1
|
Mullis DM, Padilla-Lopez A, Wang H, Zhu Y, Elde S, Bonham SA, Yajima S, Kocher ON, Krieger M, Woo YJ. Stromal cell-derived factor-1 alpha improves cardiac function in a novel diet-induced coronary atherosclerosis model, the SR-B1ΔCT/LDLR KO mouse. Atherosclerosis 2024; 395:117518. [PMID: 38627162 PMCID: PMC11254567 DOI: 10.1016/j.atherosclerosis.2024.117518] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/28/2024] [Accepted: 03/12/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND AND AIMS There are a limited number of pharmacologic therapies for coronary artery disease, and few rodent models of occlusive coronary atherosclerosis and consequent myocardial infarction with which one can rapidly test new therapeutic approaches. Here, we characterize a novel, fertile, and easy-to-use HDL receptor (SR-B1)-based model of atherogenic diet-inducible, fatal coronary atherosclerosis, the SR-B1ΔCT/LDLR KO mouse. Additionally, we test intramyocardial injection of Stromal Cell-Derived Factor-1 alpha (SDF-1α), a potent angiogenic cytokine, as a possible therapy to rescue cardiac function in this mouse. METHODS SR-B1ΔCT/LDLR KO mice were fed the Paigen diet or standard chow diet, and we determined the effects of the diets on cardiac function, histology, and survival. After two weeks of feeding either the Paigen diet (n = 24) or standard chow diet (n = 20), the mice received an intramyocardial injection of either SDF-1α or phosphate buffered saline (PBS). Cardiac function and angiogenesis were assessed two weeks later. RESULTS When six-week-old mice were fed the Paigen diet, they began to die as early as 19 days later and 50% had died by 38 days. None of the mice maintained on the standard chow diet died by day 72. Hearts from mice on the Paigen diet showed evidence of cardiomegaly, myocardial infarction, and occlusive coronary artery disease. For the five mice that survived until day 28 that underwent an intramyocardial injection of PBS on day 15, the average ejection fraction (EF) decreased significantly from day 14 (the day before injection, 52.1 ± 4.3%) to day 28 (13 days after the injection, 30.6 ± 6.8%) (paired t-test, n = 5, p = 0.0008). Of the 11 mice fed the Paigen diet and injected with SDF-1α on day 15, 8 (72.7%) survived to day 28. The average EF for these 8 mice increased significantly from 48.2 ± 7.2% on day 14 to63.6 ± 6.9% on day 28 (Paired t-test, n = 8, p = 0.003). CONCLUSIONS This new mouse model and treatment with the promising angiogenic cytokine SDF-1α may lead to new therapeutic approaches for ischemic heart disease.
Collapse
MESH Headings
- Animals
- Chemokine CXCL12/metabolism
- Chemokine CXCL12/genetics
- Disease Models, Animal
- Mice, Knockout
- Coronary Artery Disease
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Scavenger Receptors, Class B/genetics
- Male
- Neovascularization, Physiologic/drug effects
- Mice, Inbred C57BL
- Diet, Atherogenic
- Mice
- Ventricular Function, Left
- Myocardium/pathology
- Myocardium/metabolism
- Diet, High-Fat
Collapse
Affiliation(s)
- Danielle M Mullis
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | | | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Yuanjia Zhu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Stefan Elde
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Spencer A Bonham
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Shin Yajima
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Olivier N Kocher
- Department of Pathology, Beth Israel Hospital, Harvard Medical School, Boston, MA, USA
| | - Monty Krieger
- Department of Biology, Massachusetts Institute of Technology, MA, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
2
|
Zhang L, Shi Y, Liang B, Li X. An overview of the cholesterol metabolism and its proinflammatory role in the development of MASLD. Hepatol Commun 2024; 8:e0434. [PMID: 38696365 PMCID: PMC11068152 DOI: 10.1097/hc9.0000000000000434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/05/2024] [Indexed: 05/04/2024] Open
Abstract
Cholesterol is an essential lipid molecule in mammalian cells. It is not only involved in the formation of cell membranes but also serves as a raw material for the synthesis of bile acids, vitamin D, and steroid hormones. Additionally, it acts as a covalent modifier of proteins and plays a crucial role in numerous life processes. Generally, the metabolic processes of cholesterol absorption, synthesis, conversion, and efflux are strictly regulated. Excessive accumulation of cholesterol in the body is a risk factor for metabolic diseases such as cardiovascular disease, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD). In this review, we first provide an overview of the discovery of cholesterol and the fundamental process of cholesterol metabolism. We then summarize the relationship between dietary cholesterol intake and the risk of developing MASLD, and also the animal models of MASLD specifically established with a cholesterol-containing diet. In the end, the role of cholesterol-induced inflammation in the initiation and development of MASLD is discussed.
Collapse
Affiliation(s)
- Linqiang Zhang
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Yongqiong Shi
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Bin Liang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Xi Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Li G, Park HJ, Suh JH, Choi HS. 7-Ketocholesterol plays a key role in cholesterol-induced hepatitis via macrophage and neutrophil infiltration. J Nutr Biochem 2024; 125:109552. [PMID: 38134972 DOI: 10.1016/j.jnutbio.2023.109552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
This study sought to explore the role of 7-ketocholesterol (7-KC) in liver damage caused by high cholesterol intake and its potential pathological mechanism in mice. Our in vivo findings indicated that mice fed a high-cholesterol diet had elevated serum levels of 7-KC, accompanied by liver injury and inflammation, similar to human nonalcoholic steatohepatitis. Furthermore, the high-cholesterol diet induced neutrophil infiltration, which played a critical role in liver damage through myeloperoxidase (MPO) activity. Upon stimulation with 7-KC, macrophages exhibited increased expression of C-X-C motif chemokine ligand 1 (CXCL1) and CXCL2, as well as ATP-binding cassette transporter A1 (ABCA1) and ABCG1. Hepatocytes, on the other hand, exhibited increased expression of CXCL2 and ABCG1. The infiltration of neutrophils in the liver was primarily caused by CXCL1 and CXCL2, resulting in hepatocyte cell death due to elevated MPO activity. Our data also revealed that the activation of macrophages by 7-KC via ABCA1 or ABCG1 was not associated with lipid accumulation. Collectively, these findings suggest that high cholesterol-induced hepatitis in mice involves, at least partially, the recruitment of neutrophils to the liver by 7-KC-activated macrophages. This is mediated by increased expression of CXCL1 and CXCL2 through ABCA1 or ABCG1, which act as 7-KC efflux transporters. Additionally, hepatocytes contribute to this process by increased expression of CXCL2 through ABCG1. Therefore, our findings suggest that 7-KC may play a role in high cholesterol-induced hepatitis in mice by activating macrophages and hepatocytes, ultimately leading to neutrophil infiltration.
Collapse
Affiliation(s)
- Guoen Li
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Hyun-Jung Park
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Jae-Hee Suh
- Department of Pathology, Ulsan University Hospital, Ulsan, Korea
| | - Hye-Seon Choi
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea.
| |
Collapse
|
4
|
Arunachalam AR, Samuel SS, Mani A, Maynard JP, Stayer KM, Dybbro E, Narayanan S, Biswas A, Pathan S, Soni K, Kamal AHM, Ambati CSR, Putluri N, Desai MS, Thevananther S. P2Y2 purinergic receptor gene deletion protects mice from bacterial endotoxin and sepsis-associated liver injury and mortality. Am J Physiol Gastrointest Liver Physiol 2023; 325:G471-G491. [PMID: 37697947 PMCID: PMC10812707 DOI: 10.1152/ajpgi.00090.2023] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/28/2023] [Accepted: 09/11/2023] [Indexed: 09/13/2023]
Abstract
The liver plays a significant role in regulating a wide range of metabolic, homeostatic, and host-defense functions. However, the impact of liver injury on the host's ability to control bacteremia and morbidity in sepsis is not well understood. Leukocyte recruitment and activation lead to cytokine and chemokine release, which, in turn, trigger hepatocellular injury and elevate nucleotide levels in the extracellular milieu. P2Y2 purinergic receptors, G protein-coupled and activated by extracellular ATP/UTP, are expressed at the cell surface of hepatocytes and nonparenchymal cells. We sought to determine whether P2Y2 purinergic receptor function is necessary for the maladaptive host response to bacterial infection and endotoxin-mediated inflammatory liver injury and mortality in mice. We report that P2Y2 purinergic receptor knockout mice (P2Y2-/-) had attenuated inflammation and liver injury, with improved survival in response to LPS/galactosamine (LPS/GalN; inflammatory liver injury) and cecal ligation and puncture (CLP; polymicrobial sepsis). P2Y2-/- livers had attenuated c-Jun NH2-terminal kinase activation, matrix metallopeptidase-9 expression, and hepatocyte apoptosis in response to LPS/GalN and attenuated inducible nitric oxide synthase and nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain containing 3 protein expression in response to CLP. Implicating liver injury in the disruption of amino acid homeostasis, CLP led to lower serum arginine and higher bacterial load and morbidity in the WT mice, whereas serum arginine levels were comparable to sham-operated controls in P2Y2-/- mice, which had attenuated bacteremia and improved survival. Collectively, our studies highlight the pathophysiological relevance of P2Y2 purinergic receptor function in inflammatory liver injury and dysregulation of systemic amino acid homeostasis with implications for sepsis-associated immune dysfunction and morbidity in mice.NEW & NOTEWORTHY Our studies provide experimental evidence for P2Y2 purinergic receptor-mediated potentiation of inflammatory liver injury, morbidity, and mortality, in two well-established animal models of inflammatory liver injury. Our findings highlight the potential to target P2Y2 purinergic signaling to attenuate the induction of "cytokine storm" and prevent its deleterious consequences on liver function, systemic amino acid homeostasis, host response to bacterial infection, and sepsis-associated morbidity and mortality.
