51
|
Effects of cold stress on juvenile Piaractus mesopotamicus and the mitigation by β-carotene. J Therm Biol 2019; 88:102497. [PMID: 32125985 DOI: 10.1016/j.jtherbio.2019.102497] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/12/2019] [Accepted: 12/26/2019] [Indexed: 02/07/2023]
Abstract
This study investigated the effects of cold stress on morphometrical and hematological biomarkers, energy metabolism, and oxidative stress in different tissues of P. mesopotamicus, and the protective role of β-carotene. Fish were fed with a control diet (CD) and the same diet supplemented with 105 mg/kg β-carotene (BD) for 60 days. After the feeding trial, fish fed CD or BD diets were exposed to control (24 °C) and low temperature (14 °C) for 24 h. Fish (CD and BD) exposed to thermal stress showed lower hepatosomatic index. The hemoglobin increased only in CD-fed fish exposed to 14 °C. Increased glycemia, plasmatic protein depletion, and decreased hepatic glycogen were observed in fish fed the CD, while only the lipid levels in liver were augmented in BD-fed fish exposed at 14 °C. Regarding the oxidative stress, increased antioxidant enzymes activity and lipid peroxidation were observed in CD-fed fish exposed to cold. The two-way ANOVA showed an interaction between dietary treatment and temperature for glucose and oxidative stress biomarkers, with the highest values recorded in 14 °C-exposed fish fed with the CD. Our study demonstrated that cold stress had the greatest impact on fish oxidative status, and β-carotene reduces harmful effects induced by cold in P. mesopotamicus.
Collapse
|
52
|
Carpenter KJ, Valfort AC, Steinauer N, Chatterjee A, Abuirqeba S, Majidi S, Sengupta M, Di Paolo RJ, Shornick LP, Zhang J, Flaveny CA. LXR-inverse agonism stimulates immune-mediated tumor destruction by enhancing CD8 T-cell activity in triple negative breast cancer. Sci Rep 2019; 9:19530. [PMID: 31863071 PMCID: PMC6925117 DOI: 10.1038/s41598-019-56038-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/05/2019] [Indexed: 01/21/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype that is untreatable with hormonal or HER2-targeted therapies and is also typically unresponsive to checkpoint-blockade immunotherapy. Within the tumor microenvironment dysregulated immune cell metabolism has emerged as a key mechanism of tumor immune-evasion. We have discovered that the Liver-X-Receptors (LXRα and LXRβ), nuclear receptors known to regulate lipid metabolism and tumor-immune interaction, are highly activated in TNBC tumor associated myeloid cells. We therefore theorized that inhibiting LXR would induce immune-mediated TNBC-tumor clearance. Here we show that pharmacological inhibition of LXR activity induces tumor destruction primarily through stimulation of CD8+ T-cell cytotoxic activity and mitochondrial metabolism. Our results imply that LXR inverse agonists may be a promising new class of TNBC immunotherapies.
Collapse
Affiliation(s)
- Katherine J Carpenter
- The Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA
| | - Aurore-Cecile Valfort
- The Center for Clinical Pharmacology, Saint Louis College of Pharmacy, Saint Louis, MO, 63110, USA
| | - Nick Steinauer
- The Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA
| | - Arindam Chatterjee
- The Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA
| | - Suomia Abuirqeba
- The Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA
| | - Shabnam Majidi
- The Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA
| | - Monideepa Sengupta
- The Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA
| | - Richard J Di Paolo
- The Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA.,The Alvin J. Siteman Cancer Center at Barnes-Jewish and Washington University School of Medicine in Saint Louis, Saint Louis, MO, 63110, USA
| | - Laurie P Shornick
- The Department of Biology, Saint Louis University, Saint Louis, MO, 63103, USA
| | - Jinsong Zhang
- The Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA.,The Alvin J. Siteman Cancer Center at Barnes-Jewish and Washington University School of Medicine in Saint Louis, Saint Louis, MO, 63110, USA
| | - Colin A Flaveny
- The Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA. .,The Alvin J. Siteman Cancer Center at Barnes-Jewish and Washington University School of Medicine in Saint Louis, Saint Louis, MO, 63110, USA.
| |
Collapse
|
53
|
Lifsey HC, Kaur R, Thompson BH, Bennett L, Temel RE, Graf GA. Stigmasterol stimulates transintestinal cholesterol excretion independent of liver X receptor activation in the small intestine. J Nutr Biochem 2019; 76:108263. [PMID: 31759199 DOI: 10.1016/j.jnutbio.2019.108263] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 10/14/2019] [Accepted: 10/29/2019] [Indexed: 02/09/2023]
Abstract
Despite advances in healthcare, cardiovascular disease (CVD) remains the leading cause of death in the United States. Elevated levels of plasma cholesterol are highly predictive of CVD and stroke and are the principal driver of atherosclerosis. Unfortunately, current cholesterol lowering agents, such as statins, are not known to reverse atherosclerotic disease once it has been established. In preclinical models, agonists of nuclear receptor, LXR, have been shown to reduce and reverse atherosclerosis. Phytosterols are bioactive non-cholesterol sterols that act as LXR agonists and regulate cholesterol metabolism and transport. We hypothesized that stigmasterol would act as an LXR agonist and alter intestinal cholesterol secretion to promote cholesterol elimination. Mice were fed a control diet, or a diet supplemented with stigmasterol (0.3% w/w) or T0901317 (0.015% w/w), a known LXR agonist. In this experiment we analyzed the sterol content of bile, intestinal perfusate, plasma, and feces. Additionally, the liver and small intestine were analyzed for relative levels of transcripts known to be regulated by LXR. We observed that T0901317 robustly promoted cholesterol elimination and acted as a strong LXR agonist. Stigmasterol promoted transintestinal cholesterol secretion through an LXR-independent pathway.
Collapse
Affiliation(s)
| | - Rupinder Kaur
- Department of Pharmaceutical Sciences, College of Pharmacy
| | | | - Lisa Bennett
- Department of Pharmaceutical Sciences, College of Pharmacy
| | - Ryan E Temel
- Department of Physiology, College of Medicine, University of Kentucky; Saha Cardiovascular Research Center
| | - Gregory A Graf
- Department of Pharmaceutical Sciences, College of Pharmacy; Saha Cardiovascular Research Center; Barnstable Brown Diabetes and Obesity Center.
| |
Collapse
|
54
|
Chang RC, Thomas KN, Bedi YS, Golding MC. Programmed increases in LXRα induced by paternal alcohol use enhance offspring metabolic adaptation to high-fat diet induced obesity. Mol Metab 2019; 30:161-172. [PMID: 31767168 PMCID: PMC6807343 DOI: 10.1016/j.molmet.2019.09.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/25/2019] [Accepted: 09/29/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives Paternally inherited alterations in epigenetic programming are emerging as relevant factors in numerous disease states, including the growth and metabolic defects observed in fetal alcohol spectrum disorders. In rodents, chronic paternal alcohol use induces fetal growth restriction, as well as sex-specific alterations in insulin signaling and lipid homeostasis in the offspring. Based on previous studies, we hypothesized that the observed metabolic irregularities are the consequence of paternally inherited alterations liver x receptor (LXR) activity. Methods Male offspring of alcohol-exposed sires were challenged with a high-fat diet and the molecular pathways controlling glucose and lipid homeostasis assayed for LXR-induced alterations. Results Similar to findings in studies employing LXR agonists we found that the male offspring of alcohol-exposed sires display resistance to diet-induced obesity and improved glucose homeostasis when challenged with a high-fat diet. This improved metabolic adaptation is mediated by LXRα trans-repression of inflammatory cytokines, releasing IKKβ inhibition of the insulin signaling pathway. Interestingly, paternally programmed increases in LXRα expression are liver-specific and do not manifest in the pancreas or visceral fat. Conclusions These studies identify LXRα as a key mediator of the long-term metabolic alterations induced by preconception paternal alcohol use. Chronic paternal alcohol use induces up-regulation of LXRα in the male offspring. Male offspring of alcohol-exposed fathers are protected from diet-induced obesity. Paternally-inherited up-regulation of LXRα only manifests in the liver. Improved metabolic adaptation is linked to LXRα suppression of cytokine production. Male offspring exhibit the same phenotypes observed in studies of LXR agonists.
Collapse
Affiliation(s)
- Richard C Chang
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Kara N Thomas
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Yudhishtar S Bedi
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Michael C Golding
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
55
|
Naeini MB, Momtazi AA, Jaafari MR, Johnston TP, Barreto G, Banach M, Sahebkar A. Antitumor effects of curcumin: A lipid perspective. J Cell Physiol 2019; 234:14743-14758. [PMID: 30741424 DOI: 10.1002/jcp.28262] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/06/2019] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Lipid metabolism plays an important role in cancer development due to the necessities of rapidly dividing cells to increase structural, energetic, and biosynthetic demands for cell proliferation. Basically, obesity, type 2 diabetes, and other related diseases, and cancer are associated with a common hyperactivated "lipogenic state." Recent evidence suggests that metabolic reprogramming and overproduction of enzymes involved in the synthesis of fatty acids are the new hallmarks of cancer, which occur in an early phase of tumorigenesis. As the first evidence to confirm dysregulated lipid metabolism in cancer cells, the overexpression of fatty acid synthase (FAS) was observed in breast cancer patients and demonstrated the role of FAS in cancer. Other enzymes of fatty acid synthesis have recently been found to be dysregulated in cancer, including ATP-dependent citrate lyase and acetyl-CoA carboxylase, which further underscores the connection of these metabolic pathways with cancer cell survival and proliferation. The degree of overexpression of lipogenic enzymes and elevated lipid utilization in tumors is closely associated with cancer progression. The question that arises is whether the progression of cancer can be suppressed, or at least decelerated, by modulating gene expression related to fatty acid metabolism. Curcumin, due to its effects on the regulation of lipogenic enzymes, might be able to suppress, or even cause regression of tumor growth. This review discusses recent evidence concerning the important role of lipogenic enzymes in the metabolism of cancer cells and whether the inhibitory effects of curcumin on lipogenic enzymes is therapeutically efficacious.
