi Li
- Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences, Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Yunlin Feng
- Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences, Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Jin Chen
- Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences, Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Xiang Zhong
- Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences, Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Nan Wang
- Department of Nephrology, Chengdu Second People's Hospital, Chengdu, Sichuan 610041, P.R. China
| | - Wei Wang
- Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences, Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Ping Zhang
- Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences, Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Li Wang
- Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences, Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
31
|
Nyima T, Müller M, Hooiveld GJEJ, Morine MJ, Scotti M. Nonlinear transcriptomic response to dietary fat intake in the small intestine of C57BL/6J mice. BMC Genomics 2016; 17:106. [PMID: 26861690 PMCID: PMC4748552 DOI: 10.1186/s12864-016-2424-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 02/02/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND A high caloric diet, in conjunction with low levels of physical activity, promotes obesity. Many studies are available regarding the relation between dietary saturated fats and the etiology of obesity, but most focus on liver, muscle and white adipose tissue. Furthermore, the majority of transcriptomic studies seek to identify linear effects of an external stimulus on gene expression, although such an assumption does not necessarily hold. Our work assesses the dose-dependent effects of dietary fat intake on differential gene expression in the proximal, middle and distal sections of the small intestine in C57BL/6J mice. Gene expression is analyzed in terms of either linear or nonlinear responses to fat intake. RESULTS The highest number of differentially expressed genes was observed in the middle section. In all intestine sections, most of the identified processes exhibited a linear response to increasing fat intake. The relative importance of logarithmic and exponential responses was higher in the proximal and distal sections, respectively. Functional enrichment analysis highlighted a constantly linear regulation of acute-phase response along the whole small intestine, with up-regulation of Serpina1b. The study of gene expression showed that exponential down-regulation of cholesterol transport in the middle section is coupled with logarithmic up-regulation of cholesterol homeostasis. A shift from linear to exponential response was observed in genes involved in the negative regulation of caspase activity, from middle to distal section (e.g., Birc5, up-regulated). CONCLUSIONS The transcriptomic signature associated with inflammatory processes preserved a linear response in the whole small intestine (e.g., up-regulation of Serpina1b). Processes related to cholesterol homeostasis were particularly active in the middle small intestine and only the highest fat intake down-regulated cholesterol transport and efflux (with a key role played by the down-regulation of ATP binding cassette transporters). Characterization of nonlinear patterns of gene expression triggered by different levels of dietary fat is an absolute novelty in intestinal studies. This approach helps identifying which processes are overloaded (i.e., positive, logarithmic regulation) or arrested (i.e., negative, exponential regulation) in response to excessive fat intake, and can shed light on the relationships linking lipid intake to obesity and its associated molecular disturbances.
Collapse
Affiliation(s)
- Tenzin Nyima
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology, Rovereto, Italy.
| | - Michael Müller
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands. .,Norwich Medical School, University of East Anglia, Norwich, UK.
| | - Guido J E J Hooiveld
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands.
| | - Melissa J Morine
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology, Rovereto, Italy.
| | - Marco Scotti
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology, Rovereto, Italy. .,GEOMAR Helmholtz Centre for Ocean Research, Kiel, Germany.