Collapse
Affiliation(s)
- Athis R Arunachalam
- Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - Sanju S Samuel
- Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - Arunmani Mani
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - Janielle P Maynard
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - Kelsey M Stayer
- Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - Eric Dybbro
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - Subapradha Narayanan
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - Aalekhya Biswas
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - Saliha Pathan
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - Krishnakant Soni
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - Abu Hena Mostafa Kamal
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States
| | | | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States
| | - Moreshwar S Desai
- Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - Sundararajah Thevananther
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
5
|
Expression and Function of BMP and Activin Membrane-Bound Inhibitor (BAMBI) in Chronic Liver Diseases and Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:ijms24043473. [PMID: 36834884 PMCID: PMC9964332 DOI: 10.3390/ijms24043473] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
BAMBI (bone morphogenetic protein and activin membrane-bound inhibitor) is a transmembrane pseudoreceptor structurally related to transforming growth factor (TGF)-β type 1 receptors (TGF-β1Rs). BAMBI lacks a kinase domain and functions as a TGF-β1R antagonist. Essential processes such as cell differentiation and proliferation are regulated by TGF-β1R signaling. TGF-β is the best-studied ligand of TGF-βRs and has an eminent role in inflammation and fibrogenesis. Liver fibrosis is the end stage of almost all chronic liver diseases, such as non-alcoholic fatty liver disease, and at the moment, there is no effective anti-fibrotic therapy available. Hepatic BAMBI is downregulated in rodent models of liver injury and in the fibrotic liver of patients, suggesting that low BAMBI has a role in liver fibrosis. Experimental evidence convincingly demonstrated that BAMBI overexpression is able to protect against liver fibrosis. Chronic liver diseases have a high risk of hepatocellular carcinoma (HCC), and BAMBI was shown to exert tumor-promoting as well as tumor-protective functions. This review article aims to summarize relevant studies on hepatic BAMBI expression and its role in chronic liver diseases and HCC.
Collapse
|
6
|
Horn CL, Morales AL, Savard C, Farrell GC, Ioannou GN. Role of Cholesterol-Associated Steatohepatitis in the Development of NASH. Hepatol Commun 2021; 6:12-35. [PMID: 34558856 PMCID: PMC8710790 DOI: 10.1002/hep4.1801] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 07/08/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
The rising prevalence of nonalcoholic fatty liver disease (NAFLD) and NAFLD-related cirrhosis in the United States and globally highlights the need to better understand the mechanisms causing progression of hepatic steatosis to fibrosing steatohepatitis and cirrhosis in a small proportion of patients with NAFLD. Accumulating evidence suggests that lipotoxicity mediated by hepatic free cholesterol (FC) overload is a mechanistic driver for necroinflammation and fibrosis, characteristic of nonalcoholic steatohepatitis (NASH), in many animal models and also in some patients with NASH. Diet, lifestyle, obesity, key genetic polymorphisms, and hyperinsulinemia secondary to insulin resistance are pivotal drivers leading to aberrant cholesterol signaling, which leads to accumulation of FC within hepatocytes. FC overload in hepatocytes can lead to ER stress, mitochondrial dysfunction, development of toxic oxysterols, and cholesterol crystallization in lipid droplets, which in turn lead to hepatocyte apoptosis, necrosis, or pyroptosis. Activation of Kupffer cells and hepatic stellate cells by hepatocyte signaling and cholesterol loading contributes to this inflammation and leads to hepatic fibrosis. Cholesterol accumulation in hepatocytes can be readily prevented or reversed by statins. Observational studies suggest that use of statins in NASH not only decreases the substantially increased cardiovascular risk, but may ameliorate liver pathology. Conclusion: Hepatic FC loading may result in cholesterol-associated steatohepatitis and play an important role in the development and progression of NASH. Statins appear to provide significant benefit in preventing progression to NASH and NASH-cirrhosis. Randomized controlled trials are needed to demonstrate whether statins or statin/ezetimibe combination can effectively reverse steatohepatitis and liver fibrosis in patients with NASH.
Collapse
Affiliation(s)
- Christian L Horn
- Division of Gastroenterology and Hepatology, Department of Medicine, San Antonio Military Medical Center, Fort Sam Houston, TX, USA
| | - Amilcar L Morales
- Division of Gastroenterology and Hepatology, Department of Medicine, San Antonio Military Medical Center, Fort Sam Houston, TX, USA
| | - Christopher Savard
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, WA, USA.,Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Geoffrey C Farrell
- Liver Research Group, ANU Medical School, Australian National University at the Canberra Hospital, Garran, ACT, Australia
| | - George N Ioannou
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, WA, USA.,Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| |
Collapse
|
7
|
Kitamura K, Isoda K, Akita K, Miyosawa K, Kadoguchi T, Shimada K, Daida H. Lack of IκBNS promotes cholate-containing high-fat diet-induced inflammation and atherogenesis in low-density lipoprotein (LDL) receptor-deficient mice. IJC HEART & VASCULATURE 2019; 23:100344. [PMID: 30976650 PMCID: PMC6439265 DOI: 10.1016/j.ijcha.2019.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/12/2019] [Indexed: 11/21/2022]
Abstract
Background IκBNS, a nuclear IκB protein, regulates a subset of Toll-like receptor (TLR) dependent genes. A cholate-containing high-fat diet (HFD(CA(+))) induces TLR4 mediated early inflammatory response. The present study aims to clarify that the lack of IκBNS promotes atherogenesis in low-density lipoprotein receptor-deficient (LDLr−/−) mice fed HFD(CA(+)) compared with those fed a cholate-free HFD (HFD(CA(−))). Methods and results Mice that lacked IκBNS (IκBNS−/−) were crossed with LDLr−/− mice and formation of atherosclerotic lesions was analyzed after 6-week consumption of HFD(CA(+)) or HFD(CA(−)). IκBNS−/−/LDLr−/− mice fed HFD(CA(+)) (IκBNS−/−/LDLr−/−(CA(+))) showed a 3.5-fold increase of atherosclerotic lesion size in the aorta compared with LDLr−/−(CA(+)) mice (p < 0.01), whereas there was no difference between LDLr−/−(CA(−)) and IκBNS−/−/LDLr−/−(CA(−)) mice. Immunohistochemical analysis of the aortic root revealed that HFD(CA(+)) significantly increased Mac-3 (macrophage)-positive area by 1.5-fold (p < 0.01) and TLR4, interleukin-6 (IL-6) expression by 1.7-fold (p < 0.05) and 1.5-fold (p < 0.05), respectively, in IκBNS−/−/LDLr−/−(CA(+)) compared with LDLr-/-−−(CA(+)) mice. Furthermore, active STAT3 (pSTAT3)-positive cells were significantly increased by 1.7-fold in the lesions of IκBNS−/−/LDLr−/−(CA(+)) compared with LDLr−/−(CA(+)) mice (p < 0.01). These findings suggest that IκBNS deficiency and HFD(CA(+)) promote atherogenesis in LDLr−/− mice via TLR4/IL-6/STAT3 pathway. Finally, we showed that the monocytes from peripheral blood of IκBNS−/−/LDLr−/−(CA(+)) mice were found to contain the highest proportion of Ly6Chi monocytes among the four groups, suggesting that lack of IκBNS enhanced inflammation in response to HFD(CA(+)) feeding. Conclusions The present study is the first to demonstrate that the activation of innate immune system using HFD(CA(+)) induced significant inflammation and atherogenesis in IκBNS−/−/LDLr−/− compared with LDLr−/− mice.
Collapse
Affiliation(s)
| | - Kikuo Isoda
- Corresponding author at: Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | | | | | | | | | | |
Collapse
|
8
|
Yao HT, Lee PF, Lii CK, Liu YT, Chen SH. Freshwater clam extract reduces liver injury by lowering cholesterol accumulation, improving dysregulated cholesterol synthesis and alleviating inflammation in high-fat, high-cholesterol and cholic acid diet-induced steatohepatitis in mice. Food Funct 2019; 9:4876-4887. [PMID: 30160281 DOI: 10.1039/c8fo00851e] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Freshwater clam (Corbicula fluminea) is a traditional liver-protective food in Asia. Recent studies have renewed attention on high cholesterol accumulation and dysregulated cholesterol synthesis in the liver as a critical factor in the progression of nonalcoholic fatty liver disease (NAFLD) to nonalcoholic steatohepatitis (NASH). In this study, we investigated the protective effects of freshwater clam extract (FCE) and its fat fraction (FCE oil) on high-fat, high-cholesterol and cholic acid (HFHC) diet-induced lean steatohepatitis in mice. Mice were fed a HFHC diet containing FCE or FCE oil for 6 weeks. FCE, but not FCE oil, feeding reduced liver injury as indicated by decreased plasma alanine aminotransferase activity. Liver total cholesterol accumulation was reduced after FCE and FCE oil treatment. Accumulation of squalene and desmosterol, the precursors of cholesterol, in the liver was reduced by FCE but not by FCE oil. The caspase-1 (p10) and interleukin (IL)-1β (p17) protein expressions in the liver were suppressed by both FCE and FCE oil. Therefore, FCE may act as functional food that can reduce steatohepatitis and liver injury by reducing cholesterol accumulation, improving dysregulated cholesterol synthesis and attenuating inflammation.
Collapse
Affiliation(s)
- Hsien-Tsung Yao
- Department of Nutrition, China Medical University, 91 Hsueh-shih Road, Taichung 404, Taiwan.
| | | | | | | | | |
Collapse
|
9
|
Chemokines and Chemokine Receptors in the Development of NAFLD. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1061:45-53. [PMID: 29956205 DOI: 10.1007/978-981-10-8684-7_4] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chemokines are chemo-attractants for leukocyte trafficking, growth, and activation in injured and inflammatory tissues. The chemokine system is comprised of 50 chemokine ligands and 20 cognate chemokine receptors. In the context of liver diseases, leukocytes, hepatocytes, hepatic stellate cells, endothelial cells, and vascular smooth muscle cells are capable of producing chemokines. Chemokine receptors are typically expressed in various leukocyte subsets. Given that inflammation is a critical factor for the transition from simple steatosis to non-alcoholic steatohepatitis (NASH), and fibrosis, the chemokine system may play a prominent role in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Indeed, accumulating evidence shows elevated expression of chemokines and their receptors in the livers of obese patients with advanced steatosis and NASH. This chapter will discuss the underlying molecular mechanisms and the therapeutic potential of the chemokine systems in the pathogenesis of NAFLD. Among chemokines, we will highlight CCL2, CCL5, CXCL8-10, CX3CL1, and CXCL16 as pivotal mediators in the development of steatosis, NASH, and fibrosis.