Collapse
Affiliation(s)
- Mehri Bemani Naeini
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Momtazi
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri
| | - George Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C, Colombia.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Maciej Banach
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
56
|
Fitz NF, Nam KN, Koldamova R, Lefterov I. Therapeutic targeting of nuclear receptors, liver X and retinoid X receptors, for Alzheimer's disease. Br J Pharmacol 2019; 176:3599-3610. [PMID: 30924124 PMCID: PMC6715597 DOI: 10.1111/bph.14668] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/15/2019] [Accepted: 02/24/2019] [Indexed: 12/18/2022] Open
Abstract
After 15 years of research into Alzheimer's disease (AD) therapeutics, including billions of US dollars provided by federal agencies, pharmaceutical companies, and private foundations, there are still no meaningful therapies that can delay the onset or slow the progression of AD. An understanding of the proteolytic processing of amyloid precursor protein (APP) and the hypothesis that pathogenic mechanisms in familial and sporadic forms of AD are very similar led to the assumption that pharmacological inhibition of secretases or immunological approaches to clear amyloid depositions in the brain would have been the core to drug discovery strategies and successful therapies. However, there are other understudied approaches including targeting genes, gene networks, and metabolic pathways outside the proteolytic processing of APP. The advancement of newly developed sequencing technologies and mass spectrometry, as well as the availability of animal models expressing human apolipoprotein E isoforms, has been critical in rationalizing additional AD therapeutics. The purpose of this review is to present one of those approaches, based on the role of ligand-activated nuclear liver X and retinoid X receptors in the brain. This therapeutic approach was initially proposed utilizing in vitro models 15 years ago and has since been examined in numerous studies using AD-like mouse models. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Nicholas F Fitz
- Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kyong Nyon Nam
- Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Radosveta Koldamova
- Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Iliya Lefterov
- Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
57
|
Mouchiroud M, Camiré É, Aldow M, Caron A, Jubinville É, Turcotte L, Kaci I, Beaulieu MJ, Roy C, Labbé SM, Varin TV, Gélinas Y, Lamothe J, Trottier J, Mitchell PL, Guénard F, Festuccia WT, Joubert P, Rose CF, Karvellas CJ, Barbier O, Morissette MC, Marette A, Laplante M. The hepatokine Tsukushi is released in response to NAFLD and impacts cholesterol homeostasis. JCI Insight 2019; 4:129492. [PMID: 31391339 PMCID: PMC6693835 DOI: 10.1172/jci.insight.129492] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/27/2019] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) prevails in obesity and is linked to several health complications including dyslipidemia and atherosclerosis. How exactly NAFLD induces atherogenic dyslipidemia to promote cardiovascular diseases is still elusive. Here, we identify Tsukushi (TSK) as a hepatokine induced in response to NAFLD. We show that both endoplasmic reticulum stress and inflammation promote the expression and release of TSK in mice. In humans, hepatic TSK expression is also associated with steatosis, and its circulating levels are markedly increased in patients suffering from acetaminophen-induced acute liver failure (ALF), a condition linked to severe hepatic inflammation. In these patients, elevated blood TSK levels were associated with decreased transplant-free survival at hospital discharge, suggesting that TSK could have a prognostic significance. Gain- and loss-of-function studies in mice revealed that TSK impacts systemic cholesterol homeostasis. TSK reduces circulating HDL cholesterol, lowers cholesterol efflux capacity, and decreases cholesterol-to-bile acid conversion in the liver. Our data identify the hepatokine TSK as a blood biomarker of liver stress that could link NAFLD to the development of atherogenic dyslipidemia and atherosclerosis.
Collapse
Affiliation(s)
- Mathilde Mouchiroud
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Étienne Camiré
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Manal Aldow
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Alexandre Caron
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Éric Jubinville
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Laurie Turcotte
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Inès Kaci
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Marie-Josée Beaulieu
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Christian Roy
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Sébastien M. Labbé
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
- IPS Thérapeutique, Sherbrooke, Québec, Canada
| | - Thibault V. Varin
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec City, Québec, Canada
| | - Yves Gélinas
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Jennifer Lamothe
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Jocelyn Trottier
- Laboratory of Molecular Pharmacology, Endocrinology-Nephrology Axis, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec City, Québec, Canada
- Faculty of Pharmacy, Université Laval, Québec City, Québec, Canada
| | - Patricia L. Mitchell
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Frédéric Guénard
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec City, Québec, Canada
| | - William T. Festuccia
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Philippe Joubert
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
| | - Christopher F. Rose
- Hepato-Neuro Laboratory, Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Constantine J. Karvellas
- Liver Unit, Division of Gastroenterology, Department of Critical Care Medicine, School of Public Health Science, University of Alberta, Edmonton, Alberta, Canada
| | - Olivier Barbier
- Laboratory of Molecular Pharmacology, Endocrinology-Nephrology Axis, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec City, Québec, Canada
- Faculty of Pharmacy, Université Laval, Québec City, Québec, Canada
| | - Mathieu C. Morissette
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
- Département de Médecine and
| | - André Marette
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec City, Québec, Canada
- Département de Médecine and
| | - Mathieu Laplante
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec – Université Laval (CRIUCPQ), Québec City, Québec, Canada
- Département de Médecine and
- Centre de Recherche sur le Cancer de l’Université Laval, Université Laval, Québec City, Québec, Canada
| |
Collapse
|
58
|
Jalil A, Bourgeois T, Ménégaut L, Lagrost L, Thomas C, Masson D. Revisiting the Role of LXRs in PUFA Metabolism and Phospholipid Homeostasis. Int J Mol Sci 2019; 20:ijms20153787. [PMID: 31382500 PMCID: PMC6696407 DOI: 10.3390/ijms20153787] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 01/19/2023] Open
Abstract
Liver X receptors (LXRs) play a pivotal role in fatty acid (FA) metabolism. So far, the lipogenic consequences of in vivo LXR activation, as characterized by a major hepatic steatosis, has constituted a limitation to the clinical development of pharmacological LXR agonists. However, recent studies provided a different perspective. Beyond the quantitative accumulation of FA, it appears that LXRs induce qualitative changes in the FA profile and in the distribution of FAs among cellular lipid species. Thus, LXRs activate the production of polyunsaturated fatty acids (PUFAs) and their distribution into phospholipids via the control of FA desaturases, FA elongases, lysophosphatidylcholine acyltransferase (LPCAT3), and phospholipid transfer protein (PLTP). Therefore, LXRs control, in a dynamic manner, the PUFA composition and the physicochemical properties of cell membranes as well as the release of PUFA-derived lipid mediators. Recent studies suggest that modulation of PUFA and phospholipid metabolism by LXRs are involved in the control of lipogenesis and lipoprotein secretion by the liver. In myeloid cells, the interplay between LXR and PUFA metabolism affects the inflammatory response. Revisiting the complex role of LXRs in FA metabolism may open new opportunities for the development of LXR modulators in the field of cardiometabolic diseases.
Collapse
Affiliation(s)
- Antoine Jalil
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Thibaut Bourgeois
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Louise Ménégaut
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Laurent Lagrost
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Charles Thomas
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - David Masson
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France.
- INSERM, LNC UMR 1231, F-21000 Dijon, France.
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France.
| |
Collapse
|
59
|
Zwarts I, van Zutphen T, Kruit JK, Liu W, Oosterveer MH, Verkade HJ, Uhlenhaut NH, Jonker JW. Identification of the fructose transporter GLUT5 (SLC2A5) as a novel target of nuclear receptor LXR. Sci Rep 2019; 9:9299. [PMID: 31243309 PMCID: PMC6594926 DOI: 10.1038/s41598-019-45803-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 06/12/2019] [Indexed: 12/24/2022] Open
Abstract
Fructose has become a major constituent of our modern diet and is implicated as an underlying cause in the development of metabolic diseases. The fructose transporter GLUT5 (SLC2A5) is required for intestinal fructose absorption. GLUT5 expression is induced in the intestine and skeletal muscle of type 2 diabetes (T2D) patients and in certain cancers that are dependent on fructose metabolism, indicating that modulation of GLUT5 levels could have potential in the treatment of these diseases. Using an unbiased screen for transcriptional control of the human GLUT5 promoter we identified a strong and specific regulation by liver X receptor α (LXRα, NR1H3). Using promoter truncations and site-directed mutagenesis we identified a functional LXR response element (LXRE) in the human GLUT5 promoter, located at −385 bp relative to the transcriptional start site (TSS). Finally, mice treated with LXR agonist T0901317 showed an increase in Glut5 mRNA and protein levels in duodenum and adipose tissue, underscoring the in vivo relevance of its regulation by LXR. Together, our findings show that LXRα regulates GLUT5 in mice and humans. As a ligand-activated transcription factor, LXRα might provide novel pharmacologic strategies for the selective modulation of GLUT5 activity in the treatment of metabolic disease as well as cancer.
Collapse
Affiliation(s)
- Irene Zwarts
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Tim van Zutphen
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Janine K Kruit
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Weilin Liu
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Maaike H Oosterveer
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Henkjan J Verkade
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - N Henriette Uhlenhaut
- Molecular Endocrinology, Institute for Diabetes and Cancer IDC, Helmholtz Zentrum München, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany.,Gene Center, Ludwig-Maximilians-Universität München (LMU), Feodor-Lynen-Straße 25, Munich, 81377, Germany
| | - Johan W Jonker
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
60
|
Mansukhani NA, Peters EB, So MM, Albaghdadi MS, Wang Z, Karver MR, Clemons TD, Laux JP, Tsihlis ND, Stupp SI, Kibbe MR. Peptide Amphiphile Supramolecular Nanostructures as a Targeted Therapy for Atherosclerosis. Macromol Biosci 2019; 19:e1900066. [PMID: 31066494 PMCID: PMC6579116 DOI: 10.1002/mabi.201900066] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022]
Abstract
The rising prevalence of cardiovascular disease worldwide necessitates novel therapeutic approaches to manage atherosclerosis. Intravenously administered nanostructures are a promising noninvasive approach to deliver therapeutics that reduce plaque burden. The drug liver X receptor agonist GW3965 (LXR) can reduce atherosclerosis by promoting cholesterol efflux from plaque but causes liver toxicity when administered systemically at effective doses, thus preventing its clinical use. The ability of peptide amphiphile nanofibers containing apolipoprotein A1-derived targeting peptide 4F to serve as nanocarriers for LXR delivery (ApoA1-LXR PA) in vivo is investigated here. These nanostructures are found to successfully target atherosclerotic lesions in a mouse model within 24 h of injection. After 8 weeks of intravenous administration, the nanostructures significantly reduce plaque burden in both male and female mice to a similar extent as LXR alone in comparison to saline-treated controls. Furthermore, they do not cause increased liver toxicity in comparison to LXR treatments, which may be related to more controlled release by the nanostructure. These findings demonstrate the potential of supramolecular nanostructures as safe, effective drug nanocarriers to manage atherosclerosis.