| |
Collapse
|
32
|
Ngaosuwan K, Houngngam N, Limpisook P, Plengpanich W, Khovidhunkit W. Apolipoprotein A-V is not a major determinant of triglyceride levels during human sepsis. J Crit Care 2015; 30:727-31. [DOI: 10.1016/j.jcrc.2015.03.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/23/2015] [Accepted: 03/28/2015] [Indexed: 12/20/2022]
|
33
|
Warrier M, Shih DM, Burrows AC, Ferguson D, Gromovsky AD, Brown AL, Marshall S, McDaniel A, Schugar RC, Wang Z, Sacks J, Rong X, Vallim TDA, Chou J, Ivanova PT, Myers DS, Brown HA, Lee RG, Crooke RM, Graham MJ, Liu X, Parini P, Tontonoz P, Lusis AJ, Hazen SL, Temel RE, Brown JM. The TMAO-Generating Enzyme Flavin Monooxygenase 3 Is a Central Regulator of Cholesterol Balance. Cell Rep 2015; 10:326-338. [PMID: 25600868 DOI: 10.1016/j.celrep.2014.12.036] [Citation(s) in RCA: 296] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 11/24/2014] [Accepted: 12/16/2014] [Indexed: 12/15/2022] Open
Abstract
Circulating levels of the gut microbe-derived metabolite trimethylamine-N-oxide (TMAO) have recently been linked to cardiovascular disease (CVD) risk. Here, we performed transcriptional profiling in mouse models of altered reverse cholesterol transport (RCT) and serendipitously identified the TMAO-generating enzyme flavin monooxygenase 3 (FMO3) as a powerful modifier of cholesterol metabolism and RCT. Knockdown of FMO3 in cholesterol-fed mice alters biliary lipid secretion, blunts intestinal cholesterol absorption, and limits the production of hepatic oxysterols and cholesteryl esters. Furthermore, FMO3 knockdown stimulates basal and liver X receptor (LXR)-stimulated macrophage RCT, thereby improving cholesterol balance. Conversely, FMO3 knockdown exacerbates hepatic endoplasmic reticulum (ER) stress and inflammation in part by decreasing hepatic oxysterol levels and subsequent LXR activation. FMO3 is thus identified as a central integrator of hepatic cholesterol and triacylglycerol metabolism, inflammation, and ER stress. These studies suggest that the gut microbiota-driven TMA/FMO3/TMAO pathway is a key regulator of lipid metabolism and inflammation.
Collapse
Affiliation(s)
- Manya Warrier
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Diana M Shih
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amy C Burrows
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Daniel Ferguson
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Anthony D Gromovsky
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Amanda L Brown
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Stephanie Marshall
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Allison McDaniel
- Departments of Pathology and Biostatistics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Rebecca C Schugar
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Zeneng Wang
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Jessica Sacks
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Xin Rong
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | - Jeff Chou
- Departments of Pathology and Biostatistics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Pavlina T Ivanova
- Departments of Pharmacology and Biochemistry, The Vanderbilt Institute of Chemical Biology, Nashville, TN 37232, USA
| | - David S Myers
- Departments of Pharmacology and Biochemistry, The Vanderbilt Institute of Chemical Biology, Nashville, TN 37232, USA
| | - H Alex Brown
- Departments of Pharmacology and Biochemistry, The Vanderbilt Institute of Chemical Biology, Nashville, TN 37232, USA
| | - Richard G Lee
- Cardiovascular Group, Antisense Drug Discovery, Isis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Rosanne M Crooke
- Cardiovascular Group, Antisense Drug Discovery, Isis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Mark J Graham
- Cardiovascular Group, Antisense Drug Discovery, Isis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Xiuli Liu
- Department of Anatomical Pathology, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Paolo Parini
- Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, 141 86 Stockholm, Sweden
| | - Peter Tontonoz
- Howard Hughes Medical Institute; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aldon J Lusis
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stanley L Hazen
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Ryan E Temel
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536-0509, USA
| | - J Mark Brown
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA.