Collapse
|
10
|
Eblimit Z, Thevananther S, Karpen SJ, Taegtmeyer H, Moore DD, Adorini L, Penny DJ, Desai MS. TGR5 activation induces cytoprotective changes in the heart and improves myocardial adaptability to physiologic, inotropic, and pressure-induced stress in mice. Cardiovasc Ther 2018; 36:e12462. [PMID: 30070769 DOI: 10.1111/1755-5922.12462] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/12/2018] [Accepted: 07/30/2018] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Administration of cholic acid, or its synthetic derivative, 6-alpha-ethyl-23(S)-methylcholic acid (INT-777), activates the membrane GPCR, TGR5, influences whole body metabolism, reduces atherosclerosis, and benefits the cardiovascular physiology in mice. Direct effects of TGR5 agonists, and the role for TGR5, on myocardial cell biology and stress response are unknown. METHODS Mice were fed chow supplemented with 0.5% cholic acid (CA) or 0.025% INT-777, a specific TGR5 agonist, or regular chow for 3 weeks. Anthropometric, biochemical, physiologic (electrocardiography and echocardiography), and molecular analysis was performed at baseline. CA and INT-777 fed mice were challenged with acute exercise-induced stress, acute catecholamine-induced stress, and hemodynamic stress induced by transverse aortic constriction (TAC) for a period of 8 weeks. In separate experiments, mice born with constitutive deletion of TGR5 in cardiomyocytes (CM-TGR5del ) were exposed to exercise, inotropic, and TAC-induced stress. RESULTS Administration of CA and INT-777 supplemented diets upregulated TGR5 expression and activated Akt, PKA, and ERK1/2 in the heart. CA and INT-777 fed mice showed improved exercise tolerance, improved sensitivity to catecholamine and attenuation in pathologic remodeling of the heart under hemodynamic stress. In contrast, CM-TGR5del showed poor response to exercise and catecholamine challenge as well as higher mortality and signs of accelerated cardiomyopathy under hemodynamic stress. CONCLUSIONS Bile acids, specifically TGR5 agonists, induce cytoprotective changes in the heart and improve myocardial response to physiologic, inotropic, and hemodynamic stress in mice. TGR5 plays a critical role in myocardial adaptability, and TGR5 activation may represent a potentially attractive treatment option in heart failure.
Collapse
Affiliation(s)
- Zeena Eblimit
- Section of Pediatric Critical Care, Baylor College of Medicine, Houston, Texas
| | | | - Saul J Karpen
- Pediatric Gastroenterology, Emory School of Medicine, Atlanta, Georgia
| | - Heinrich Taegtmeyer
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas
| | - David D Moore
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | | | - Daniel J Penny
- Department of Pediatric Cardiology, Baylor College of Medicine, Houston, Texas
| | - Moreshwar S Desai
- Section of Pediatric Critical Care, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
11
|
Understanding the Impact of Dietary Cholesterol on Chronic Metabolic Diseases through Studies in Rodent Models. Nutrients 2018; 10:nu10070939. [PMID: 30037080 PMCID: PMC6073247 DOI: 10.3390/nu10070939] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 01/07/2023] Open
Abstract
The development of certain chronic metabolic diseases has been attributed to elevated levels of dietary cholesterol. However, decades of research in animal models and humans have demonstrated a high complexity with respect to the impact of dietary cholesterol on the progression of these diseases. Thus, recent investigations in non-alcoholic fatty liver disease (NAFLD) point to dietary cholesterol as a key factor for the activation of inflammatory pathways underlying the transition from NAFLD to non-alcoholic steatohepatitis (NASH) and to hepatic carcinoma. Dietary cholesterol was initially thought to be the key factor for cardiovascular disease development, but its impact on the disease depends partly on the capacity to modulate plasmatic circulating low-density lipoprotein (LDL) cholesterol levels. These studies evidence a complex relationship between these chronic metabolic diseases and dietary cholesterol, which, in certain conditions, might promote metabolic complications. In this review, we summarize rodent studies that evaluate the impact of dietary cholesterol on these two prevalent chronic diseases and their relevance to human pathology.
Collapse
|
12
|
Zimny S, Pohl R, Rein-Fischboeck L, Haberl EM, Krautbauer S, Weiss TS, Buechler C. Chemokine (CC-motif) receptor-like 2 mRNA is expressed in hepatic stellate cells and is positively associated with characteristics of non-alcoholic steatohepatitis in mice and men. Exp Mol Pathol 2017; 103:1-8. [DOI: 10.1016/j.yexmp.2017.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 05/29/2017] [Accepted: 06/05/2017] [Indexed: 02/07/2023]
|
13
|
Tu LN, Showalter MR, Cajka T, Fan S, Pillai VV, Fiehn O, Selvaraj V. Metabolomic characteristics of cholesterol-induced non-obese nonalcoholic fatty liver disease in mice. Sci Rep 2017; 7:6120. [PMID: 28733574 PMCID: PMC5522413 DOI: 10.1038/s41598-017-05040-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 05/30/2017] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) in non-obese patients remains a clinical condition with unclear etiology and pathogenesis. Using a metabolomics approach in a mouse model that recapitulates almost all the characteristic features of non-obese NAFLD, we aimed to advance mechanistic understanding of this disorder. Mice fed high fat, high cholesterol, cholate (HFHCC) diet for three weeks consistently developed hepatic pathology similar to NAFLD and nonalcoholic steatohepatitis (NASH) without changes to body weight or fat pad weights. Gas- and liquid chromatography/mass spectrometry-based profiling of lipidomic and primary metabolism changes in the liver and plasma revealed that systemic mechanisms leading to steatosis and hepatitis in this non-obese NAFLD model were driven by a combination of effects directed by elevated free cholesterol, cholesterol esters and cholic acid, and associated changes to metabolism of sphingomyelins and phosphatidylcholines. These results demonstrate that mechanisms underlying cholesterol-induced non-obese NAFLD are distinct from NAFLD occurring as a consequence of metabolic syndrome. In addition, this investigation provides one of the first metabolite reference profiles for interpreting effects of dietary and hepatic cholesterol in human non-obese NAFLD/NASH patients.
Collapse
Affiliation(s)
- Lan N Tu
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Megan R Showalter
- West Coast Metabolomics Center, University of California Davis Genome Center, Davis, CA, 95616, USA
| | - Tomas Cajka
- West Coast Metabolomics Center, University of California Davis Genome Center, Davis, CA, 95616, USA
| | - Sili Fan
- West Coast Metabolomics Center, University of California Davis Genome Center, Davis, CA, 95616, USA
| | - Viju V Pillai
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California Davis Genome Center, Davis, CA, 95616, USA
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Vimal Selvaraj
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
14
|
Bloomer SA, Olivier AK, Bergmann OM, Mathahs MM, Broadhurst KA, Hicsasmaz H, Brown KE. Strain- and time-dependent alterations in hepatic iron metabolism in a murine model of nonalcoholic steatohepatitis. Cell Biochem Funct 2017; 34:628-639. [PMID: 27935134 DOI: 10.1002/cbf.3238] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 01/29/2023]
Abstract
Nonalcoholic steatohepatitis is a common liver disease that is often accompanied by dysregulated iron metabolism. The aim of the study was to test the hypothesis that aberrant iron metabolism in nonalcoholic steatohepatitis is modulated by genetic susceptibility to inflammation and oxidative stress. Hepatic histology and iron content were assessed in 3 inbred strains of mice (C57BL/6, BALB/c, and C3H/HeJ) fed an atherogenic diet (AD). Hepatic expression of genes relevant to iron metabolism, inflammation, and oxidative stress were quantitated by real-time reverse transcription-polymerase chain reaction. At 6 weeks on the AD, histologic injury and induction of inflammatory and oxidative stress-associated gene expression were most pronounced in C57BL/6. At 18 weeks on the AD, these parameters were similar in C57BL/6 and BALB/c. Atherogenic diet-fed C3H/HeJ showed milder responses at both time points. The AD was associated with decreased hepatic iron concentrations in all strains at 6 and 18 weeks. The decrease in hepatic iron concentrations did not correlate with changes in hepcidin expression and was not associated with altered expression of iron transporters. These findings are similar to those observed in models of obesity-induced steatosis and indicate that hepatic steatosis can be associated with depletion of iron stores that is not explained by upregulation of hepcidin expression by inflammation. SIGNIFICANCE OF THE STUDY Nonalcoholic steatohepatitis (NASH) is a common liver disease that often accompanies the metabolic syndrome. The latter condition has been linked to iron deficiency and diminished intestinal iron absorption, likely the result of hepcidin upregulation by chronic inflammation. Paradoxically, some NASH patients accumulate excess hepatic iron, which may increase fibrosis and cancer risk. Iron accumulation has been attributed to suppression of hepcidin by oxidative stress. The objective of this study was to investigate the contributions of inflammation and oxidative stress to altered hepatic iron metabolism in a murine model of NASH using inbred strains of mice with differing susceptibilities to injury.
Collapse
Affiliation(s)
- Steven A Bloomer
- Division of Science and Engineering, Penn State Abington, Abington, PA, USA
| | - Alicia K Olivier
- Division of Comparative Pathology, Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Pathobiology and Population Medicine, Mississippi State University, Starkville, MS, USA
| | - Ottar M Bergmann
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Internal Medicine, Section of Gastroenterology and Hepatology, The National University Hospital of Iceland, Reykjavik, Iceland
| | - M Meleah Mathahs
- Iowa City Veterans Administration Medical Center, Iowa City, IA, USA
| | | | | | - Kyle E Brown
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Iowa City Veterans Administration Medical Center, Iowa City, IA, USA.,Program in Free Radical and Radiation Biology, Department of Radiation Oncology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
15
|
Wang Y, Song E, Bai B, Vanhoutte PM. Toll-like receptors mediating vascular malfunction: Lessons from receptor subtypes. Pharmacol Ther 2015; 158:91-100. [PMID: 26702901 DOI: 10.1016/j.pharmthera.2015.12.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Toll-like receptors (TLR) are a subfamily of pattern recognition receptors (PRR) implicated in a variety of vascular abnormalities. However, the pathophysiological role and the interplay between different TLR-mediated innate and adaptive immune responses during the development of vascular diseases remain largely unspecified. TLR are widely distributed in both immune and nonimmune cells in the blood vessel wall. The expressions and locations of TLR are dynamically regulated in response to distinct molecular patterns derived from pathogens or damaged host cells. As a result, the outcome of TLR signaling is agonist- and cell type-dependent. A better understanding of discrete TLR signaling pathways in the vasculature will provide unprecedented opportunities for the discovery of novel therapies in many inflammatory vascular diseases. The present brief review discusses the role of individual TLR in controlling cellular functions of the vascular system, by focusing on the inflammatory responses within the blood vessel wall which contribute to the development of hypertension and atherosclerosis.
Collapse
Affiliation(s)
- Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| | - Erfei Song
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Bo Bai
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Paul M Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
16
|
Nozako M, Koyama T, Nagano C, Sato M, Matsumoto S, Mitani K, Yasufuku R, Kohashi M, Yoshikawa T. An Atherogenic Paigen-Diet Aggravates Nephropathy in Type 2 Diabetic OLETF Rats. PLoS One 2015; 10:e0143979. [PMID: 26606054 PMCID: PMC4659596 DOI: 10.1371/journal.pone.0143979] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/11/2015] [Indexed: 11/18/2022] Open
Abstract
Diabetic nephropathy develops in association with hyperglycemia, is aggravated by atherogenic factors such as dyslipidemia, and is sometimes initiated before obvious hyperglycemia is seen. However, the precise mechanisms of progression are still unclear. In this study, we investigated the influence of an atherogenic Paigen diet (PD) on the progression of nephropathy in spontaneous type 2 diabetic OLETF rats. Feeding PD to male OLETF rats for 12 weeks caused an extensive increase in excretion of urinary albumin and markers of tubular injury such as KIM-1 and L-FABP, accompanied by mesangial expansion and tubular atrophy. PD significantly increased plasma total cholesterol concentration, which correlates well with increases in urine albumin excretion and mesangial expansion. Conversely, PD did not change plasma glucose and free fatty acid concentrations. PD enhanced renal levels of mRNA for inflammatory molecules such as KIM-1, MCP-1, TLR4 and TNF-α and promoted macrophage infiltration and lipid accumulation in the tubulointerstitium and glomeruli in OLETF rats. Intriguingly, PD had little effect on urine albumin excretion and renal morphology in normal control LETO rats. This model may be useful in studying the complex mechanisms that aggravate diabetic nephropathy in an atherogenic environment.