Collapse
Affiliation(s)
- Neel A. Mansukhani
- Division of Vascular Surgery, Department of Surgery, Feinberg School of Medicine, 251 East Huron St., Galter 3-150, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, 303 E. Superior, Suite 11-131, Northwestern University, Chicago, IL 60611, USA
| | - Erica B. Peters
- Department of Surgery, 3001 Burnett-Womack Building, 101 Manning Drive, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Miranda M. So
- Simpson Querrey Institute, 303 E. Superior, Suite 11-131, Northwestern University, Chicago, IL 60611, USA
- Department of Materials Science and Engineering, McCormick School of Engineering and Applied Science, 2220 Campus Drive, Room 2036, Northwestern University, Evanston, IL 60208, USA
| | - Mazen S. Albaghdadi
- Division of Vascular Surgery, Department of Surgery, Feinberg School of Medicine, 251 East Huron St., Galter 3-150, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, 303 E. Superior, Suite 11-131, Northwestern University, Chicago, IL 60611, USA
| | - Zheng Wang
- Division of Vascular Surgery, Department of Surgery, Feinberg School of Medicine, 251 East Huron St., Galter 3-150, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, 303 E. Superior, Suite 11-131, Northwestern University, Chicago, IL 60611, USA
| | - Mark R. Karver
- Simpson Querrey Institute, 303 E. Superior, Suite 11-131, Northwestern University, Chicago, IL 60611, USA
| | - Tristan D. Clemons
- Simpson Querrey Institute, 303 E. Superior, Suite 11-131, Northwestern University, Chicago, IL 60611, USA
- Department of Chemistry, 2145 N. Sheridan Rd., Tech K148, Northwestern University, Evanston, IL 60208, USA
| | - Jeffrey P. Laux
- Biostatistics, Epidemiology and Research Design, NC Translational and Clinical Sciences Institute, University of North Carolina, Chapel Hill, NC 27599-7420
| | - Nick D. Tsihlis
- Division of Vascular Surgery, Department of Surgery, Feinberg School of Medicine, 251 East Huron St., Galter 3-150, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, 303 E. Superior, Suite 11-131, Northwestern University, Chicago, IL 60611, USA
- Department of Surgery, 3001 Burnett-Womack Building, 101 Manning Drive, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Samuel I. Stupp
- Simpson Querrey Institute, 303 E. Superior, Suite 11-131, Northwestern University, Chicago, IL 60611, USA
- Department of Materials Science and Engineering, McCormick School of Engineering and Applied Science, 2220 Campus Drive, Room 2036, Northwestern University, Evanston, IL 60208, USA
- Department of Chemistry, 2145 N. Sheridan Rd., Tech K148, Northwestern University, Evanston, IL 60208, USA
- Department of Medicine, 676 N. St. Clair St., Arkes Suite 2330, Northwestern University, Chicago, IL 60611, USA
- Department of Biomedical Engineering, 2145 N. Sheridan Rd., Technological Institute, Northwestern University, Evanston, IL 60208, USA
| | - Melina R. Kibbe
- Division of Vascular Surgery, Department of Surgery, Feinberg School of Medicine, 251 East Huron St., Galter 3-150, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, 303 E. Superior, Suite 11-131, Northwestern University, Chicago, IL 60611, USA
- Department of Surgery, 3001 Burnett-Womack Building, 101 Manning Drive, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Bioengineering, 333 South Columbia Street, University of North Carolina, Chapel Hill, NC, 27514, USA
| |
Collapse
|
61
|
Pastore M, Grimaudo S, Pipitone RM, Lori G, Raggi C, Petta S, Marra F. Role of Myeloid-Epithelial-Reproductive Tyrosine Kinase and Macrophage Polarization in the Progression of Atherosclerotic Lesions Associated With Nonalcoholic Fatty Liver Disease. Front Pharmacol 2019; 10:604. [PMID: 31191323 PMCID: PMC6548874 DOI: 10.3389/fphar.2019.00604] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022] Open
Abstract
Recent lines of evidence highlight the involvement of myeloid-epithelial-reproductive tyrosine kinase (MerTK) in metabolic disease associated with liver damage. MerTK is mainly expressed in anti-inflammatory M2 macrophages where it mediates transcriptional changes including suppression of proinflammatory cytokines and enhancement of inflammatory repressors. MerTK is regulated by metabolic pathways through nuclear sensors including LXRs, PPARs, and RXRs, in response to apoptotic bodies or to other sources of cholesterol. Nonalcoholic fatty liver disease (NAFLD) is one of the most serious public health problems worldwide. It is a clinicopathological syndrome closely related to obesity, insulin resistance, and oxidative stress. It includes a spectrum of conditions ranging from simple steatosis, characterized by hepatic fat accumulation with or without inflammation, to nonalcoholic steatohepatitis (NASH), defined by hepatic fat deposition with hepatocellular damage, inflammation, and accumulating fibrosis. Several studies support an association between NAFLD and the incidence of cardiovascular diseases including atherosclerosis, a major cause of death worldwide. This pathological condition consists in a chronic and progressive inflammatory process in the intimal layer of large- and medium-sized arteries. The complications of advanced atherosclerosis include chronic or acute ischemic damage in the tissue perfused by the affected artery, leading to cellular death. By identifying specific targets influencing lipid metabolism and cardiovascular-related diseases, the present review highlights the role of MerTK in NAFLD-associated atherosclerotic lesions as a potential innovative therapeutic target. Therapeutic advantages might derive from the use of compounds selective for nuclear receptors targeting PPARs rather than LXRs regulating macrophage lipid metabolism and macrophage mediated inflammation, by favoring the expression of MerTK, which mediates an immunoregulatory action with a reduction in inflammation and in atherosclerosis.
Collapse
Affiliation(s)
- Mirella Pastore
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Stefania Grimaudo
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Palermo, Italy
| | - Rosaria Maria Pipitone
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Palermo, Italy
| | - Giulia Lori
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Chiara Raggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Humanitas Clinical and Research Center, Rozzano, Italy
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Palermo, Italy
| | - Fabio Marra
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
62
|
Liang X, Cao Y, Xiang S, Xiang Z. LXRα-mediated downregulation of EGFR suppress colorectal cancer cell proliferation. J Cell Biochem 2019; 120:17391-17404. [PMID: 31104333 DOI: 10.1002/jcb.29003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
Abstract
Liver X receptors (LXRs) are members of the nuclear receptor family, including the LXRα (NR1H3) and LXRβ (NR1H2) subtypes, which are related to the metabolism of glucose and cholesterol and possess anti-inflammatory functions. Mounting evidence has linked LXRs to the inhibition of cell proliferation in a variety of cancers. We revealed a differential distribution for NR1H3, but not for NR1H2, in colorectal cancer and adjacent normal tissues. We found that NR1H3 enhanced the inhibitory action of GW3965, an agonist of LXRs, on the proliferation of colorectal cancer cells. Upregulation of NR1H3 enhanced the inhibition of cell proliferation by GW3965 while silencing of NR1H3 attenuated the inhibitory effect of GW3965 on cell proliferation. Bioinformatic prediction and luciferase assays showed that NR1H3 was able to inhibit the activity of the epidermal growth factor receptor (EGFR) promoter. Moreover, we demonstrated that activation of NR1H3 inhibited the growth of transplanted tumors in an animal experiment, with the inhibition accompanied by downregulation of EGFR. Our findings suggest that NR1H3 controls cell proliferation by affecting EGFR promoter activity. The high expression of EGFR was due to the downregulation of NR1H3 which is a novel molecular mechanism in the development of colorectal cancer.
Collapse
Affiliation(s)
- Xiaolong Liang
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Cao
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Song Xiang
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zheng Xiang
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
63
|
Chen J, Yue J, Liu J, Liu Y, Jiao KL, Teng MY, Hu CY, Zhen J, Wu MX, Zhou M, Li Z, Li Y. Salvianolic acids improve liver lipid metabolism in ovariectomized rats via blocking STAT-3/SREBP1 signaling. Chin J Nat Med 2019; 16:838-845. [PMID: 30502765 DOI: 10.1016/s1875-5364(18)30125-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Indexed: 12/17/2022]
Abstract
Postmenopausal women, who have reduced circulating estrogen levels, are more prone to develop obesity and related metabolic diseases than premenopausal women. The absence of safe and effective treatments for postmenopausal obesity has changed the focus to natural products as alternative remedies. Total salvianolic acids (TSA) are the major water-soluble ingredients of Danshen. Salvianolic acid (SA) is the major constituent of the TSA. Salvianolic acids, including TSA and SA, are widely used in traditional Chinese medicine. In the present study, ovariectomized rats and LO2 cells were used to study the effects of salvianolic acids on body weight gain and hepatic steatosis. Salvianolic acids reduced ovariectomy (OVX)-induced body weight gain, attenuated the expressions of hepatic lipogenic genes, such as sterol regulatory element binding protein (SREBP)1, fatty acid synthase (FAS), and stearoyl-CoA desaturase (SCD)1, and decreased the liver triglyceride (TG) and total cholesterol (TC). For the molecular mechanisms, OVX and high glucose-induced phosphorylation of signal transducer and activator of transcription (STAT)-3 was inhibited by salvianolic acids treatment. In LO2 cells, inhibition of STAT-3 by siRNA attenuated the increased expression of SREBP1 and TG induced by high glucose. Salvianolic acids reduced the upregulation of SREBP1 and TG induced by high glucose in LO2 cells. In conclusion, these findings illustrated that salvianolic acids markedly alleviated the lipid metabolism disorders and protected against the postmenopausal obesity. The underlying mechanism was probably associated with the regulation of STAT-3 signaling.