| |
Collapse
|
34
|
Liu W, Yin Y, Zhou Z, He M, Dai Y. OxLDL-induced IL-1 beta secretion promoting foam cells formation was mainly via CD36 mediated ROS production leading to NLRP3 inflammasome activation. Inflamm Res 2014; 63:33-43. [PMID: 24121974 DOI: 10.1007/s00011-013-0667-3] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 09/13/2013] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE IL-1β is a master switch of inflammation and plays an important role in the pathogenesis of vascular disease. During early atherosclerosis development, it is not clearly understood how oxidized low density lipoprotein (oxLDL)induced signaling pathways control NLRP3 inflammasome activation and produce IL-1β and promote foam cells formation. METHODS The study used THP-1 macrophage as cell model. Western blot quantified the oxLDL-induced NLRP3 inflammasome related proteins. The FACS detected the expression of SR-A and CD36 receptors on the cells, and caspase-1 activation in the cells. The DCFH-DA assayed the reactive oxygen species (ROS). Oil red O staining techniques examined the intracellular lipid droplet. RESULTS The OxLDL remarkably increased not only IL-1β mRNA transcription and pro-IL-1β protein synthesis but also IL-1β secretion in human macrophages. The activation of the NLRP3 inflammasome depended on oxLDL-induced generation of ROS, potassium efflux and cathepsin B activity. The OxLDL-induced ROS production that mediates IL-1β maturation mainly depended on the scavenger receptor of CD36 but not SR-A. The secreted IL-1β served as an autocrine function for promoting macrophage foam cells formation. CONCLUSIONS These findings suggest that oxLDL-induced NLRP3 inflammasome activation mainly depends on CD36 involved in the progression of atherosclerosis by promoting oxLDL-mediated inflammation and foam cell formation.
Collapse
|
35
|
Thirunavukkarasu S, de Silva K, Plain KM, J Whittington R. Role of host- and pathogen-associated lipids in directing the immune response in mycobacterial infections, with emphasis on Mycobacterium avium subsp. paratuberculosis. Crit Rev Microbiol 2014; 42:262-75. [PMID: 25163812 DOI: 10.3109/1040841x.2014.932327] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mycobacteria have a complex cell wall with a high lipid content that confers unique advantages for bacterial survival in the hostile host environment, leading to long-term infection. There is a wealth of evidence suggesting the role cell wall-associated lipid antigens play at the host-pathogen interface by contributing to bacterial virulence. One pathway that pathogenic mycobacteria use to subvert host immune pathways to their advantage is host cholesterol/lipid homeostasis. This review focuses on the possible role of pathogen- and host-associated lipids in the survival and persistence of pathogenic mycobacteria with emphasis on Mycobacterium avium subsp. paratuberculosis. We draw upon literature in diverse areas of infectious and metabolic diseases and explain a mechanism by which mycobacterial-induced changes in the host cellular energy state could account for phenomena that are a hallmark of chronic mycobacterial diseases.
Collapse
Affiliation(s)
| | - Kumudika de Silva
- a Faculty of Veterinary Science , University of Sydney , Camden , Australia
| | - Karren M Plain
- a Faculty of Veterinary Science , University of Sydney , Camden , Australia
| | | |
Collapse
|
36
|
Serban C, Muntean D, Mikhailids DP, Toth PP, Banach M. Dysfunctional HDL: the journey from savior to slayer. ACTA ACUST UNITED AC 2014. [DOI: 10.2217/clp.13.83] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
37
|
Feng X, Yuan Y, Wang C, Feng J, Yuan Z, Zhang X, Sui W, Hu P, Zheng P, Ye J. Autophagy involved in lipopolysaccharide-induced foam cell formation is mediated by adipose differentiation-related protein. Lipids Health Dis 2014; 13:10. [PMID: 24405744 PMCID: PMC3896829 DOI: 10.1186/1476-511x-13-10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/07/2014] [Indexed: 02/02/2023] Open
Abstract