Collapse
Affiliation(s)
- Masanori Nozako
- Free Radical Research Project, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
- Department of Toxicology, Drug Safety Research Center, Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
| | - Takashi Koyama
- Free Radical Research Project, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
- Department of Toxicology, Drug Safety Research Center, Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
| | - Chifumi Nagano
- Free Radical Research Project, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
- Department of Toxicology, Drug Safety Research Center, Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
| | - Makoto Sato
- Department of Toxicology, Drug Safety Research Center, Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
| | - Satoshi Matsumoto
- Department of Toxicology, Drug Safety Research Center, Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
| | - Kiminobu Mitani
- Free Radical Research Project, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
| | - Reiko Yasufuku
- Free Radical Research Project, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
| | - Masayuki Kohashi
- Free Radical Research Project, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
| | - Tomohiro Yoshikawa
- Free Radical Research Project, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
- * E-mail:
| |
Collapse
|
17
|
Woollard SM, Kanmogne GD. Maraviroc: a review of its use in HIV infection and beyond. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5447-68. [PMID: 26491256 PMCID: PMC4598208 DOI: 10.2147/dddt.s90580] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The human immunodeficiency virus-1 (HIV-1) enters target cells by binding its envelope glycoprotein gp120 to the CD4 receptor and/or coreceptors such as C-C chemokine receptor type 5 (CCR5; R5) and C-X-C chemokine receptor type 4 (CXCR4; X4), and R5-tropic viruses predominate during the early stages of infection. CCR5 antagonists bind to CCR5 to prevent viral entry. Maraviroc (MVC) is the only CCR5 antagonist currently approved by the United States Food and Drug Administration, the European Commission, Health Canada, and several other countries for the treatment of patients infected with R5-tropic HIV-1. MVC has been shown to be effective at inhibiting HIV-1 entry into cells and is well tolerated. With expanding MVC use by HIV-1-infected humans, different clinical outcomes post-approval have been observed with MVC monotherapy or combination therapy with other antiretroviral drugs, with MVC use in humans infected with dual-R5- and X4-tropic HIV-1, infected with different HIV-1 genotype or infected with HIV-2. This review discuss the role of CCR5 in HIV-1 infection, the development of the CCR5 antagonist MVC, its pharmacokinetics, pharmacodynamics, drug–drug interactions, and the implications of these interactions on treatment outcomes, including viral mutations and drug resistance, and the mechanisms associated with the development of resistance to MVC. This review also discusses available studies investigating the use of MVC in the treatment of other diseases such as cancer, graft-versus-host disease, and inflammatory diseases.
Collapse
Affiliation(s)
- Shawna M Woollard
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Georgette D Kanmogne
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
18
|
Goodwin JE, Zhang X, Rotllan N, Feng Y, Zhou H, Fernández-Hernando C, Yu J, Sessa WC. Endothelial glucocorticoid receptor suppresses atherogenesis--brief report. Arterioscler Thromb Vasc Biol 2015; 35:779-782. [PMID: 25810297 DOI: 10.1161/atvbaha.114.304525] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The purpose of this study was to determine the role of the endothelial glucocorticoid receptor in the pathogenesis of atherosclerosis. APPROACH AND RESULTS Control mice and mice lacking the endothelial glucocorticoid receptor were bred onto an Apoe knockout background and subjected to high-fat diet feeding for 12 weeks. Assessment of body weight and total cholesterol and triglycerides before and after the diet revealed no differences between the 2 groups of mice. However, mice lacking the endothelial glucocorticoid receptor developed more severe atherosclerotic lesions in the aorta, brachiocephalic artery, and aortic sinus, as well as a heightened inflammatory milieu as evidenced by increased macrophage recruitment in the lesions. CONCLUSIONS These data suggest that the endothelial glucocorticoid receptor is important for tonic inhibition of inflammation and limitation of atherosclerosis progression in this model.
Collapse
Affiliation(s)
- Julie E Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Xinbo Zhang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT.,Department of Internal Medicine (Cardiology), Yale University School of Medicine, New Haven, CT
| | - Noemi Rotllan
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT
| | - Yan Feng
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Han Zhou
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Carlos Fernández-Hernando
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT
| | - Jun Yu
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT.,Department of Internal Medicine (Cardiology), Yale University School of Medicine, New Haven, CT
| | - William C Sessa
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT.,Department of Pharmacology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
19
|
Schuck RN, Zha W, Edin ML, Gruzdev A, Vendrov KC, Miller TM, Xu Z, Lih FB, DeGraff LM, Tomer KB, Jones HM, Makowski L, Huang L, Poloyac SM, Zeldin DC, Lee CR. The cytochrome P450 epoxygenase pathway regulates the hepatic inflammatory response in fatty liver disease. PLoS One 2014; 9:e110162. [PMID: 25310404 PMCID: PMC4195706 DOI: 10.1371/journal.pone.0110162] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/08/2014] [Indexed: 12/15/2022] Open
Abstract
Fatty liver disease is an emerging public health problem without effective therapies, and chronic hepatic inflammation is a key pathologic mediator in its progression. Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to biologically active epoxyeicosatrienoic acids (EETs), which have potent anti-inflammatory effects. Although promoting the effects of EETs elicits anti-inflammatory and protective effects in the cardiovascular system, the contribution of CYP-derived EETs to the regulation of fatty liver disease-associated inflammation and injury is unknown. Using the atherogenic diet model of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis (NAFLD/NASH), our studies demonstrated that induction of fatty liver disease significantly and preferentially suppresses hepatic CYP epoxygenase expression and activity, and both hepatic and circulating levels of EETs in mice. Furthermore, mice with targeted disruption of Ephx2 (the gene encoding soluble epoxide hydrolase) exhibited restored hepatic and circulating EET levels and a significantly attenuated induction of hepatic inflammation and injury. Collectively, these data suggest that suppression of hepatic CYP-mediated EET biosynthesis is an important pathological consequence of fatty liver disease-associated inflammation, and that the CYP epoxygenase pathway is a central regulator of the hepatic inflammatory response in NAFLD/NASH. Future studies investigating the utility of therapeutic strategies that promote the effects of CYP-derived EETs in NAFLD/NASH are warranted.
Collapse
Affiliation(s)
- Robert N. Schuck
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Weibin Zha
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Matthew L. Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Artiom Gruzdev
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Kimberly C. Vendrov
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Tricia M. Miller
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Zhenghong Xu
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Fred B. Lih
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Laura M. DeGraff
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Kenneth B. Tomer
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - H. Michael Jones
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Liza Makowski
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Leaf Huang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Samuel M. Poloyac
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Darryl C. Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Craig R. Lee
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
20
|
Pérez-Martínez L, Pérez-Matute P, Aguilera-Lizarraga J, Rubio-Mediavilla S, Narro J, Recio E, Ochoa-Callejero L, Oteo JA, Blanco JR. Maraviroc, a CCR5 antagonist, ameliorates the development of hepatic steatosis in a mouse model of non-alcoholic fatty liver disease (NAFLD). J Antimicrob Chemother 2014; 69:1903-10. [PMID: 24651825 DOI: 10.1093/jac/dku071] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in the general population. The NAFLD spectrum ranges from simple steatosis to cirrhosis. The chemokine CCL5/RANTES plays an important role in the progression of hepatic inflammation and fibrosis. The objective of this study was to examine the effects of maraviroc, a CCR5 antagonist, on liver pathology in a NAFLD mouse model. METHODS A total of 32 male C57BL/6 mice were randomly assigned to one of four groups: (i) control group (chow diet plus tap water); (ii) maraviroc group (chow diet plus maraviroc in drinking water); (iii) high-fat diet (HFD) group (HFD plus tap water); and (iv) maraviroc/HFD group (HFD plus maraviroc). All mice were sacrificed 16 weeks after the beginning of the experiment. Biochemical analyses and liver examinations were performed. RESULTS Mice in the HFD group showed a tendency towards increased body mass gain and liver damage compared with the maraviroc/HFD group. Moreover, liver weight in the HFD group was significantly higher than in the maraviroc/HFD group. Hepatic triglyceride concentration in the maraviroc/HFD group was significantly lower than in the HFD group. Interestingly, the maraviroc/HFD group exhibited a lower degree of steatosis. Furthermore, hepatic CCL5/RANTES expression was significantly lower in the maraviroc/HFD group than in the HFD group. Overall, no differences were observed between the control group and the maraviroc group. CONCLUSIONS Maraviroc ameliorates hepatic steatosis in an experimental model of NAFLD.
Collapse
Affiliation(s)
- Laura Pérez-Martínez
- Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Patricia Pérez-Matute
- Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | | | | | - Judit Narro
- Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Emma Recio
- Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | | | - José-Antonio Oteo
- Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - José-Ramón Blanco
- Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| |
Collapse
|
21
|
Effect of maternal diabetes and hypercholesterolemia on fetal liver of albino Wistar rats. Nutrition 2014; 30:326-36. [DOI: 10.1016/j.nut.2013.08.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 08/19/2013] [Accepted: 08/22/2013] [Indexed: 12/30/2022]
|
22
|
Renaud HJ, Cui JY, Lu H, Klaassen CD. Effect of diet on expression of genes involved in lipid metabolism, oxidative stress, and inflammation in mouse liver-insights into mechanisms of hepatic steatosis. PLoS One 2014; 9:e88584. [PMID: 24551121 PMCID: PMC3925138 DOI: 10.1371/journal.pone.0088584] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 01/08/2014] [Indexed: 12/11/2022] Open
Abstract
Nutritional intake is a fundamental determinant of health. Many studies have correlated excess caloric intake, as well as a high ratio of n-6:n-3 fatty acids, with detrimental health outcomes, such as the metabolic syndrome. In contrast, low-calorie diets have beneficial health effects. Despite these associations, our understanding of the causal relationship between diet and health remains largely elusive. The present study examined the molecular changes elicited by nine diets with varying fat, sugar, cholesterol, omega-3 fatty acids, omega-6 fatty acids, and calories in C57BL/6 male mice. Microarray analyses were conducted on liver samples from three mice per diet and detected 20,449 genes of which 3,734 were responsive to changes in dietary components. Principal component analysis showed that diet restriction correlated the least with the other diets and also affected more genes than any other diet. Interestingly, Gene Set Enrichment Analysis (GSEA) identified gene sets involved in glutathione metabolism, immune response, fatty acid metabolism, cholesterol metabolism, ABC transporters, and oxidative phosphorylation as being highly responsive to changes in diet composition. On the gene level, this study reveals novel findings such as the induction of the drug efflux pump Abcb1a (p-glycoprotein) by diet restriction and an atherogenic diet, as well as the suppression of the rate limiting step of bile acid synthesis, Cyp7a1, by a high fructose diet. This study provides considerable insight into the molecular changes incurred by a variety of diets and furthers our understanding of the causal relationships between diet and health.