Collapse
Affiliation(s)
- Juan Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jia Yue
- Department of Nutrition and Food Hygiene, School of Public Health, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Jiao Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yun Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Kai-Lin Jiao
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Meng-Ying Teng
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Chun-Yan Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jing Zhen
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Mao-Xuan Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ming Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| | - Yuan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
64
|
Dietary Sargassum fusiforme improves memory and reduces amyloid plaque load in an Alzheimer's disease mouse model. Sci Rep 2019; 9:4908. [PMID: 30894635 PMCID: PMC6426980 DOI: 10.1038/s41598-019-41399-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 03/05/2019] [Indexed: 12/12/2022] Open
Abstract
Activation of liver X receptors (LXRs) by synthetic agonists was found to improve cognition in Alzheimer's disease (AD) mice. However, these LXR agonists induce hypertriglyceridemia and hepatic steatosis, hampering their use in the clinic. We hypothesized that phytosterols as LXR agonists enhance cognition in AD without affecting plasma and hepatic triglycerides. Phytosterols previously reported to activate LXRs were tested in a luciferase-based LXR reporter assay. Using this assay, we found that phytosterols commonly present in a Western type diet in physiological concentrations do not activate LXRs. However, a lipid extract of the 24(S)-Saringosterol-containing seaweed Sargassum fusiforme did potently activate LXRβ. Dietary supplementation of crude Sargassum fusiforme or a Sargassum fusiforme-derived lipid extract to AD mice significantly improved short-term memory and reduced hippocampal Aβ plaque load by 81%. Notably, none of the side effects typically induced by full synthetic LXR agonists were observed. In contrast, administration of the synthetic LXRα activator, AZ876, did not improve cognition and resulted in the accumulation of lipid droplets in the liver. Administration of Sargassum fusiforme-derived 24(S)-Saringosterol to cultured neurons reduced the secretion of Aβ42. Moreover, conditioned medium from 24(S)-Saringosterol-treated astrocytes added to microglia increased phagocytosis of Aβ. Our data show that Sargassum fusiforme improves cognition and alleviates AD pathology. This may be explained at least partly by 24(S)-Saringosterol-mediated LXRβ activation.
Collapse
|
65
|
Xu N, Li J, Gao Y, Zhou N, Ma Q, Wu M, Zhang Y, Sun X, Xie J, Shen G, Yang M, Tu Q, Xu X, Zhu J, Tao J. Apoptotic cell-mimicking gold nanocages loaded with LXR agonist for attenuating the progression of murine systemic lupus erythematosus. Biomaterials 2019; 197:380-392. [DOI: 10.1016/j.biomaterials.2019.01.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 12/31/2018] [Accepted: 01/20/2019] [Indexed: 02/07/2023]
|
66
|
Hoekstra M, Nahon JE, de Jong LM, Kröner MJ, de Leeuw LR, Van Eck M. Inhibition of PRMT3 activity reduces hepatic steatosis without altering atherosclerosis susceptibility in apoE knockout mice. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1402-1409. [PMID: 30776415 DOI: 10.1016/j.bbadis.2019.02.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 02/07/2019] [Accepted: 02/12/2019] [Indexed: 12/22/2022]
Abstract
The nuclear receptor liver X receptor (LXR) impacts on cholesterol metabolism as well as hepatic lipogenesis via transcriptional regulation. It is proposed that inhibition of the protein arginine methyltransferase 3 (PRMT3) uncouples these two transcriptional pathways in vivo by acting as a specific lipogenic coactivator of LXR. Here we validated the hypothesis that treatment with the allosteric PRMT3 inhibitor SGC707 will diminish the hepatic steatosis extent, while leaving global cholesterol metabolism, important in cholesterol-driven pathologies like atherosclerosis, untouched. For this purpose, 12-week old hyperlipidemic apolipoprotein E knockout mice were fed a Western-type diet for six weeks to induce both hepatic steatosis and atherosclerosis. The mice received 3 intraperitoneal injections with SGC707 or solvent control per week. Mice chronically treated with SGC707 developed less severe hepatic steatosis as exemplified by the 51% reduced (P < 0.05) liver triglyceride levels. In contrast, the extent of in vivo macrophage foam cell formation and aortic root atherosclerosis was not affected by SGC707 treatment. Interestingly, SGC707-treated mice gained 94% less body weight (P < 0.05), which was paralleled by changes in white adipose tissue morphology, i.e. reduction in adipocyte size and browning. In conclusion, we have shown that through PRMT3 inhibitor treatment specific functions of LXR involved in respectively the development of fatty liver disease and atherosclerosis can be uncoupled, resulting in an overall diminished hepatic steatosis extent without a negative impact on atherosclerosis susceptibility. As such, our studies highlight that PRMT3 inhibition may constitute a novel therapeutic approach to limit the development of fatty liver disease in humans.
Collapse
Affiliation(s)
- Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands..
| | - Joya E Nahon
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Laura M de Jong
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Mara J Kröner
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Lidewij R de Leeuw
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Miranda Van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands
| |
Collapse
|
67
|
Nguyen LT, Chen H, Zaky A, Pollock C, Saad S. SIRT1 overexpression attenuates offspring metabolic and liver disorders as a result of maternal high-fat feeding. J Physiol 2018; 597:467-480. [PMID: 30381838 DOI: 10.1113/jp276957] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/28/2018] [Indexed: 12/22/2022] Open
Abstract
KEY POINTS Maternal high-fat diet (MHF) consumption led to metabolic and liver disorders in male offspring, which are associated with reduced sirtuin (SIRT)1 expression and activity in the offspring liver SIRT1 overexpression in MHF offspring reduced their body weight and adiposity and normalized lipid metabolic markers in epididymal and retroperitoneal adipose tissues SIRT1 overexpression in MHF offspring improved glucose tolerance, as well as systemic and hepatic insulin sensitivity SIRT1 overexpression ameliorated MHF-induced lipogenesis, oxidative stress and fibrogenesis in the liver of offspring. ABSTRACT Maternal obesity can increase the risk of metabolic disorders in the offspring. However, the underlying mechanism responsible for this is not clearly understood. Previous evidence implied that sirtuin (SIRT)1, a potent regulator of energy metabolism and stress responses, may play an important role. In the present study, we have shown, in C57BL/6 mice, that maternal high-fat diet (HFD) consumption can induce a pre-diabetic and non-alcoholic fatty liver disease phenotype in the offspring, associated with reduced SIRT1 expression in the hypothalamus, white adipose tissues (WAT) and liver. Importantly, the overexpression of SIRT1 in these offspring significantly attenuated the excessive accumulation of epididymal (Epi) white adipose tissue (WAT) and retroperitoneal (Rp)WAT (P < 0.001), glucose intolerance and insulin resistance (both P < 0.05) at weaning age. These changes were associated with the suppression of peroxisome proliferator-activated receptor gamma (PPAR)γ (P < 0.01), PPARγ-coactivator 1-alpha (P < 0.05) and sterol regulatory element-binding protein-1c in EpiWAT (P < 0.01), whereas there was increased expression of PPARγ in RpWAT (P < 0.05). In the liver, PPARγ mRNA expression, as well as Akt protein expression and activity, were increased (P < 0.05), whereas fatty acid synthase and carbohydrate response element binding protein were downregulated (P < 0.05), supporting increased insulin sensitivity and reduced lipogenesis in the liver. In addition, hepatic expression of endogenous anti-oxidants, including glutathione peroxidase 1 and catalase, was increased (P < 0.01 and P < 0.05 respectively), whereas collagen and fibronectin deposition was suppressed (P < 0.01). Collectively, the present study provides direct evidence of the mechanistic significance of SIRT1 in maternal HFD-induced metabolic dysfunction in offspring and suggests that SIRT1 is a promising target for fetal reprogramming.
Collapse
Affiliation(s)
- Long T Nguyen
- Renal medicine, Kolling Institute, Royal North Shore Hospital, University of Sydney, Sydney, NSW, Australia.,School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Amgad Zaky
- Renal medicine, Kolling Institute, Royal North Shore Hospital, University of Sydney, Sydney, NSW, Australia
| | - Carol Pollock
- Renal medicine, Kolling Institute, Royal North Shore Hospital, University of Sydney, Sydney, NSW, Australia
| | - Sonia Saad
- Renal medicine, Kolling Institute, Royal North Shore Hospital, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
68
|
López-Lluch G, Hernández-Camacho JD, Fernández-Ayala DJM, Navas P. Mitochondrial dysfunction in metabolism and ageing: shared mechanisms and outcomes? Biogerontology 2018; 19:461-480. [PMID: 30143941 DOI: 10.1007/s10522-018-9768-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/21/2018] [Indexed: 12/15/2022]
Abstract
Mitochondria are key in the metabolism of aerobic organisms and in ageing progression and age-related diseases. Mitochondria are essential for obtaining ATP from glucose and fatty acids but also in many other essential functions in cells including aminoacids metabolism, pyridine synthesis, phospholipid modifications and calcium regulation. On the other hand, the activity of mitochondria is also the principal source of reactive oxygen species in cells. Ageing and chronic age-related diseases are associated with the deregulation of cell metabolism and dysfunction of mitochondria. Cell metabolism is controlled by three major nutritional sensors: mTOR, AMPK and Sirtuins. These factors control mitochondrial biogenesis and dynamics by regulating fusion, fission and turnover through mito- and autophagy. A complex interaction between the activity of these nutritional sensors, mitochondrial biogenesis rate and dynamics exists and affect ageing, age-related diseases including metabolic disease. Further, mitochondria maintain a constant communication with nucleus modulating gene expression and modifying epigenetics. In this review we highlight the importance of mitochondria in ageing and the repercussion in the progression of age-related diseases and metabolic disease.