Background Autophagy is an essential process for breaking down macromolecules and aged/damaged cellular organelles to maintain cellular energy balance and cellular nutritional status. The idea that autophagy regulates lipid metabolism is an emerging concept with important implications for atherosclerosis. However, the potential role of autophagy and its relationship with lipid metabolism in foam cell formation remains unclear. In this study, we found that autophagy was involved in the lipopolysaccharide (LPS)-induced the formation of foam cells and was at least partially dependent on adipose differentiation-related protein (ADRP). Method Foam cell formation was evaluated by Oil red O staining. Autophagic activity was determined by immunofluorescence and Western blotting. ADRP gene expression of ADRP was examined by real-time PCR (RT-PCR). The protein expression of ADRP and LC3 was measured using Western blotting analysis. Intracellular cholesterol and triglyceride levels in foam cells were quantitatively measured by enzymatic colorimetric assays. Results LPS promoted foam cell formation by inducing lipid accumulation in macrophages. The activation of autophagy with rapamycin (Rap) decreased intracellular cholesterol and triglyceride levels, whereas the inhibition of autophagy with 3-methyladenine (3MA) enhanced the accumulation of lipid droplets. Overexpression of ADRP alone increased the formation of foam cells and consequently autophagic activity. In contrast, the inhibitory effects of ADRP activity with siRNA suppressed the activation of autophagy. Taken together, we propose a novel role for ADRP in the regulation of macrophage autophagy during LPS stimulation. Conclusion We defined a new molecular pathway in which LPS-induced foam cell formation is regulated through autophagy. These findings facilitate the understanding of the role of autophagy in the development of atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jing Ye
- Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
38
|
Yu XH, Qian K, Jiang N, Zheng XL, Cayabyab FS, Tang CK. ABCG5/ABCG8 in cholesterol excretion and atherosclerosis. Clin Chim Acta 2014; 428:82-8. [DOI: 10.1016/j.cca.2013.11.010] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 11/07/2013] [Accepted: 11/09/2013] [Indexed: 12/23/2022]
|
39
|
Involvement of lipid droplets in hepatic responses to lipopolysaccharide treatment in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1357-67. [DOI: 10.1016/j.bbalip.2013.04.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 04/05/2013] [Accepted: 04/30/2013] [Indexed: 01/07/2023]
|
40
|
Salminen A, Pussinen PJ, Payne JB, Stoner JA, Jauhiainen M, Golub LM, Lee HM, Thompson DM, Sorsa T. Subantimicrobial-dose doxycycline treatment increases serum cholesterol efflux capacity from macrophages. Inflamm Res 2013; 62:711-20. [PMID: 23649042 DOI: 10.1007/s00011-013-0626-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 03/07/2013] [Accepted: 04/16/2013] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE Subantimicrobial-dose doxycycline (SDD) treatment has been reported to reduce the severity of chronic inflammation and to increase serum high-density lipoprotein cholesterol. In a double-blind, placebo-controlled clinical trial, we determined whether SDD affects the ability of serum to facilitate cholesterol removal from macrophages. METHODS Forty-five postmenopausal osteopenic women with periodontitis were randomly assigned to take placebo (n = 26) or doxycycline hyclate (20 mg, n = 19) tablets twice daily for 2 years. Serum samples were collected at baseline, 1-, and 2-year appointments. The cholesterol efflux capacity of serum from cultured human macrophages (THP-1) was measured. RESULTS SDD subjects demonstrated a significant increase in serum-mediated cholesterol efflux from macrophages at both time points compared to baseline (p < 0.04 for each). Mean cholesterol efflux levels over the first year of follow-up were 3.0 percentage points (unit change) higher among SDD subjects compared to placebo subjects (p = 0.010), while there was no significant difference in 2-year changes. There were no significant differences in the changes of apolipoprotein A-I, apolipoprotein A-II, or serum amyloid A levels between the groups. CONCLUSIONS Our results suggest that SDD treatment may reduce the risk of cardiovascular disease in this patient group by increasing the cholesterol efflux capacity of serum.