Collapse
Affiliation(s)
- Helen J Renaud
- Department of Internal Medicine, Kansas University Medical Center, Kansas City, Kansas, United States of America
| | - Julia Y Cui
- Department of Internal Medicine, Kansas University Medical Center, Kansas City, Kansas, United States of America
| | - Hong Lu
- Department of Pharmacology, State University of New York, Syracuse, New York, United States of America
| | - Curtis D Klaassen
- Department of Internal Medicine, Kansas University Medical Center, Kansas City, Kansas, United States of America
| |
Collapse
|
23
|
Kosters A, Felix JC, Desai MS, Karpen SJ. Impaired bile acid handling and aggravated liver injury in mice expressing a hepatocyte-specific RXRα variant lacking the DNA-binding domain. J Hepatol 2014; 60:362-9. [PMID: 24120911 PMCID: PMC3946967 DOI: 10.1016/j.jhep.2013.09.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 09/20/2013] [Accepted: 09/30/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Retinoid X Receptor α (RXRα) is the principal heterodimerization partner of class II Nuclear Receptors (NRs), and a major regulator of gene expression of numerous hepatic processes, including bile acid (BA) homeostasis through multiple partners. Specific contributions of hepatic RXRα domains in heterodimer function in response to either BA load or ductular cholestasis are not fully characterized. METHODS Wild-type (WT) mice and mice expressing a hepatocyte-specific RXRα lacking the DNA-Binding-Domain (hs-RxrαΔex4(-/-)), which retains partial ability to heterodimerize with its partners, were fed a 1% cholic acid (CA) diet for 5 days, a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet for 3 weeks, or control diet. RESULTS Serum ALT (6.5-fold; p<0.05), AST (9.3-fold; p=0.06) and BA (2.8-fold; p<0.05) were increased in CA-fed hs-RxαΔex4(-/-) mice compared to CA-fed WT mice, but were equally induced between genotypes by DDC-feeding. CA-feeding elevated total (4.4-fold; p=0.06) and unconjugated (2.2-fold; p<0.02) bilirubin levels in hs-RxrαΔex4(-/-) mice compared to WT mice, but not in DDC-fed hs-RxrαΔex4(-/-) mice. Increased necrosis and inflammation was observed in CA-fed, but not in DDC-fed hs-RxrαΔex4(-/-) mice. Apoptotic markers DR5, CK8, CK18 RNA were increased in CA- and DDC-fed hs-RxrαΔex4(-/-) mice. Cleaved caspase 3, CK18 and p-JNK protein were elevated in CA-fed but not in DDC-fed hs-RxrαΔex4(-/-) mice. Induction of Ostβ and Cyp2b10 RNA was impaired in CA-fed and DDC-fed hs-RxrαΔex4(-/-) mice. Surprisingly, DDC-fed hs-RxrαΔex4(-/-) mice showed attenuated fibrosis compared to DDC-fed WT mice. CONCLUSIONS These two models of cholestasis identify common and injury-specific roles for RXRα heterodimers and the functional relevance of an intact RXRα-DBD in the hepatocytic adaptive cholestatic response.
Collapse
Affiliation(s)
- Astrid Kosters
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta GA, 30322
| | - Julio C. Felix
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Moreshwar S. Desai
- Division of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Saul J. Karpen
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta GA, 30322
| |
Collapse
|
24
|
Tintut Y, Demer LL. Effects of bioactive lipids and lipoproteins on bone. Trends Endocrinol Metab 2014; 25:53-9. [PMID: 24183940 PMCID: PMC3946677 DOI: 10.1016/j.tem.2013.10.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 09/30/2013] [Accepted: 10/02/2013] [Indexed: 12/11/2022]
Abstract
Although epidemiological studies from the past two decades show a link between atherosclerotic vascular disease and bone loss, that is independent of age, the mechanism is still unclear. This review focuses on evidence that suggests a role for atherogenic lipids and lipoproteins in the pathogenesis of bone loss, including direct effects of these bioactive lipids/lipoproteins on bone cells, inhibiting osteoblastic differentiation and promoting osteoclastic differentiation. It also addresses recent evidence that suggests that bioactive lipids blunt the effects of bone anabolic agents such as teriparatide and bone morphogenetic proteins. Systemic and intracellular oxidant stress and inflammation are implicated in mediating the effects of bioactive lipids/lipoproteins.
Collapse
Affiliation(s)
- Yin Tintut
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Linda L Demer
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Physiology, University of California, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
25
|
Moreira ELG, de Oliveira J, Engel DF, Walz R, de Bem AF, Farina M, Prediger RDS. Hypercholesterolemia induces short-term spatial memory impairments in mice: up-regulation of acetylcholinesterase activity as an early and causal event? J Neural Transm (Vienna) 2013; 121:415-26. [DOI: 10.1007/s00702-013-1107-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/16/2013] [Indexed: 12/23/2022]
|
26
|
Mehedint MG, Zeisel SH. Choline's role in maintaining liver function: new evidence for epigenetic mechanisms. Curr Opin Clin Nutr Metab Care 2013; 16:339-45. [PMID: 23493015 PMCID: PMC3729018 DOI: 10.1097/mco.0b013e3283600d46] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Humans eating diets low in choline develop fatty liver and liver damage. Rodents fed choline-methionine-deficient diets not only develop fatty liver, but also progress to develop fibrosis and hepatocarcinoma. This review focuses on the role of choline in liver function, with special emphasis on the epigenetic mechanisms of action. RECENT FINDINGS Dietary intake of methyl donors like choline influences the methylation of DNA and histones, thereby altering the epigenetic regulation of gene expression. The liver is the major organ within which methylation reactions occur, and many of the hepatic genes involved in pathways for the development of fatty liver, hepatic fibrosis, and hepatocarcinomas are epigenetically regulated. SUMMARY Dietary intake of choline varies over a three-fold range and many humans have genetic polymorphisms that increase their demand for choline. Choline is an important methyl donor needed for the generation of S-adenosylmethionine. Dietary choline intake is an important modifier of epigenetic marks on DNA and histones, and thereby modulates the gene expression in many of the pathways involved in liver function and dysfunction.
Collapse
Affiliation(s)
- Mihai G Mehedint
- Nutrition Research Institute at Kannapolis, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
27
|
Mild hypercholesterolemia blunts the proinflammatory and prothrombotic effects of hypertension on the cerebral microcirculation. J Cereb Blood Flow Metab 2013; 33:483-9. [PMID: 23281427 PMCID: PMC3618387 DOI: 10.1038/jcbfm.2012.194] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Although an increased leukocyte and platelet adhesion is observed in cerebral venules of mice with either hypertension (HTN) or hypercholesterolemia (HCh), it remains unclear whether the combination of HTN and HCh exerts a comparable effect on leukocyte and platelet recruitment in the cerebral microvasculature. Thus, we examined whether HCh alters platelet and leukocyte adhesion, and blood-brain barrier (BBB) permeability, in cerebral venules in two models of murine HTN: DOCA salt-induced and angiotensin II (Ang II) induced. In both models, the mice were placed on either a normal or cholesterol-enriched diet. An enhanced recruitment of adherent leukocytes and platelets in cerebral venules was noted in both HTN models in the absence of HCh, but not in its presence. The Ang II-induced increase in BBB permeability was attenuated by HCh as well. Both total and high-density lipoprotein (HDL) cholesterol levels were elevated in the HCh mice. The HTN-induced increase in leukocyte and platelet adhesion was attenuated in apolipoprotein A-I transgenic mice (ApoA1-Tg) and blunted in wild-type mice treated with the ApoA1 mimetic peptide, 4F. Our findings indicate that mild HCh significantly blunts the cerebral microvascular responses to HTN and that HDL may have a role in mediating this beneficial effect of HCh.
Collapse
|
28
|
Changes in the hepatic mitochondrial and membrane proteome in mice fed a non-alcoholic steatohepatitis inducing diet. J Proteomics 2013; 80:107-22. [PMID: 23313215 DOI: 10.1016/j.jprot.2012.12.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 11/30/2012] [Accepted: 12/17/2012] [Indexed: 12/20/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) accounts for a large proportion of cryptic cirrhosis in the Western societies. Nevertheless, we lack a deeper understanding of the underlying pathomolecular processes, particularly those preceding hepatic inflammation and fibrosis. In order to gain novel insights into early NASH-development from the first appearance of proteomic alterations to the onset of hepatic inflammation and fibrosis, we conducted a time-course analysis of proteomic changes in liver mitochondria and membrane-enriched fractions of female C57Bl/6N mice fed either a mere steatosis or NASH inducing diet. This data was complemented by quantitative measurements of hepatic glycerol-containing lipids, cholesterol and intermediates of the methionine cycle. Aside from energy metabolism and stress response proteins, enzymes of the urea cycle and methionine metabolism were found regulated. Alterations in the methionine cycle occur early in disease progression preceding molecular signs of inflammation. Proteins that hold particular promise in the early distinction between benign steatosis and NASH are methyl-transferase Mettl7b, the glycoprotein basigin and the microsomal glutathione-transferase.
Collapse
|
29
|
Thomas A, Stevens AP, Klein MS, Hellerbrand C, Dettmer K, Gronwald W, Oefner PJ, Reinders J. Early changes in the liver-soluble proteome from mice fed a nonalcoholic steatohepatitis inducing diet. Proteomics 2012; 12:1437-51. [PMID: 22589191 DOI: 10.1002/pmic.201100628] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Despite the increasing incidence of nonalcoholic steatohepatitis (NASH) with the rise in lifestyle-related diseases such as the metabolic syndrome, little is known about the changes in the liver proteome that precede the onset of inflammation and fibrosis. Here, we investigated early changes in the liver-soluble proteome of female C57BL/6N mice fed an NASH-inducing diet by 2D-DIGE and nano-HPLC-MS/MS. In parallel, histology and measurements of hepatic content of triglycerides, cholesterol and intermediates of the methionine cycle were performed. Hepatic steatosis manifested itself after 2 days of feeding, albeit significant changes in the liver-soluble proteome were not evident before day 10 in the absence of inflammatory or fibrotic signs. Proteomic alterations affected mainly energy and amino acid metabolism, detoxification processes, urea cycle, and the one-carbon/S-adenosylmethionine pathways. Additionally, intermediates of relevant affected pathways were quantified from liver tissue, confirming the findings from the proteomic analysis.