Collapse
Affiliation(s)
- Guillermo López-Lluch
- Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide, Carretera de Utrera km. 1, 41013, Seville, Spain.
| | - Juan Diego Hernández-Camacho
- Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide, Carretera de Utrera km. 1, 41013, Seville, Spain
| | - Daniel J Moreno Fernández-Ayala
- Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide, Carretera de Utrera km. 1, 41013, Seville, Spain
| | - Plácido Navas
- Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide, Carretera de Utrera km. 1, 41013, Seville, Spain
| |
Collapse
|
69
|
Grefhorst A, van den Beukel JC, Dijk W, Steenbergen J, Voortman GJ, Leeuwenburgh S, Visser TJ, Kersten S, Friesema ECH, Themmen APN, Visser JA. Multiple effects of cold exposure on livers of male mice. J Endocrinol 2018; 238:91-106. [PMID: 29743343 DOI: 10.1530/joe-18-0076] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/09/2018] [Indexed: 02/02/2023]
Abstract
Cold exposure of mice is a common method to stimulate brown adipose tissue (BAT) activity and induce browning of white adipose tissue (WAT) that has beneficial effects on whole-body lipid metabolism, including reduced plasma triglyceride (TG) concentrations. The liver is a key regulatory organ in lipid metabolism as it can take up as well as oxidize fatty acids. The liver can also synthesize, store and secrete TGs in VLDL particles. The effects of cold exposure on murine hepatic lipid metabolism have not been addressed. Here, we report the effects of 24-h exposure to 4°C on parameters of hepatic lipid metabolism of male C57BL/6J mice. Cold exposure increased hepatic TG concentrations by 2-fold (P < 0.05) but reduced hepatic lipogenic gene expression. Hepatic expression of genes encoding proteins involved in cholesterol synthesis and uptake such as the LDL receptor (LDLR) was significantly increased upon cold exposure. Hepatic expression of Cyp7a1 encoding the rate-limiting enzyme in the classical bile acid (BA) synthesis pathway was increased by 4.3-fold (P < 0.05). Hepatic BA concentrations and fecal BA excretion were increased by 2.8- and 1.3-fold, respectively (P < 0.05 for both). VLDL-TG secretion was reduced by approximately 50% after 24 h of cold exposure (P < 0.05). In conclusion, cold exposure has various, likely intertwined effects on the liver that should be taken into account when studying the effects of cold exposure on whole-body metabolism.
Collapse
Affiliation(s)
- Aldo Grefhorst
- Section of EndocrinologyDepartment of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Johanna C van den Beukel
- Section of EndocrinologyDepartment of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Wieneke Dijk
- Division of Human NutritionNutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands
| | - Jacobie Steenbergen
- Section of EndocrinologyDepartment of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Gardi J Voortman
- Section of PharmacologyVascular and Metabolic Diseases, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Selmar Leeuwenburgh
- Section of EndocrinologyDepartment of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Theo J Visser
- Section of EndocrinologyDepartment of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Sander Kersten
- Division of Human NutritionNutrition, Metabolism, and Genomics Group, Wageningen University, Wageningen, The Netherlands
| | - Edith C H Friesema
- Section of PharmacologyVascular and Metabolic Diseases, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Axel P N Themmen
- Section of EndocrinologyDepartment of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jenny A Visser
- Section of EndocrinologyDepartment of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
70
|
Nahon JE, Groeneveldt C, Geerling JJ, van Eck M, Hoekstra M. Inhibition of protein arginine methyltransferase 3 activity selectively impairs liver X receptor-driven transcription of hepatic lipogenic genes in vivo. Br J Pharmacol 2018; 175:3175-3183. [PMID: 29774529 PMCID: PMC6031883 DOI: 10.1111/bph.14361] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Agonists for the liver X receptor (LXR) are considered promising therapeutic moieties in cholesterol-driven diseases by promoting cellular cholesterol efflux pathways. However, current clinical application of these agents is hampered by concomitant LXR-induced activation of a lipogenic transcriptional network, leading to hepatic steatosis. Recent studies have suggested that protein arginine methyltransferase 3 (PRMT3) may act as a selective co-activator of LXR activity. Here, we verified the hypothesis that PRMT3 inhibition selectively disrupts the ability of LXR to stimulate lipogenesis while maintaining its capacity to modulate macrophage cholesterol homeostasis. EXPERIMENTAL APPROACH A combination of the LXR agonist T0901317 and palm oil was administered to C57BL/6 mice to maximally stimulate LXR and PRMT3 activity. PRMT3 activity was inhibited using the allosteric inhibitor SGC707. KEY RESULTS Treatment with SGC707 did not negatively influence the T0901317/palm oil-induced up-regulation of the cholesterol efflux ATP-binding cassette transporter genes, ABCA1 and ABCG1, in peritoneal cells. In contrast, SGC707 treatment was associated with a significant decrease in the hepatic expression of the lipogenic gene fatty acid synthase (-64%). A similar trend was observed for stearoyl-coenzyme A desaturase and acetyl CoA carboxylase expression (-43%; -56%). This obstruction of lipogenic gene transcription coincided with a significant 2.3-fold decrease in liver triglyceride content as compared with the T0901317 and palm oil-treated control group. CONCLUSION AND IMPLICATIONS We showed that inhibition of PRMT3 activity by SGC707 treatment selectively impairs LXR-driven transcription of hepatic lipogenic genes, while the positive effect of LXR stimulation on macrophage cholesterol efflux pathways is maintained.
Collapse
Affiliation(s)
- Joya E Nahon
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Leiden, The Netherlands
| | - Christianne Groeneveldt
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Leiden, The Netherlands
| | - Janine J Geerling
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Leiden, The Netherlands
| | - Miranda van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Leiden, The Netherlands
| | - Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Leiden, The Netherlands
| |
Collapse
|
71
|
Sengupta M, Griffett K, Flaveny CA, Burris TP. Inhibition of Hepatotoxicity by a LXR Inverse Agonist in a Model of Alcoholic Liver Disease. ACS Pharmacol Transl Sci 2018; 1:50-60. [PMID: 31696159 DOI: 10.1021/acsptsci.8b00003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alcohol abuse is a major cause of liver disease and mortality worldwide and is a significant public health issue. Patients with alcoholic liver disease (ALD) have severe hepatic lipid accumulation, inflammation, and fibrosis. Therapies for ALD are very limited and even abstinence from alcohol consumption does not necessarily protect patients from progression of the disease. We sought to evaluate the efficacy of a liver X receptor (LXR) inverse agonist, SR9238, in an animal model of ALD. SR9238 suppresses hepatic lipogenesis, a pathological hallmark of ALD, and we hypothesized that targeting suppression of hepatic metabolic pathways that are activated in ALD may be an effective treatment for the disease. A chronic ethanol diet with or without a final ethanol binge treatment was used to induce ALD in mice. Mice were administered the liver specific LXR inverse agonist SR9238 for 4 weeks after the mice had been maintained on the ethanol diet for 14 days. Mice developed all the hallmarks of advanced ALD demonstrating significant pathophysiology and hepatotoxicity. SR9238 significantly attenuated liver injury and hepatic steatosis and fibrosis was nearly eliminated in SR9238 treated mice. SR9238 treatment reversed the damage associated with chronic ethanol use returning the liver to near normal morphology. These results indicate that inhibiting LXR activity using the inverse agonist has a hepatoprotective effect in rodent models of ALD; thus, this pharmacological approach may be efficacious for treatment of ALD in humans.
Collapse
Affiliation(s)
- Monideepa Sengupta
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Kristine Griffett
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Colin A Flaveny
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Thomas P Burris
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| |
Collapse
|
72
|
Li YW, Wang CH, Chen CJ, Wang CCN, Lin CL, Cheng WK, Shen HY, Lim YP. Effects of antiepileptic drugs on lipogenic gene regulation and hyperlipidemia risk in Taiwan: a nationwide population-based cohort study and supporting in vitro studies. Arch Toxicol 2018; 92:2829-2844. [DOI: 10.1007/s00204-018-2263-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 04/25/2018] [Indexed: 11/30/2022]
|
73
|
Blocking of STAT-3/SREBP1-mediated glucose-lipid metabolism is involved in dietary phytoestrogen-inhibited ovariectomized-induced body weight gain in rats. J Nutr Biochem 2018; 61:17-23. [PMID: 30179725 DOI: 10.1016/j.jnutbio.2018.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 05/23/2018] [Accepted: 06/25/2018] [Indexed: 12/24/2022]
Abstract
Postmenopausal women have a decline in circulating estrogen levels and are more prone to obesity and its related metabolic diseases than premenopausal women are. The absence of safe and effective conventional treatments for postmenopausal obesity has changed the focus to natural products as alternative remedies. Here, ovariectomized rats and LO2 cells were used to study the molecular basis of the effect of dietary phytoestrogens on body weight gain and hepatic steatosis. Dietary phytoestrogens can inhibit ovariectomy (OVX)-induced body weight gain, blood glucose concentration, expression of hepatic lipogenic genes, such as sterol regulatory element binding protein (SREBP)1, acetyl-CoA carboxylase (ACC)1, fatty acid synthase (FAS), and stearoyl-CoA desaturase (SCD)1, and decrease liver triglyceride (TG) content, but later estradiol withdrawal increased expression of SREBP1. Histological analysis of liver showed that dietary phytoestrogens improved OVX-induced morphological abnormalities. OVX and high glucose-induced phosphorylation of signal transducer and activator of transcription (STAT)-3 were inhibited by phytoestrogens treatment. In LO2 cells, inhibition of STAT-3 by siRNA attenuated the increased TG content and expression of SREBP1 induced by high glucose. Phytoestrogens reduced the upregulation of SREBP1 and TG induced by high glucose in LO2 cells. In conclusion, these findings illustrated that dietary phytoestrogens markedly alleviated the derangement of lipid metabolism. The underlying mechanism is probably associated with regulating STAT-3/SREBP1 signaling.