Collapse
Affiliation(s)
- Aino Salminen
- Institute of Dentistry, University of Helsinki, P.O. Box 63, 00014 Helsinki, Finland.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Dikkers A, Tietge UJF. The neglected cousin of the hepatocyte: how gallbladder epithelial cells might contribute to cholesterol gallstone formation. Dig Dis Sci 2013; 58:296-8. [PMID: 23371007 DOI: 10.1007/s10620-012-2541-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 12/19/2012] [Indexed: 12/09/2022]
|
42
|
Gao M, Zeng Y, Guan Y, Hu Z, Zhong D, Shen X, Zhang L, Xu Z, Gong W, Zhang Y, Zhang M, Zheng Y, He F. Activation of liver X receptor attenuates endothelin-1 expression in vascular endothelial cells. Int J Biochem Cell Biol 2012; 44:2299-307. [PMID: 23018104 DOI: 10.1016/j.biocel.2012.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 08/31/2012] [Accepted: 09/14/2012] [Indexed: 10/27/2022]
Abstract
Endothelin-1 (ET-1), predominantly produced by vascular endothelial cells (VECs), plays an important role in the pathogenesis of inflammatory diseases. Liver X receptor (LXR), a typical nuclear receptor, is known for inhibiting expression of inflammatory molecules. However, it remains unclear whether LXR suppresses ET-1 expression. In the present study, we showed that pretreatment with GW3965, a specific ligand of LXR, significantly attenuated lipopolysaccharide (LPS)-induced ET-1 in mice plasma. The in vitro experiments showed that both LXRα and β were expressed in human VECs, and they are functional as demonstrated by induction of the target gene ABCA1 after treatment with GW3965. Moreover, activation of LXR with GW3965 in human VECs dramatically attenuated the basal and LPS-stimulated ET-1 production at both transcriptional and translational levels. Luciferase reporter assays indicated that LXR activation suppressed the transcriptional activity of the human ET-1 gene promoter, and repressed the activity of a heterologous promoter driven by the response elements of activator-1 (AP-1) or nuclear factor-κB (NF-κB). Electrophoretic mobility shift and chromatin immunoprecipitation assays showed that activation of LXR reduced the binding of the transcriptional factors AP-1 and NF-κB to the ET-1 gene promoter region. In conclusion, activation of LXR represses ET-1 expression in vivo and in vitro, which may be involved in the negatively interfering with AP-1/NF-κB signaling. These results suggest that LXRs may serve as a novel molecular target for modulating ET-1 expression in VECs, and even for the treatment of ET-1-associated inflammatory diseases.
Collapse
Affiliation(s)
- Min Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
ten Freyhaus H, Calay ES, Yalcin A, Vallerie SN, Yang L, Calay ZZ, Saatcioglu F, Hotamisligil GS. Stamp2 controls macrophage inflammation through nicotinamide adenine dinucleotide phosphate homeostasis and protects against atherosclerosis. Cell Metab 2012; 16:81-9. [PMID: 22704678 PMCID: PMC4163924 DOI: 10.1016/j.cmet.2012.05.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 03/06/2012] [Accepted: 05/11/2012] [Indexed: 01/21/2023]
Abstract
The six-transmembrane protein Stamp2 plays an important role in metabolically triggered inflammation and insulin action. We report that Stamp2 is expressed in human and mouse macrophages, is regulated upon differentiation or activation, acts as an anti-inflammatory protein, and regulates foam cell formation. Absence of Stamp2 results in significant increases in cellular NADPH levels, and both NADPH homeostasis and the exaggerated inflammatory response of Stamp2(-/-) macrophages are rescued by exogenous wild-type but not by a reductase-deficient Stamp2 molecule. Chemical and genetic suppression of NADPH production in Stamp2(-/-) macrophages reverts the heightened inflammatory response. Stamp2 is detected in mouse and human atherosclerotic plaques, and its deficiency promotes atherosclerosis in mice. Furthermore, bone marrow transplantation experiments demonstrated that Stamp2 in myeloid cells is sufficient to protect against atherosclerosis. Our data reveal a role of Stamp2 in controlling intermediary metabolites to regulate inflammatory responses in macrophages and in progression of atherosclerosis.