Collapse
Affiliation(s)
- Anja Thomas
- Institute of Functional Genomics, University of Regensburg, Josef-Engert-Strasse 9, Regensburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Bikman BT. A role for sphingolipids in the pathophysiology of obesity-induced inflammation. Cell Mol Life Sci 2012; 69:2135-46. [PMID: 22294100 PMCID: PMC11114706 DOI: 10.1007/s00018-012-0917-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 01/01/2012] [Accepted: 01/04/2012] [Indexed: 12/20/2022]
Abstract
Following the initial discovery that adipose tissue actively synthesizes and secretes cytokines, obesity-induced inflammation has been implicated in the etiology of a host of disease states related to obesity, including cardiovascular disease and type II diabetes. Interestingly, a growing body of evidence similarly implicates sphingolipids as prime instigators in these same diseases. From the recent discovery that obesity-related inflammatory pathways modulate sphingolipid metabolism comes a novel perspective—sphingolipids may act as the dominant mediators of deleterious events stemming from obesity-induced inflammation. This paradigm may identify sphingolipids as an effective target for future therapeutics aimed at ameliorating diseases associated with chronic inflammation.
Collapse
Affiliation(s)
- Benjamin T Bikman
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
31
|
Braunersreuther V, Viviani GL, Mach F, Montecucco F. Role of cytokines and chemokines in non-alcoholic fatty liver disease. World J Gastroenterol 2012; 18:727-35. [PMID: 22371632 PMCID: PMC3286135 DOI: 10.3748/wjg.v18.i8.727] [Citation(s) in RCA: 260] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 07/27/2011] [Accepted: 08/04/2011] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) includes a variety of histological conditions (ranging from liver steatosis and steatohepatitis, to fibrosis and hepatocarcinoma) that are characterized by an increased fat content within the liver. The accumulation/deposition of fat within the liver is essential for diagnosis of NAFLD and might be associated with alterations in the hepatic and systemic inflammatory state. Although it is still unclear if each histological entity represents a different disease or rather steps of the same disease, inflammatory processes in NAFLD might influence its pathophysiology and prognosis. In particular, non-alcoholic steatohepatitis (the most inflamed condition in NAFLDs, which more frequently evolves towards chronic and serious liver diseases) is characterized by a marked activation of inflammatory cells and the upregulation of several soluble inflammatory mediators. Among several mediators, cytokines and chemokines might play a pivotal active role in NAFLD and are considered as potential therapeutic targets. In this review, we will update evidence from both basic research and clinical studies on the potential role of cytokines and chemokines in the pathophysiology of NAFLD.
Collapse
|
32
|
Watanabe S, Tsuneyama K. Cattle bile but not bear bile or pig bile induces lipid profile changes and fatty liver injury in mice: mediation by cholic acid. J Toxicol Sci 2012; 37:105-21. [DOI: 10.2131/jts.37.105] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Shiro Watanabe
- Division of Clinical Application, Department of Clinical Sciences, Institute of Natural Medicine, University of Toyama
| | - Koichi Tsuneyama
- Department of Molecular Pathology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama
| |
Collapse
|
33
|
Conditional transgenic expression of TIR-domain-containing adaptor-inducing interferon-β (TRIF) in the adult mouse heart is protective in acute viral myocarditis. Basic Res Cardiol 2011; 106:1159-71. [DOI: 10.1007/s00395-011-0226-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 09/10/2011] [Accepted: 09/19/2011] [Indexed: 12/29/2022]
|
34
|
Lopes PA, Martins SV, Viana RSJ, Ramalho RM, Alfaia CM, Pinho MS, Jerónimo E, Bessa RJB, Castro MF, Rodrigues CMP, Prates JAM. Contrasting apoptotic responses of conjugated linoleic acid in the liver of obese Zucker rats fed palm oil or ovine fat. Prostaglandins Leukot Essent Fatty Acids 2011; 85:89-96. [PMID: 21600750 DOI: 10.1016/j.plefa.2011.04.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 04/04/2011] [Accepted: 04/09/2011] [Indexed: 12/15/2022]
Abstract
We hypothesized that reducing weight properties of conjugated linoleic acid (CLA) are due to adipocyte apoptosis and that CLA differentially modulates the apoptotic responses in hepatic lipotoxicity from rats fed saturated fat diets. Obese Zucker rats were fed atherogenic diets (2%w/w of cholesterol) formulated with high (15%w/w) saturated fat, from vegetable or animal origin, supplemented or not with 1% of a mixture (1:1) of cis-9, trans-11 and trans-10, cis-12 CLA isomers for 14 weeks. CLA induced no changes on retroperitoneal fat depot weight, which was in line with similar levels of apoptosis. Interestingly, CLA had a contrasting effect on cell death in the liver according to the dietary fat. CLA increased hepatocyte apoptosis, associated with upregulation of Fas protein in rats fed palm oil, compared to rats receiving palm oil alone. However, rats fed ovine fat alone displayed the highest levels of hepatic cell death, which were decreased in rats fed ovine fat plus CLA. This reducing effect of CLA was related to positively restoring endoplasmic reticulum (ER) ATF-6α, BiP and CHOP protein levels and increasing phosphorylated c-Jun NH(2)-terminal kinase (JNK) and c-Jun, thus suggesting an adaptive response of cell survival. These findings reinforce the role of CLA as regulator of apoptosis in the liver. Moreover, the dietary fat composition is a key factor in activation of apoptosis.
Collapse
Affiliation(s)
- Paula A Lopes
- CIISA, Faculdade de Medicina Veterinária, Universidade Técnica de Lisboa, Pólo Universitário do Alto da Ajuda, Lisboa, Portugal.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Drake C, Boutin H, Jones MS, Denes A, McColl BW, Selvarajah JR, Hulme S, Georgiou RF, Hinz R, Gerhard A, Vail A, Prenant C, Julyan P, Maroy R, Brown G, Smigova A, Herholz K, Kassiou M, Crossman D, Francis S, Proctor SD, Russell JC, Hopkins SJ, Tyrrell PJ, Rothwell NJ, Allan SM. Brain inflammation is induced by co-morbidities and risk factors for stroke. Brain Behav Immun 2011; 25:1113-22. [PMID: 21356305 PMCID: PMC3145158 DOI: 10.1016/j.bbi.2011.02.008] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 02/11/2011] [Accepted: 02/12/2011] [Indexed: 01/22/2023] Open
Abstract
Chronic systemic inflammatory conditions, such as atherosclerosis, diabetes and obesity are associated with increased risk of stroke, which suggests that systemic inflammation may contribute to the development of stroke in humans. The hypothesis that systemic inflammation may induce brain pathology can be tested in animals, and this was the key objective of the present study. First, we assessed inflammatory changes in the brain in rodent models of chronic, systemic inflammation. PET imaging revealed increased microglia activation in the brain of JCR-LA (corpulent) rats, which develop atherosclerosis and obesity, compared to the control lean strain. Immunostaining against Iba1 confirmed reactive microgliosis in these animals. An atherogenic diet in apolipoprotein E knock-out (ApoE(-/-)) mice induced microglial activation in the brain parenchyma within 8 weeks and increased expression of vascular adhesion molecules. Focal lipid deposition and neuroinflammation in periventricular and cortical areas and profound recruitment of activated myeloid phagocytes, T cells and granulocytes into the choroid plexus were also observed. In a small, preliminary study, patients at risk of stroke (multiple risk factors for stroke, with chronically elevated C-reactive protein, but negative MRI for brain pathology) exhibited increased inflammation in the brain, as indicated by PET imaging. These findings show that brain inflammation occurs in animals, and tentatively in humans, harbouring risk factors for stroke associated with elevated systemic inflammation. Thus a "primed" inflammatory environment in the brain may exist in individuals at risk of stroke and this can be adequately recapitulated in appropriate co-morbid animal models.
Collapse
Affiliation(s)
- Caroline Drake
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Hervé Boutin
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Matthew S. Jones
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Adam Denes
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Barry W. McColl
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Johann R. Selvarajah
- Clinical Neurosciences Group and Stroke Medicine, Salford Royal Foundation Trust, UK
| | - Sharon Hulme
- Clinical Neurosciences Group and Stroke Medicine, Salford Royal Foundation Trust, UK
| | - Rachel F. Georgiou
- Clinical Neurosciences Group and Stroke Medicine, Salford Royal Foundation Trust, UK
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Alexander Gerhard
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Andy Vail
- Health Methodology Research Group, University of Manchester, UK
| | - Christian Prenant
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Peter Julyan
- North Western Medical Physics, Christie Hospital, Manchester, UK
| | | | - Gavin Brown
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Alison Smigova
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Karl Herholz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Michael Kassiou
- Brain and Mind Research Institute, University of Sydney, NSW 2050, Australia,Discipline of Medical Radiation Sciences, University of Sydney, NSW 1825, Australia,School of Chemistry, University of Sydney, NSW 2006, Australia
| | - David Crossman
- NIHR Biomedical Research Unit, University of Sheffield, Sheffield, UK
| | - Sheila Francis
- Department of Cardiovascular Science, University of Sheffield, UK
| | - Spencer D. Proctor
- Metabolic and Cardiovascular Diseases Laboratory, Alberta Institute for Human Nutrition, University of Alberta, Edmonton, Alberta, Canada
| | - James C. Russell
- Metabolic and Cardiovascular Diseases Laboratory, Alberta Institute for Human Nutrition, University of Alberta, Edmonton, Alberta, Canada
| | - Stephen J. Hopkins
- Clinical Neurosciences Group and Stroke Medicine, Salford Royal Foundation Trust, UK
| | - Pippa J. Tyrrell
- Clinical Neurosciences Group and Stroke Medicine, Salford Royal Foundation Trust, UK
| | | | - Stuart M. Allan
- Faculty of Life Sciences, University of Manchester, Manchester, UK,Corresponding author. Address: Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, UK. Fax: +44 (0) 161 275 3938.
| |
Collapse
|
36
|
Du RH, Li EG, Cao Y, Song YC, Tan RX. Fumigaclavine C inhibits tumor necrosis factor α production via suppression of toll-like receptor 4 and nuclear factor κB activation in macrophages. Life Sci 2011; 89:235-40. [PMID: 21762706 DOI: 10.1016/j.lfs.2011.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 05/29/2011] [Accepted: 06/04/2011] [Indexed: 01/23/2023]
Abstract
AIMS Atherosclerosis is the main cause of cardiovascular disease and is widely treated with statins. However, there are a few cases of intolerable adverse reactions by statins; thus, there is still a need for new drugs to prevent atherosclerosis. The inflammation associated with the activation of Toll-like receptor 4 (TLR4) and nuclear factor κB (NFκB) has been shown to be an important factor in the development of atherosclerosis. In the current study, we investigated the anti-inflammatory action of the fungal alkaloid fumigaclavine C (FC), its effects on the TLR4 and NFκB signaling pathway, and its potential relevance as an anti-atherosclerotic agent. MAIN METHODS The inhibitory effects of FC on tumor necrosis factor α (TNFα) production were determined by enzyme-linked immunosorbent assays (ELISA). The mRNA and protein expression of TLR4 and p65NFκB were detected by quantitative real-time polymerase chain reaction and western blot analysis, respectively. The effect of FC on NFκB was determined using the Dual-Luciferase reporter assay. KEY FINDINGS FC reduced TNFα production in LPS-stimulated human whole blood and RAW 264.7 macrophages via reduced IκBα phosphorylation associated with the decreased expression of p65NFκB. FC also suppressed LPS-induced TLR4 overexpression at the mRNA and protein level. SIGNIFICANCE FC attenuated TNFα via the TLR4-NFκB signaling transduction pathway, suggesting that this alkaloid might serve as a promising molecule for anti-inflammatory treatment of atherosclerosis.