Collapse
|
74
|
Catani MV, Gasperi V, Bisogno T, Maccarrone M. Essential Dietary Bioactive Lipids in Neuroinflammatory Diseases. Antioxid Redox Signal 2018; 29:37-60. [PMID: 28637354 PMCID: PMC5984567 DOI: 10.1089/ars.2016.6958] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 06/19/2017] [Accepted: 06/20/2017] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Under physiological conditions, neurons and glia are in a healthy, redox-balanced environment; when injury perturbs this equilibrium, a neuroinflammatory state is established by activated microglia that triggers pro-inflammatory responses and alters the oxidant/antioxidant balance, thus leading to neuronal loss and neurodegeneration. In neurodegenerative diseases (such as Alzheimer's disease, Parkinson's disease, amyothrophic lateral sclerosis, and multiple sclerosis), the brain is in a constitutively self-sustaining cycle of inflammation and oxidative stress that prompts and amplifies brain damage. Recent Advances: Recently, an increasing amount of scientific data highlight the ability of specific nutrients to cross the blood-brain barrier, and to modulate inflammatory and oxidative pathways. Therefore, nutritional approaches may contribute to restore the lost equilibrium among factors accounting for neurodegeneration. CRITICAL ISSUES Herein, we critically examine how essential lipids (including fatty acids, liposoluble vitamins and phytosterols) might contribute to accelerate or prevent the onset and progression of such pathologies. In particular, we highlight that experimental and clinical findings, although promising, are still inadequate to draw definitive conclusions. FUTURE DIRECTIONS More research is warranted in order to establish the real impact of lipid intake on brain health, especially when redox balance and inflammatory responses have been already compromised. In the future, it would be hoped to gain a detailed knowledge of chemical modifications and dynamic properties of such nutrients, before planning to exploit them as potential therapeutics. Antioxid. Redox Signal. 29, 37-60.
Collapse
Affiliation(s)
- Maria Valeria Catani
- Department of Experimental Medicine and Surgery, Tor Vergata University of Rome, Rome, Italy
| | - Valeria Gasperi
- Department of Experimental Medicine and Surgery, Tor Vergata University of Rome, Rome, Italy
| | - Tiziana Bisogno
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Italy
- Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
| | - Mauro Maccarrone
- Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
- European Center for Brain Research/Santa Lucia Foundation IRCCS, Rome, Italy
| |
Collapse
|
75
|
Angrish MM, McQueen CA, Cohen-Hubal E, Bruno M, Ge Y, Chorley BN. Editor's Highlight: Mechanistic Toxicity Tests Based on an Adverse Outcome Pathway Network for Hepatic Steatosis. Toxicol Sci 2018; 159:159-169. [PMID: 28903485 DOI: 10.1093/toxsci/kfx121] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Risk assessors use liver endpoints in rodent toxicology studies to assess the safety of chemical exposures. Yet, rodent endpoints may not accurately reflect human responses. For this reason and others, human-based invitro models are being developed and anchored to adverse outcome pathways to better predict adverse human health outcomes. Here, a networked adverse outcome pathway-guided selection of biology-based assays for lipid uptake, lipid efflux, fatty acid oxidation, and lipid accumulation were developed. These assays were evaluated in a metabolically competent human hepatocyte cell model (HepaRG) exposed to compounds known to cause steatosis (amiodarone, cyclosporine A, and T0901317) or activate lipid metabolism pathways (troglitazone, Wyeth-14,643, and 22(R)-hydroxycholesterol). All of the chemicals activated at least one assay, however, only T0901317 and cyclosporin A dose-dependently increased lipid accumulation. T0901317 and cyclosporin A increased fatty acid uptake, decreased lipid efflux (inferred from apolipoprotein B100 levels), and increased fatty acid synthase protein levels. Using this biologically-based evaluation of key events regulating hepatic lipid levels, we demonstrated dysregulation of compensatory pathways that normally balance hepatic lipid levels. This approach may provide biological plausibility and data needed to increase confidence in linking invitro-based measurements to chemical effects on adverse human health outcomes.
Collapse
Affiliation(s)
- Michelle M Angrish
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Charlene A McQueen
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Elaine Cohen-Hubal
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Maribel Bruno
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Yue Ge
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Brian N Chorley
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| |
Collapse
|
76
|
Transcriptional control of intestinal cholesterol absorption, adipose energy expenditure and lipid handling by Sortilin. Sci Rep 2018; 8:9006. [PMID: 29899496 PMCID: PMC5998044 DOI: 10.1038/s41598-018-27416-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/31/2018] [Indexed: 12/11/2022] Open
Abstract
The sorting receptor Sortilin functions in the regulation of glucose and lipid metabolism. Dysfunctional lipid uptake, storage, and metabolism contribute to several major human diseases including atherosclerosis and obesity. Sortilin associates with cardiovascular disease; however, the role of Sortilin in adipose tissue and lipid metabolism remains unclear. Here we show that in the low-density lipoprotein receptor-deficient (Ldlr−/−) atherosclerosis model, Sortilin deficiency (Sort1−/−) in female mice suppresses Niemann-Pick type C1-Like 1 (Npc1l1) mRNA levels, reduces body and white adipose tissue weight, and improves brown adipose tissue function partially via transcriptional downregulation of Krüppel-like factor 4 and Liver X receptor. Female Ldlr−/−Sort1−/− mice on a high-fat/cholesterol diet had elevated plasma Fibroblast growth factor 21 and Adiponectin, an adipokine that when reduced is associated with obesity and cardiovascular disease-related factors. Additionally, Sort1 deficiency suppressed cholesterol absorption in both female mice ex vivo intestinal tissue and human colon Caco-2 cells in a similar manner to treatment with the NPC1L1 inhibitor ezetimibe. Together our findings support a novel role of Sortilin in energy regulation and lipid homeostasis in female mice, which may be a potential therapeutic target for obesity and cardiovascular disease.
Collapse
|
77
|
Rational application of macrophage-specific LXR agonists avoids the pitfalls of SREBP-induced lipogenesis. Proc Natl Acad Sci U S A 2018; 115:5051-5053. [PMID: 29712848 DOI: 10.1073/pnas.1805128115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
78
|
Liver X receptor β in the hippocampus: A potential novel target for the treatment of major depressive disorder? Neuropharmacology 2018; 135:514-528. [PMID: 29654801 DOI: 10.1016/j.neuropharm.2018.04.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 12/13/2022]
Abstract
Liver X receptors (LXRs), including LXRα and LXRβ isoforms, have been implicated in multiple physiological functions including promoting neurogenesis, improving synaptic plasticity, preventing neurodegeneration, inhibiting inflammation as well as regulating cholesterol metabolism. However, a potential role of LXRs in the treatment of major depressive disorder (MDD) has never been investigated previously. Our present results demonstrated that levels of hippocampal LXRβ but not LXRα were down-regulated in rats exposed to chronic unpredictable stress (CUS) and were negatively correlated with the severity of CUS-induced depressive-like behaviors. Furthermore, rats with LXRβ knockdown by short hairpin RNA (shRNA) in hippocampus displayed depressive-like behaviors and impaired hippocampal neurogenesis similar to those observed after CUS exposure. Conversely, LXRs activation by GW3965 (GW), a synthetic dual agonist for both LXRα and LXRβ isoforms, could improve depression-like behaviors and reverse the impaired hippocampal neurogenesis in rats exposed to CUS. LXRβ knockdown by shRNA completely abrogated the antidepressant and hippocampal neurogenesis-promoting effects of GW, suggesting that LXRβ isoform mediated the antidepressant and hippocampal neurogenesis-promoting effects of the LXRα/β dual agonist. However, ablation of hippocampal neurogenesis with x-irradiation only partly but not completely abolished the antidepressant effects of GW in the behavioral tests, implying that the antidepressant effects mediated by LXRβ isoform are likely through both neurogenesis-dependent and -independent pathways. Thus, our findings suggest that LXRβ activation may represent a potential novel target for the treatment of MDD and also provide a novel insight into the underlying mechanisms of MDD.
Collapse
|
79
|
Cell-specific discrimination of desmosterol and desmosterol mimetics confers selective regulation of LXR and SREBP in macrophages. Proc Natl Acad Sci U S A 2018; 115:E4680-E4689. [PMID: 29632203 PMCID: PMC5960280 DOI: 10.1073/pnas.1714518115] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The beneficial effects of LXR-pathway activation have long been appreciated, but clinical application of synthetic LXR ligands has been limited by coactivation of SREBP1c and consequent hypertriglyceridemia. Natural LXR ligands such as desmosterol do not promote hypertriglyceridemia because of coordinate down-regulation of the SREBP pathway. Here we demonstrate that synthetic desmosterol mimetics activate LXR in macrophages both in vitro and in vivo while suppressing SREBP target genes. Unexpectedly, desmosterol and synthetic desmosterol mimetics have almost no effect on LXR activity in hepatocytes in comparison with conventional synthetic LXR ligands. These findings reveal cell-specific differences in LXR responses to natural and synthetic ligands in macrophages and liver cells that provide a conceptually new basis for future drug development. Activation of liver X receptors (LXRs) with synthetic agonists promotes reverse cholesterol transport and protects against atherosclerosis in mouse models. Most synthetic LXR agonists also cause marked hypertriglyceridemia by inducing the expression of sterol regulatory element-binding protein (SREBP)1c and downstream genes that drive fatty acid biosynthesis. Recent studies demonstrated that desmosterol, an intermediate in the cholesterol biosynthetic pathway that suppresses SREBP processing by binding to SCAP, also binds and activates LXRs and is the most abundant LXR ligand in macrophage foam cells. Here we explore the potential of increasing endogenous desmosterol production or mimicking its activity as a means of inducing LXR activity while simultaneously suppressing SREBP1c-induced hypertriglyceridemia. Unexpectedly, while desmosterol strongly activated LXR target genes and suppressed SREBP pathways in mouse and human macrophages, it had almost no activity in mouse or human hepatocytes in vitro. We further demonstrate that sterol-based selective modulators of LXRs have biochemical and transcriptional properties predicted of desmosterol mimetics and selectively regulate LXR function in macrophages in vitro and in vivo. These studies thereby reveal cell-specific discrimination of endogenous and synthetic regulators of LXRs and SREBPs, providing a molecular basis for dissociation of LXR functions in macrophages from those in the liver that lead to hypertriglyceridemia.