Collapse
Affiliation(s)
- Henrik ten Freyhaus
- Department of Genetics, Harvard School of Public Health, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Ma CIJ, Beckstead JA, Thompson A, Hafiane A, Wang RHL, Ryan RO, Kiss RS. Tweaking the cholesterol efflux capacity of reconstituted HDL. Biochem Cell Biol 2012; 90:636-45. [PMID: 22607224 DOI: 10.1139/o2012-015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mechanisms to increase plasma high-density lipoprotein (HDL) or to promote egress of cholesterol from cholesterol-loaded cells (e.g., foam cells from atherosclerotic lesions) remain an important target to regress heart disease. Reconstituted HDL (rHDL) serves as a valuable vehicle to promote cellular cholesterol efflux in vitro and in vivo. rHDL were prepared with wild type apolipoprotein (apo) A-I and the rare variant, apoA-I Milano (M), and each apolipoprotein was reconstituted with phosphatidylcholine (PC) or sphingomyelin (SM). The four distinct rHDL generated were incubated with CHO cells, J774 macrophages, and BHK cells in cellular cholesterol efflux assays. In each cell type, apoA-I(M) SM-rHDL promoted the greatest cholesterol efflux. In BHK cells, the cholesterol efflux capacities of all four distinct rHDL were greatly enhanced by increased expression of ABCG1. Efflux to PC-containing rHDL was stimulated by transfection of a nonfunctional ABCA1 mutant (W590S), suggesting that binding to ABCA1 represents a competing interaction. This interpretation was confirmed by binding experiments. The data show that cholesterol efflux activity is dependent upon the apoA-I protein employed, as well as the phospholipid constituent of the rHDL. Future studies designed to optimize the efflux capacity of therapeutic rHDL may improve the value of this emerging intervention strategy.
Collapse
Affiliation(s)
- Cheng-I J Ma
- Cardiovascular Research Laboratories, Department of Medicine, Royal Victoria Hospital, 687 Pine Avenue West, Montreal, QC H3A 1A1, Canada
| | | | | | | | | | | | | |
Collapse
|
45
|
Azzam KM, Fessler MB. Crosstalk between reverse cholesterol transport and innate immunity. Trends Endocrinol Metab 2012; 23:169-78. [PMID: 22406271 PMCID: PMC3338129 DOI: 10.1016/j.tem.2012.02.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 01/30/2012] [Accepted: 02/01/2012] [Indexed: 02/06/2023]
Abstract
Although lipid metabolism and host defense are widely considered to be very divergent disciplines, compelling evidence suggests that host cell handling of self- and microbe-derived (e.g. lipopolysaccharide, LPS) lipids may have common evolutionary roots, and that they indeed may be inseparable processes. The innate immune response and the homeostatic network controlling cellular sterol levels are now known to regulate each other reciprocally, with important implications for several common diseases, including atherosclerosis. In the present review we discuss recent discoveries that provide new insight into the bidirectional crosstalk between reverse cholesterol transport and innate immunity, and highlight the broader implications of these findings for the development of therapeutics.
Collapse
Affiliation(s)
- Kathleen M Azzam
- Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | | |
Collapse
|
46
|
Abstract
Serum amyloid A (SAA), a protein originally of interest primarily to investigators focusing on AA amyloidogenesis, has become a subject of interest to a very broad research community. SAA is still a major amyloid research topic because AA amyloid, for which SAA is the precursor, is the prototypic model of in vivo amyloidogenesis and much that has been learned with this model has been applicable to much more common clinical types of amyloid. However, SAA has also become a subject of considerable interest to those studying (i) the synthesis and regulation of acute phase proteins, of which SAA is a prime example, (ii) the role that SAA plays in tissue injury and inflammation, a situation in which the plasma concentration of SAA may increase a 1000-fold, (iii) the influence that SAA has on HDL structure and function, because during inflammation the majority of SAA is an apolipoprotein of HDL, (iv) the influence that SAA may have on HDL's role in reverse cholesterol transport, and therefore, (v) SAA's potential role in atherogenesis. However, no physiological role for SAA, among many proposed, has been widely accepted. None the less from an evolutionary perspective SAA must have a critical physiological function conferring survival-value because SAA genes have existed for at least 500 million years and SAA's amino acid sequence has been substantially conserved. An examination of the published literature over the last 40 years reveals a great deal of conflicting data and interpretation. Using SAA's conserved amino acid sequence and the physiological effects it has while in its native structure, namely an HDL apolipoprotein, we argue that much of the confounding data and interpretation relates to experimental pitfalls not appreciated when working with SAA, a failure to appreciate the value of physiologic studies done in the 1970-1990 and a current major focus on putative roles of SAA in atherogenesis and chronic disease. When viewed from an evolutionary perspective, published data suggest that acute-phase SAA is part of a systemic response to injury to recycle and reuse cholesterol from destroyed and damaged cells. This is accomplished through SAA's targeted delivery of HDL to macrophages, and its suppression of ACAT, the enhancement of neutral cholesterol esterase and ABC transporters in macrophages. The recycling of cholesterol during serious injury, when dietary intake is restricted and there is an immediate and critical requirement of cholesterol in the generation of myriads of cells involved in inflammation and repair responses, is likely SAA's important survival role. Data implicating SAA in atherogenesis are not relevant to its evolutionary role. Furthermore, in apoE(-/-) mice, domains near the N- and C- termini of SAA inhibit the initiation and progression of aortic lipid lesions illustrating the conflicting nature of these two sets of data.