Collapse
Affiliation(s)
- Rong Hui Du
- School of Medicine, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, PR China
| | | | | | | | | |
Collapse
|
37
|
Wanninger J, Neumeier M, Bauer S, Weiss TS, Eisinger K, Walter R, Dorn C, Hellerbrand C, Schäffler A, Buechler C. Adiponectin induces the transforming growth factor decoy receptor BAMBI in human hepatocytes. FEBS Lett 2011; 585:1338-44. [PMID: 21496456 DOI: 10.1016/j.febslet.2011.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 04/01/2011] [Accepted: 04/01/2011] [Indexed: 02/07/2023]
Abstract
Transforming growth factor (TGF) β is the central cytokine in fibrotic liver diseases. We analyzed whether hepatoprotective adiponectin directly interferes with TGFβ1 signaling in primary human hepatocytes (PHH). Adiponectin induces the TGFβ decoy receptor BMP-and activin-membrane-bound inhibitor (BAMBI) in PHH. Overexpression of BAMBI in hepatoma cells impairs TGFβ-mediated phosphorylation of SMAD2 and induction of connective tissue growth factor. BAMBI is lower in human fatty liver with a higher susceptibility to liver fibrosis and negatively correlates with BMI of the donors. Hepatic BAMBI is reduced in rodent models of liver inflammation and fibrosis. In summary, the current data show that hepatoprotective effects of adiponectin include induction of BAMBI which is reduced in human fatty liver and rodent models of metabolic liver injury.
Collapse
Affiliation(s)
- Josef Wanninger
- Department of Internal Medicine I, University Hospital of Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Atherosclerosis aggravates ischemia/reperfusion injury in the gut and remote damage in the liver and the lung. Inflamm Res 2011; 60:555-67. [DOI: 10.1007/s00011-010-0304-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2010] [Revised: 12/13/2010] [Accepted: 12/16/2010] [Indexed: 10/18/2022] Open
|
39
|
Erdely A, Kepka-Lenhart D, Salmen-Muniz R, Chapman R, Hulderman T, Kashon M, Simeonova PP, Morris SM. Arginase activities and global arginine bioavailability in wild-type and ApoE-deficient mice: responses to high fat and high cholesterol diets. PLoS One 2010; 5:e15253. [PMID: 21151916 PMCID: PMC2997799 DOI: 10.1371/journal.pone.0015253] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 11/02/2010] [Indexed: 12/03/2022] Open
Abstract
Increased catabolism of arginine by arginase is increasingly viewed as an important pathophysiological factor in cardiovascular disease, including atherosclerosis induced by high cholesterol diets. Whereas previous studies have focused primarily on effects of high cholesterol diets on arginase expression and arginine metabolism in specific blood vessels, there is no information regarding the impact of lipid diets on arginase activity or arginine bioavailability at a systemic level. We, therefore, evaluated the effects of high fat (HF) and high fat-high cholesterol (HC) diets on arginase activity in plasma and tissues and on global arginine bioavailability (defined as the ratio of plasma arginine to ornithine + citrulline) in apoE−/− and wild-type C57BL/6J mice. HC and HF diets led to reduced global arginine bioavailability in both strains. The HC diet resulted in significantly elevated plasma arginase in both strains, but the HF diet increased plasma arginase only in apoE−/− mice. Elevated plasma arginase activity correlated closely with increased alanine aminotransferase levels, indicating that liver damage was primarily responsible for elevated plasma arginase. The HC diet, which promotes atherogenesis, also resulted in increased arginase activity and expression of the type II isozyme of arginase in multiple tissues of apoE−/− mice only. These results raise the possibility that systemic changes in arginase activity and global arginine bioavailability may be contributing factors in the initiation and/or progression of cardiovascular disease.
Collapse
Affiliation(s)
- Aaron Erdely
- Toxicology and Molecular Biology Branch, National Institute for Occupational Safety and Health, Morgantown, West Virginia, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Kirovski G, Gäbele E, Dorn C, Moleda L, Niessen C, Weiss TS, Wobser H, Schacherer D, Buechler C, Wasmuth HE, Hellerbrand C. Hepatic steatosis causes induction of the chemokine RANTES in the absence of significant hepatic inflammation. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2010. [PMID: 20830238 DOI: 10.1055/s-0029-1246351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum ranging from simple steatosis to cirrhosis. Hepatocellular lipid accumulation is a hallmark of both nonalcoholic steatosis and steatohepatitis (NASH). The latter develops upon pro-inflammatory cell infiltration and is widely considered as the first relevant pathophysiological step in NAFLD-progression. The chemokine CCL5/RANTES plays an important role in the progression of hepatic inflammation and fibrosis. We here aimed to investigate its expression in NAFLD. Incubation of primary human hepatocytes with palmitic acid induced a dose-dependent lipid accumulation, and corresponding dose-dependent RANTES induction in vitro. Furthermore, we observed significantly elevated hepatic RANTES expression in a dietary model of NAFLD, in which mice were fed a high-fat diet for 12 weeks. This diet induced significant hepatic steatosis but only minimal inflammation. In contrast to the liver, RANTES expression was not induced in visceral adipose tissue of the group fed with high-fat diet. Finally, RANTES serum levels were elevated in patients with ultrasound-diagnosed NAFLD. In conclusion, our data indicate hepatocytes as cellular source of elevated hepatic as well as circulating RANTES levels in response to hepatic steatosis. Noteworthy, upregulation of RANTES in response to lipid accumulation occurs in the absence of relevant inflammation, which further indicates that hepatic steatosis per se has pathophysiological relevance and should not be considered as benign.
Collapse
Affiliation(s)
- Georgi Kirovski
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Swindell WR, Johnston A, Gudjonsson JE. Transcriptional profiles of leukocyte populations provide a tool for interpreting gene expression patterns associated with high fat diet in mice. PLoS One 2010; 5:e11861. [PMID: 20686622 PMCID: PMC2912331 DOI: 10.1371/journal.pone.0011861] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Accepted: 07/05/2010] [Indexed: 12/24/2022] Open
Abstract
Background Microarray experiments in mice have shown that high fat diet can lead to elevated expression of genes that are disproportionately associated with immune functions. These effects of high fat (atherogenic) diet may be due to infiltration of tissues by leukocytes in coordination with inflammatory processes. Methodology/Principal Findings The Novartis strain-diet-sex microarray database (GSE10493) was used to evaluate the hepatic effects of high fat diet (4 weeks) in 12 mouse strains and both genders. We develop and apply an algorithm that identifies “signature transcripts” for many different leukocyte populations (e.g., T cells, B cells, macrophages) and uses this information to derive an in silico “inflammation profile”. Inflammation profiles highlighted monocytes, macrophages and dendritic cells as key drivers of gene expression patterns associated with high fat diet in liver. In some strains (e.g., NZB/BINJ, B6), we estimate that 50–60% of transcripts elevated by high fat diet might be due to hepatic infiltration by these cell types. Interestingly, DBA mice appeared to exhibit resistance to localized hepatic inflammation associated with atherogenic diet. A common characteristic of infiltrating cell populations was elevated expression of genes encoding components of the toll-like receptor signaling pathway (e.g., Irf5 and Myd88). Conclusions/Significance High fat diet promotes infiltration of hepatic tissue by leukocytes, leading to elevated expression of immune-associated transcripts. The intensity of this effect is genetically controlled and sensitive to both strain and gender. The algorithm developed in this paper provides a framework for computational analysis of tissue remodeling processes and can be usefully applied to any in vivo setting in which inflammatory processes play a prominent role.
Collapse
Affiliation(s)
- William R Swindell
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America.
| | | | | |
Collapse
|
42
|
Lewis DK, Bake S, Thomas K, Jezierski MK, Sohrabji F. A high cholesterol diet elevates hippocampal cytokine expression in an age and estrogen-dependent manner in female rats. J Neuroimmunol 2010; 223:31-8. [PMID: 20435353 DOI: 10.1016/j.jneuroim.2010.03.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 03/05/2010] [Accepted: 03/31/2010] [Indexed: 02/07/2023]
Abstract
BACKGROUND While the effects of a proatherogenic diet have been widely studied in the context of systemic inflammation, much less is known about its effects on central or brain inflammation and its modulation with age. In this study, we examined the effect of a high cholesterol/choline diet in adult and older acyclic females to assess its impact on systemic and central inflammatory markers. Moreover, since the loss of ovarian hormones at menopause may predispose women to increased production of pro-inflammatory cytokines, we also tested the impact of estrogen replacement to adult and older females in diet-induced inflammation. METHODS Ovariectomized adult female rats and older (reproductive senescent) female rats were replaced with estrogen or a control pellet and maintained thereafter on a diet containing either 4% cholesterol/1% choline or control chow for 10 weeks. Interleukin 1beta (IL-1beta) expression in the liver was used as a marker of systemic inflammation, while a panel of cytokine/chemokines were used to examine the effects of diet on the hippocampus. RESULTS IL-1beta expression was elevated in the liver of adult and reproductive senescent females fed with the high cholesterol diet, although this was restricted to groups that were ovariectomized and not replaced with estrogen. Estrogen-treated animals of both ages did not have elevated IL-1beta levels when fed the high cholesterol diet. Diet-induced changes in cytokine/chemokine expression in the hippocampus however were critically age dependent and restricted to the reproductive senescent females. In this group, the high cholesterol diet led to an increase in interleukin (IL)-4, IL-6, IL-12p70, IL-13, RANTES (Regulated on Activation, Normal T Expressed and Secreted) and VEGF (vascular endothelial growth factor). Moreover, estrogen treatment to reproductive senescent females suppressed diet-induced expression of specific cytokines (RANTES, VEGF, IL-6) and attenuated the expression of others (IL-4, IL-12p70, and IL-13). CONCLUSIONS These data indicate that a proatherogenic diet presents a significant risk for central inflammation in older females that are deprived of estrogen treatment.