Collapse
|
80
|
Liu P, Peng L, Zhang H, Tang PMK, Zhao T, Yan M, Zhao H, Huang X, Lan H, Li P. Tangshen Formula Attenuates Diabetic Nephropathy by Promoting ABCA1-Mediated Renal Cholesterol Efflux in db/db Mice. Front Physiol 2018; 9:343. [PMID: 29681863 PMCID: PMC5897509 DOI: 10.3389/fphys.2018.00343] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/20/2018] [Indexed: 01/01/2023] Open
Abstract
The commonly prescribed Tangshen Formula (TSF) is a traditional Chinese formulation that has been shown to reduce plasma lipid metabolism and proteinuria and improve the estimated glomerular filtration rate (eGFR) in patients with diabetic kidney disease. This study investigated the underlying mechanism whereby TSF regulates renal lipid accumulation and ameliorates diabetic renal injuries in spontaneous diabetic db/db mice and in vitro in sodium palmitate (PA)-stimulated and Abca1-SiRNA-transfected mouse tubular epithelial cells (mTECs). The results revealed that TSF treatment significantly ameliorated the renal injuries by lowering urinary albumin excretion and improving renal tissue injuries in diabetic (db/db) mice. Interestingly, the treatment with TSF also resulted in decreased cholesterol levels in the renal tissues of db/db mice, which was associated with increased expression of the peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α), the Liver X receptors (LXR), and ATP-binding cassette subfamily A member 1 (ABCA1), suggesting that TSF might attenuate diabetic kidney injury via a mechanism associated with improving cholesterol efflux in the diabetic kidney. This was investigated in vitro in mTECs, and the results showed that TSF reduced the PA-stimulated cholesterol accumulation in mTECs. Mechanistically, the addition of TSF was capable of reversing PA-induced downregulation of PGC-1α, LXR, and ABCA1 expression and cholesterol accumulation in mTECs, suggesting that TSF might act the protection via the PGC-1α-LXR-ABCA1 pathway to improve the cholesterol efflux in the renal tissues of db/db mice. This was further confirmed by silencing ABCA1 to block the promotive effect of TSF on cholesterol efflux in vitro. In conclusion, TSF might ameliorate diabetic kidney injuries by promoting ABCA1-mediated renal cholesterol efflux.
Collapse
Affiliation(s)
- Peng Liu
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.,Li Ka Shing Institute of Health Sciences and Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Liang Peng
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Haojun Zhang
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Tingting Zhao
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Meihua Yan
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Hailing Zhao
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoru Huang
- Li Ka Shing Institute of Health Sciences and Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Huiyao Lan
- Li Ka Shing Institute of Health Sciences and Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Ping Li
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
81
|
Tamura S, Okada M, Kato S, Shinoda Y, Shioda N, Fukunaga K, Ui-Tei K, Ueda M. Ouabagenin is a naturally occurring LXR ligand without causing hepatic steatosis as a side effect. Sci Rep 2018; 8:2305. [PMID: 29396543 PMCID: PMC5797171 DOI: 10.1038/s41598-018-20663-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 01/23/2018] [Indexed: 12/23/2022] Open
Abstract
Ouabagenin (OBG) is an aglycone of the cardiotonic steroid ouabain and until now was considered a biologically inactive biosynthetic precursor. Herein, we revealed that OBG functions as a novel class of ligand for the liver X receptor (LXR). Luciferase reporter assays and in silico docking studies suggested that OBG has LXR-selective agonistic activity. In addition, OBG repressed the expression of epithelial sodium channel (ENaC), a LXR target gene, without causing hepatic steatosis, a typical side effect of conventional LXR ligands. This remarkable biological activity can be attributed to a unique mode of action; the LXR agonist activity mainly proceeds through the LXRβ subtype without affecting LXRα, unlike conventional LXR ligands. Thus, OBG is a novel class of LXR ligand that does not cause severe side effects, with potential for use as an antihypertensive diuretic or a tool compound for exploring LXR subtype-specific biological functions.
Collapse
Affiliation(s)
- Satoru Tamura
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Miyagi, 980-8578, Japan.,School of Pharmacy, Iwate Medical University, Shiwa-gun, Iwate, 028-3694, Japan
| | - Maiko Okada
- Institute of Medical Science, St. Marianna University Graduate School of Medicine, Kawasaki, Kanagawa, 970-8551, Japan.,Genome regulation and Molecular Pharmacogenomics, School of Bioscience and Biotechnology, Tokyo University of Technology, Hachioji, Tokyo, 192-0982, Japan
| | - Shigeaki Kato
- Iwaki Meisei University, Iwaki, Fukushima, 970-8551, Japan.,Research Institute of Innovative Medicine, Tokiwa Foundation, Iwaki, Fukushima, 972-8322, Japan
| | - Yasuharu Shinoda
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Norifumi Shioda
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Kohji Fukunaga
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Kumiko Ui-Tei
- Graduate School of Science, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Minoru Ueda
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Miyagi, 980-8578, Japan.
| |
Collapse
|
82
|
Liu Y, Wei Z, Ma X, Yang X, Chen Y, Sun L, Ma C, Miao QR, Hajjar DP, Han J, Duan Y. 25-Hydroxycholesterol activates the expression of cholesterol 25-hydroxylase in an LXR-dependent mechanism. J Lipid Res 2018; 59:439-451. [PMID: 29298812 DOI: 10.1194/jlr.m080440] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/06/2017] [Indexed: 12/13/2022] Open
Abstract
Cholesterol 25-hydroxylase (CH25H) catalyzes the production of 25-hydroxycholesterol (25-HC), an oxysterol that can play an important role in different biological processes. However, the mechanisms regulating CH25H expression have not been fully elucidated. In this study, we determined that CH25H is highly expressed in mouse liver and peritoneal macrophages. We identified several liver X receptor (LXR) response elements (LXREs) in the human CH25H promoter. In HepG2 cells, activation of LXR by 25-HC or other oxysterols and synthetic ligands [T0901317 (T317) and GW3965] induced CH25H protein expression, which was associated with increased CH25H mRNA expression. 25-HC or T317 activated CH25H transcription in an LXRE-dependent manner. Thus, high-expressing LXRα or LXRβ activated CH25H expression, and the activation was further enhanced by LXR ligands. In contrast, inhibition of LXRα/β expression attenuated 25-HC or T317-induced CH25H expression. Deficiency of interferon γ expression reduced, but did not block, LXR ligand-induced hepatic CH25H expression. Activation of LXR also substantially induced macrophage CH25H expression. In vivo, administration of GW3965 to mice increased CH25H expression in both liver and peritoneal macrophages. Taken together, our study demonstrates that 25-HC can activate CH25H expression in an LXR-dependent manner, which may be an important mechanism to exert the biological actions of 25-HC.
Collapse
Affiliation(s)
- Ying Liu
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China.,Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China
| | - Zhuo Wei
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xingzhe Ma
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Xiaoxiao Yang
- Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China
| | - Yuanli Chen
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Lei Sun
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Chuanrui Ma
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Qing R Miao
- Departments of Surgery and Pathology, Medical College of Wisconsin, Milwaukee, WI
| | - David P Hajjar
- Department of Pathology, Weill Cornell Medical College of Cornell University, New York, NY
| | - Jihong Han
- Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China .,College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yajun Duan
- Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China .,College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
83
|
Iqbal F, Baker WS, Khan MI, Thukuntla S, McKinney KH, Abate N, Tuvdendorj D. Current and future therapies for addressing the effects of inflammation on HDL cholesterol metabolism. Br J Pharmacol 2017; 174:3986-4006. [PMID: 28326542 PMCID: PMC5660004 DOI: 10.1111/bph.13743] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/16/2017] [Accepted: 02/02/2017] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Inflammatory processes arising from metabolic abnormalities are known to precipitate the development of CVD. Several metabolic and inflammatory markers have been proposed for predicting the progression of CVD, including high density lipoprotein cholesterol (HDL-C). For ~50 years, HDL-C has been considered as the atheroprotective 'good' cholesterol because of its strong inverse association with the progression of CVD. Thus, interventions to increase the concentration of HDL-C have been successfully tested in animals; however, clinical trials were unable to confirm the cardiovascular benefits of pharmaceutical interventions aimed at increasing HDL-C levels. Based on these data, the significance of HDL-C in the prevention of CVD has been called into question. Fundamental in vitro and animal studies suggest that HDL-C functionality, rather than HDL-C concentration, is important for the CVD-preventive qualities of HDL-C. Our current review of the literature positively demonstrates the negative impact of systemic and tissue (i.e. adipose tissue) inflammation in the healthy metabolism and function of HDL-C. Our survey indicates that HDL-C may be a good marker of adipose tissue health, independently of its atheroprotective associations. We summarize the current findings on the use of anti-inflammatory drugs to either prevent HDL-C clearance or improve the function and production of HDL-C particles. It is evident that the therapeutic agents currently available may not provide the optimal strategy for altering HDL-C metabolism and function, and thus, further research is required to supplement this mechanistic approach for preventing the progression of CVD. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Fatima Iqbal
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Wendy S Baker
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Madiha I Khan
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Shwetha Thukuntla
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Kevin H McKinney
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Nicola Abate
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Demidmaa Tuvdendorj
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| |
Collapse
|
84
|
Kim SJ, Kim JE, Kim YW, Kim JY, Park SY. Nutritional regulation of renal lipogenic factor expression in mice: comparison to regulation in the liver and skeletal muscle. Am J Physiol Renal Physiol 2017; 313:F887-F898. [DOI: 10.1152/ajprenal.00594.2016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 05/08/2017] [Accepted: 05/18/2017] [Indexed: 12/16/2022] Open
Abstract
Regulation of lipogenesis by pathophysiological factors in the liver and skeletal muscle is well understood; however, regulation in the kidney is still unclear. To elucidate nutritional regulation of lipogenic factors in the kidney, we measured the renal expression of lipogenic transcriptional factors and enzymes during fasting and refeeding in chow-fed and high-fat-fed mice. We also examined the regulatory effect of the liver X receptor (LXR) on the expression of lipogenic factors. The renal gene expression of sterol regulatory element-binding protein (SREBP)-1c and fatty acid synthase (FAS) was reduced by fasting for 48 h and restored by refeeding, whereas the mRNA levels of forkhead box O (FOXO)1/3 were increased by fasting and restored by refeeding. Accordingly, protein levels of SREBP-1, FAS, and phosphorylated FOXO1/3 were reduced by fasting and restored by refeeding. The patterns of lipogenic factors expression in the kidney were similar to those in the liver and skeletal muscle. However, this phasic regulation of renal lipogenic gene expression was blunted in diet-induced obese mice. LXR agonist TO901317 increased the lipogenic gene expression and the protein levels of SREBP-1 precursor and FAS but not nuclear SREBP-1. Moreover, increases in insulin-induced gene mRNA and nuclear carbohydrate-responsive element binding protein (ChREBP) levels were observed in the TO901317-treated mice. These results suggest that the kidney shows flexible suppression and restoration of lipogenic factors following fasting and refeeding in lean mice, but this is blunted in obese mice. LXR is involved in the renal expression of lipogenic enzymes, and ChREBP may mediate the response.