Collapse
|
47
|
Park Y, Pham TX, Lee J. Lipopolysaccharide represses the expression of ATP-binding cassette transporter G1 and scavenger receptor class B, type I in murine macrophages. Inflamm Res 2012; 61:465-72. [PMID: 22240665 DOI: 10.1007/s00011-011-0433-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 11/16/2011] [Accepted: 12/29/2011] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE AND DESIGN To investigate the regulation of cholesterol transporters, including ATP-binding cassette transporter A1 (ABCA1), ABCG1 and scavenger receptor class B, type I (SR-BI), by inflammatory stimuli in macrophages. MATERIALS AND TREATMENTS: RAW 264.7 macrophages and mouse peritoneal macrophages were treated with inflammatory stimuli with or without rosiglitazone, a peroxisome proliferator activated receptor γ (PPARγ) agonist, or T0901317, a liver X receptor (LXR) agonist. METHODS Real-time PCR and Western blotting for cholesterol transporters as well as cellular cholesterol efflux to high-density lipoprotein 2 (HDL(2)) were determined. RESULTS In RAW 264.7 macrophages, lipopolysaccharide (LPS) significantly reduced ABCG1 and PPARγ as well as cholesterol efflux to HDL(2). Rosiglitazone and T0901317 induced ABCA1 and ABCG1 several-fold, but LPS reduced only ABCG1. ABCG1 and SR-BI proteins, but not ABCA1, were decreased by LPS. In mouse peritoneal macrophages, LPS, tumor necrosis factor α and interleukin-1β decreased ABCG1, SR-BI, LXRα and PPARγ mRNA. The agonists increased ABC transporter expression but LPS reduced mRNA of T0901317-induced ABCA1 as well as basal and agonists-induced ABCG1. SR-BI protein was increased by rosiglitazone but LPS decreased the levels. CONCLUSION The data suggest that inflammatory insults repress ABCG1 and SR-BI expression partly dependent on PPARγ with a minimal effect on ABCA1 expression.
Collapse
Affiliation(s)
- Youngki Park
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| | | | | |
Collapse
|
48
|
Xie X, Wang S, Xiao L, Zhang J, Wang J, Liu J, Shen X, He D, Zheng X, Zhai Y. DBZ blocks LPS-induced monocyte activation and foam cell formation via inhibiting nuclear factor-ĸB. Cell Physiol Biochem 2011; 28:649-62. [PMID: 22178877 DOI: 10.1159/000335760] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS It has been widely accepted that chronic inflammation plays important roles in the atherogenesis. Danshensu Bingpian Zhi (DBZ) is a novel synthetic compound derived from the traditional Chinese medicine (TCM) formula Fu Fang Dan Shen (FFDS), which is effective on atherosclerosis clinically. We hypothesized that DBZ possessed the anti-atherosclerosis potentials. Here, we examined the inhibitory effects of DBZ on LPS-induced monocyte activation and foam cell formation. METHODS The effects of DBZ were assessed on LPS-induced inflammatory factors expression in monocyte/macrophage. Activation of NF-κB and AP-1 was analyzed by luciferase reporter assay and signaling pathway of NF-κB was investigated to elucidate mechanisms underlying DBZ mediated anti-inflammatory activity. Effects of DBZ on macrophage lipid accumulation were evaluated in native LDL and LPS co-incubated macrophages. RESULTS DBZ inhibited LPS-induced inflammatory factors expression dose dependently in monocytes. DBZ inhibited NF-κB activation strongly and AP-1 slightly. DBZ suppressed the LPS-induced degradation of IκBα, thereby decreasing the translocation of p65 to nucleus. Furthermore, DBZ suppressed LPS-activated macrophages lipid accumulation, partly due to inhibiting the expression of LPS-induced aP2 and ADRP in macrophges. CONCLUSION These results demonstrate that DBZ has potentials on anti-atherosclerosis by suppressing monocyte activation and foam cell formation.