Collapse
Affiliation(s)
- Danielle K Lewis
- Department of Neuroscience and Experimental Therapeutics and Women's Health in Neuroscience Program, TAM Health Science Center, College Station, TX 77843-1114, USA.
| | | | | | | | | |
Collapse
|
43
|
Peterson CY, Costantini TW, Loomis WH, Putnam JG, Wolf P, Bansal V, Eliceiri BP, Baird A, Coimbra R. Toll-like receptor-4 mediates intestinal barrier breakdown after thermal injury. Surg Infect (Larchmt) 2010; 11:137-44. [PMID: 20374005 DOI: 10.1089/sur.2009.053] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Toll-like receptor 4 (TLR-4) activation after sterile injury leads to organ dysfunction at distant sites. We have shown previously that intestinal barrier breakdown and alteration of tight junction proteins follows thermal injury; however, the role of TLR-4 in this process remains unclear. We hypothesized that increased intestinal permeability and barrier breakdown after burns is a TLR-4 dependent process; hence, knocking down the TLR-4 gene would have a protective effect on burn-induced intestinal dysfunction. METHODS Male C57BL/6J (TLR-4 wild type [WT]) and C57BL/10ScN (TLR-4 knockout [KO]) mice were assigned randomly to either 30% total body surface area steam burn or sham injury. At 4 h, permeability to intraluminally administered fluorescein isothiocyanate (FITC)-dextran was assessed by measuring the fluorescence of the serum. Intestinal samples were analyzed for the presence of the tight junction protein occludin by immunoblotting and immunohistochemistry. Tumor necrosis factor (TNF)-alpha concentrations in the serum and intestines were measured by enzyme-linked immunosorbent assay at 2 h post-burn. RESULTS Serum concentrations of FITC-dextran were decreased in TLR-4 KO mice compared with TLR-4 WT mice after burn injury (92.0 micrograms/mL and 264.5 micrograms/mL, respectively; p < 0.05). After injury, no difference in intestinal permeability was observed between the TLR-4 KO mice and the TLR-4 WT sham-treated mice. The TLR-4 KO mice had preservation of occludin concentrations after thermal injury in both immunoblot and immunohistochemistry assays, but concentrations were decreased in TLR-4 WT animals. The serum concentrations of TNF-alpha serum were higher in TLR-4 WT burned animals than in the sham-treated mice. The TLR-4 KO animals had unmeasurable concentrations of TNF-alpha. No differences in TNF-alpha were observed in the intestinal tissue at 2 h. CONCLUSIONS Mice with TLR-4 KO have less intestinal permeability to FITC-dextran than do TLR-4 WT mice after burn injury as a result of alterations in the tight junction protein occludin. These findings suggest that the greater intestinal permeability and barrier breakdown after burn injury is a TLR-4-dependent process. Toll-like receptor 4 may provide a useful target for the prevention and treatment of systemic inflammatory response syndrome and multisystem organ failure after injury.
Collapse
Affiliation(s)
- Carrie Y Peterson
- Division of Trauma, Critical Care and Burns, Department of Surgery, University of California, San Diego, San Diego, California 92103-8896, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Samokhin AO, Wilson S, Nho B, Lizame MLG, Musenden OEE, Brömme D. Cholate-containing high-fat diet induces the formation of multinucleated giant cells in atherosclerotic plaques of apolipoprotein E-/- mice. Arterioscler Thromb Vasc Biol 2010; 30:1166-73. [PMID: 20203298 DOI: 10.1161/atvbaha.110.203976] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To determine the role of multinucleated giant cells (MGCs) in cardiovascular diseases. METHODS AND RESULTS MGCs are a hallmark of giant cell arteritis. They are also described in atherosclerotic plaques from aortic aneurysms and carotid and coronary arteries. Herein, we demonstrate that the cholate-containing Paigen diet yields many MGCs in atherosclerotic plaques of apolipoprotein E-/- mice. These mice revealed a 4-fold increase in MGC numbers when compared with mice on a Western or Paigen diet without cholate. Most of the MGCs stained intensively for cathepsin K and were located at fibrous caps and close to damaged elastic laminae, with associated medial smooth muscle cell depletion. During in vitro experiments, MGCs demonstrated a 6-fold increase in elastolytic activity when compared with macrophages and facilitated transmigration of smooth muscle cells through a collagen-elastin matrix. An elastin-derived hexapeptide (Val-Gly-Val-Ala-Pro-Gly [VGVAPG]) significantly increased the rate of macrophage fusion, providing a possible mechanism of in vivo MGC formation. Comparable to the mouse model, human specimens from carotid arteries and aortic aneurysms contained cathepsin K-positive MGCs. CONCLUSIONS Apolipoprotein E-/- mice fed a Paigen diet provide a model to analyze the tissue-destructive role of MGCs in vascular diseases.
Collapse
Affiliation(s)
- Andriy O Samokhin
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | |
Collapse
|
45
|
Chronic intake of a high-cholesterol diet resulted in hepatic steatosis, focal nodular hyperplasia and fibrosis in non-obese mice. Br J Nutr 2009; 103:378-85. [DOI: 10.1017/s0007114509991772] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We investigated the effects of a high-cholesterol (HC) diet administered long term (25 or 55 weeks) on metabolic disorders including hepatic damage in mice. The mice were fed the HC diet (15 % milk fat, 1·5 % cholesterol and 0·1 % cholic acid, w/w) for 25 or 55 weeks. Body and adipose tissue weights were similar to those of mice fed a control diet. Consumption of the HC diet long term resulted in hypercholesterolaemia, hepatic steatosis and gallstones. In addition, focal nodular hyperplasia (FNH) and mild fibrosis of the liver developed in all mice fed the HC diet for 55 weeks. Plasma levels of monocyte chemoattractant protein (MCP)-1 were elevated, and the level of hepatic platelet-derived growth factor (PDGF)-B protein was increased in mice fed the HC diet compared with those fed the control diet. Thus, it seems likely that the liver fibrosis and FNH caused by the long-term consumption of a HC diet may be partly due to an elevation of plasma MCP-1 and hepatic PDGF expression.
Collapse
|
46
|
Masson D, Koseki M, Ishibashi M, Larson CJ, Miller SG, King BD, Tall AR. Increased HDL cholesterol and apoA-I in humans and mice treated with a novel SR-BI inhibitor. Arterioscler Thromb Vasc Biol 2009; 29:2054-60. [PMID: 19815817 DOI: 10.1161/atvbaha.109.191320] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Increasing HDL levels is a potential strategy for the treatment of atherosclerosis. METHODS AND RESULTS ITX5061, a molecule initially characterized as a p38 MAPK inhibitor, increased HDL-C levels by 20% in a human population of hypertriglyceridemic subjects with low HDL levels. ITX5061 also moderately increased apoA-I but did not affect VLDL/LDL cholesterol or plasma triglyceride concentrations. ITX5061 increased HDL-C in WT and human apoA-I transgenic mice, and kinetic experiments showed that ITX5061 decreased the fractional catabolic rate of HDL-CE and reduced its hepatic uptake. In transfected cells, ITX5061 inhibited SR-BI-dependent uptake of HDL-CE. Moreover, ITX5061 failed to increase HDL-C levels in SR-BI(-/-) mice. To assess effects on atherosclerosis, ITX5061 was given to atherogenic diet-fed Ldlr(+/-) mice with or without CETP expression for 18 weeks. In both the control and CETP-expressing groups, ITX5061-treated mice displayed reductions of early atherosclerotic lesions in the aortic arch -40%, P<0.05), and a nonsignificant trend to reduced lesion area in the proximal aorta. CONCLUSIONS Our data indicate that ITX5061 increases HDL-C levels by inhibition of SR-BI activity. This suggests that pharmacological inhibition of SR-BI has the potential to raise HDL-C and apoA-I levels without adverse effects on VLDL/LDL cholesterol levels in humans.
Collapse
Affiliation(s)
- David Masson
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Lohmann C, Schäfer N, von Lukowicz T, Sokrates Stein MA, Borén J, Rütti S, Wahli W, Donath MY, Lüscher TF, Matter CM. Atherosclerotic mice exhibit systemic inflammation in periadventitial and visceral adipose tissue, liver, and pancreatic islets. Atherosclerosis 2009; 207:360-7. [PMID: 19481752 DOI: 10.1016/j.atherosclerosis.2009.05.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2008] [Revised: 04/16/2009] [Accepted: 05/05/2009] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Atherosclerosis is a chronic inflammatory disease of major conduit arteries. Similarly, obesity and type 2 diabetes mellitus are associated with accumulation of macrophages in visceral white adipose tissue and pancreatic islets. Our goal was to characterize systemic inflammation in atherosclerosis with hypercholesterolemia, but without obesity. METHODS AND RESULTS We compared 22-week-old apolipoprotein E knockout (ApoE(-/-)) with wild-type mice kept for 14 weeks on a high cholesterol (1.25%) diet (CD, n=8) and 8-week-old ApoE(-/-) with wild-type mice kept on a normal diet (ND, n=8). Hypercholesterolemic, atherosclerotic ApoE(-/-) mice on CD exhibited increased macrophages and T-cells in plaques and periadventitial adipose tissue that revealed elevated expression of MIP-1alpha, IL-1beta, IL-1 receptor, and IL-6. Mesenteric adipose tissue and pancreatic islets in ApoE(-/-) mice showed increased macrophages. Expression of IL-1beta was enhanced in mesenteric adipose tissue of ApoE(-/-) mice on CD. Furthermore, these mice exhibited steatohepatitis with macrophage and T-cell infiltrations as well as increased MIP-1alpha and IL-1 receptor expression. Blood glucose, insulin and total body weight did not differ between the groups. CONCLUSIONS In hypercholesterolemic lean ApoE(-/-) mice, inflammation extends beyond atherosclerotic plaques to the periadventitial and visceral adipose tissue, liver, and pancreatic islets without affecting glucose homeostasis.
Collapse
Affiliation(s)
- Christine Lohmann
- Cardiovascular Research, Institute of Physiology, Zurich University and Cardiology, Cardiovascular Center, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|