Collapse
Affiliation(s)
- Xinni Xie
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry and College of Life Sciences, Beijing Normal University, Beijing, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kushiyama A, Okubo H, Sakoda H, Kikuchi T, Fujishiro M, Sato H, Kushiyama S, Iwashita M, Nishimura F, Fukushima T, Nakatsu Y, Kamata H, Kawazu S, Higashi Y, Kurihara H, Asano T. Xanthine oxidoreductase is involved in macrophage foam cell formation and atherosclerosis development. Arterioscler Thromb Vasc Biol 2011; 32:291-8. [PMID: 22095983 DOI: 10.1161/atvbaha.111.234559] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Hyperuricemia is common in patients with metabolic syndrome. We investigated the role of xanthine oxidoreductase (XOR) in atherosclerosis development, and the effects of the XOR inhibitor allopurinol on this process. METHODS AND RESULTS Oral administration of allopurinol to ApoE knockout mice markedly ameliorated lipid accumulation and calcification in the aorta and aortic root. In addition, allopurinol treatment or siRNA-mediated gene knockdown of XOR suppressed transformation of J774.1 murine macrophage cells, treated with acetylated LDL or very low density lipoprotein (VLDL) into foam cells. This inhibitory effect of allopurinol was also observed in primary cultured human macrophages. In contrast, overexpression of XOR promoted transformation of J774.1 cells into foam cells. Interestingly, SR-A1, SR-B1, SR-B II, and VLDL receptors in J774.1 cells were reduced by XOR knockdown, and increased by XOR overexpression. Conversely, expressions of ABCA1 and ABCG1 were increased by XOR knockdown and suppressed by XOR overexpression. Finally, productions of inflammatory cytokines accompanied by foam cell formation were also reduced by allopurinol administration. CONCLUSIONS These results strongly suggest XOR activity and/or its expression level to contribute to macrophage foam cell formation. Thus, XOR inhibitors may be useful for preventing atherosclerosis.
Collapse
|
50
|
Wang H, Peng DQ. New insights into the mechanism of low high-density lipoprotein cholesterol in obesity. Lipids Health Dis 2011; 10:176. [PMID: 21988829 PMCID: PMC3207906 DOI: 10.1186/1476-511x-10-176] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2011] [Accepted: 10/12/2011] [Indexed: 02/06/2023] Open
Abstract
Obesity, a significant risk factor for various chronic diseases, is universally related to dyslipidemia mainly represented by decreasing high-density lipoprotein cholesterol (HDL-C), which plays an indispensible role in development of cardiovascular disease (CVD). However, the mechanisms underlying obesity and low HDL-C have not been fully elucidated. Previous studies have focused on the alteration of HDL catabolism in circulation following elevated triglyceride (TG). But recent findings suggested that liver and fat tissue played pivotal role in obesity related low HDL-C. Some new molecular pathways like microRNA have also been proposed in the regulation of HDL metabolism in obesity. This article will review recent advances in understanding of the potential mechanism of low HDL-C in obesity.
Collapse
Affiliation(s)
- Hao Wang
- Departments of Cardiology, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, PR China
| | | |
Collapse
|