Collapse
Affiliation(s)
- Suk-Jeong Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea; and
- Smart-Aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Ji-Eun Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea; and
- Smart-Aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Yong-Woon Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea; and
| | - Jong-Yeon Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea; and
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea; and
- Smart-Aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| |
Collapse
|
85
|
Yu M, Amengual J, Menon A, Kamaly N, Zhou F, Xu X, Saw PE, Lee SJ, Si K, Ortega CA, Choi WI, Lee IH, Bdour Y, Shi J, Mahmoudi M, Jon S, Fisher EA, Farokhzad OC. Targeted Nanotherapeutics Encapsulating Liver X Receptor Agonist GW3965 Enhance Antiatherogenic Effects without Adverse Effects on Hepatic Lipid Metabolism in Ldlr -/- Mice. Adv Healthc Mater 2017; 6:10.1002/adhm.201700313. [PMID: 28730752 PMCID: PMC5656530 DOI: 10.1002/adhm.201700313] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/27/2017] [Indexed: 11/11/2022]
Abstract
The pharmacological manipulation of liver X receptors (LXRs) has been an attractive therapeutic strategy for atherosclerosis treatment as they control reverse cholesterol transport and inflammatory response. This study presents the development and efficacy of nanoparticles (NPs) incorporating the synthetic LXR agonist GW3965 (GW) in targeting atherosclerotic lesions. Collagen IV (Col IV) targeting ligands are employed to functionalize the NPs to improve targeting to the atherosclerotic plaque, and formulation parameters such as the length of the polyethylene glycol (PEG) coating molecules are systematically optimized. In vitro studies indicate that the GW-encapsulated NPs upregulate the LXR target genes and downregulate proinflammatory mediator in macrophages. The Col IV-targeted NPs encapsulating GW (Col IV-GW-NPs) successfully reaches atherosclerotic lesions when administered for 5 weeks to mice with preexisting lesions, substantially reducing macrophage content (≈30%) compared to the PBS group, which is with greater efficacy versus nontargeting NPs encapsulating GW (GW-NPs) (≈18%). In addition, mice administered the Col IV-GW-NPs do not demonstrate increased hepatic lipid biosynthesis or hyperlipidemia during the treatment period, unlike mice injected with the free GW. These findings suggest a new form of LXR-based therapeutics capable of enhanced delivery of the LXR agonist to atherosclerotic lesions without altering hepatic lipid metabolism.
Collapse
Affiliation(s)
- Mikyung Yu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jaume Amengual
- Division of Cardiology, Department of Medicine, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Arjun Menon
- Division of Cardiology, Department of Medicine, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Nazila Kamaly
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Micro and Nanotechnology, Technical University of Denmark, DTU Nanotech, 2800 Kgs. Lyngby, Denmark
| | - Felix Zhou
- Division of Cardiology, Department of Medicine, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Xiaoding Xu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Phei Er Saw
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Seung-Joo Lee
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - Kevin Si
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Carleena Angelica Ortega
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Won Il Choi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Center for Convergence Bioceramic Materials, Convergence R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-guCheongju, Chungbuk, 28160, Republic of Korea
| | - In-Hyun Lee
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yazan Bdour
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Morteza Mahmoudi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Sangyong Jon
- KAIST Institute for the BioCentury, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- College of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
86
|
Gu M, Zhang Y, Liu C, Wang D, Feng L, Fan S, Yang B, Tong Q, Ji G, Huang C. Morin, a novel liver X receptor α/β dual antagonist, has potent therapeutic efficacy for nonalcoholic fatty liver diseases. Br J Pharmacol 2017. [PMID: 28646531 DOI: 10.1111/bph.13933] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Morin is a natural occurring flavonoid in many dietary plants and has a wide range of beneficial effects on metabolism; however, the mechanism underlying its action remains elusive. EXPERIMENTAL APPROACH A reporter assay and the time-resolved FRET assay were used to identify morin as a dual antagonist of liver X receptor (LXR)-α and -β. Morin (100 mg. 100 g-1 diet) was administered to high-fat diet-induced obese or LXRβ-/- mice. The pharmacological effects and mechanism of action of morin were evaluated by Western blot and RT-PCR analyses. KEY RESULTS From the in vitro assays, morin was shown to be a dual antagonist of LXRα and LXRβ. In vivo, morin blunted the development of liver hepatic steatosis, reduced body weight gains, lowered triglyceride levels and improved glucose and insulin tolerance in mice fed a high-fat diet. Mechanistically, morin inhibited 3T3-L1 adipocyte differentiation and lipid formation in human hepatic HepG2 cells and suppressed the mRNA expression of genes downstream of LXR. Consistently, the effects of morin on metabolic disorders were attenuated in LXRβ-/- mice. CONCLUSION AND IMPLICATIONS Our data reveal that morin is a dual antagonist of LXRα and LXRβ and suggest that morin may alleviate hepatic steatosis and other associated metabolic disorders via the suppression of LXR signalling and, therefore, shows promise as a novel therapy or nutraceutical for nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Ming Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Chuhe Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dongshan Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Brown Foundation Institute of Molecular Medicine and Program in Neuroscience, Graduate School of Biological Sciences, University of Texas McGovern Medical School, Houston, TX, USA
| | - Baican Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine and Program in Neuroscience, Graduate School of Biological Sciences, University of Texas McGovern Medical School, Houston, TX, USA
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
87
|
Peng L, Zhao XS, Peng D. Anti-obesity Effect of a Novel Potent Synthetic Steroidal Liver X Receptor α (LXRα)-selective Agonist in Male ob/ob C57BL/6 Mice. INT J PHARMACOL 2017. [DOI: 10.3923/ijp.2017.636.642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
88
|
Ducheix S, Montagner A, Polizzi A, Lasserre F, Régnier M, Marmugi A, Benhamed F, Bertrand-Michel J, Mselli-Lakhal L, Loiseau N, Martin PG, Lobaccaro JM, Ferrier L, Postic C, Guillou H. Dietary oleic acid regulates hepatic lipogenesis through a liver X receptor-dependent signaling. PLoS One 2017; 12:e0181393. [PMID: 28732092 PMCID: PMC5521785 DOI: 10.1371/journal.pone.0181393] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 06/22/2017] [Indexed: 12/13/2022] Open
Abstract
Olive oil consumption is beneficial for health as it is associated with a decreased prevalence of cancer and cardiovascular diseases. Oleic acid is, by far, the most abundant component of olive oil. Since it can be made through de novo synthesis in animals, it is not an essential fatty acid. While it has become clear that dietary oleic acid regulates many biological processes, the signaling pathway involved in these regulations remains poorly defined. In this work we tested the impact of an oleic acid-rich diet on hepatic gene expression. We were particularly interested in addressing the contribution of Liver X Receptors (LXR) in the control of genes involved in hepatic lipogenesis, an essential process in whole body energy homeostasis. We used wild-type mice and transgenic mice deficient for both α and β Liver X Receptor isoforms (LXR-/-) fed a control or an oleate enriched diet. We observed that hepatic-lipid accumulation was enhanced as well as the expression of lipogenic genes in the liver of wild-type mice fed the oleate enriched diet. In contrast, none of these changes occurred in the liver of LXR-/- mice. Strikingly, oleate-rich diet reduced cholesterolemia in wild-type mice and induced signs of liver inflammation and damage in LXR-/- mice but not in wild-type mice. This work suggests that dietary oleic acid reduces cholesterolemia while promoting LXR-dependent hepatic lipogenesis without detrimental effects to the liver.
Collapse
Affiliation(s)
- Simon Ducheix
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Alexandra Montagner
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Arnaud Polizzi
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Frédéric Lasserre
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Marion Régnier
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Alice Marmugi
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Fadila Benhamed
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France
| | | | - Laila Mselli-Lakhal
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Nicolas Loiseau
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Pascal G Martin
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Jean-Marc Lobaccaro
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, Clermont-Ferrand, France.,CNRS, UMR 6293, GReD, Aubière, France.,INSERM, U1103, GReD, Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Laurent Ferrier
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Catherine Postic
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Hervé Guillou
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| |
Collapse
|
89
|
Benítez-Santana T, Hugo SE, Schlegel A. Role of Intestinal LXRα in Regulating Post-prandial Lipid Excursion and Diet-Induced Hypercholesterolemia and Hepatic Lipid Accumulation. Front Physiol 2017; 8:280. [PMID: 28536535 PMCID: PMC5422522 DOI: 10.3389/fphys.2017.00280] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022] Open
Abstract
Post-prandial hyperlipidemia has emerged as a cardiovascular risk factor with limited therapeutic options. The Liver X receptors (Lxrs) are nuclear hormone receptors that regulate cholesterol elimination. Knowledge of their role in regulating the absorption and handling of dietary fats is incomplete. The purpose of this study was to determine the role of intestinal Lxrα in post-prandial intestinal lipid transport. Using Lxrα knockout (nr1h3−/−) and intestine-limited Lxrα over-expressing [Tg(fabp2a:EGFP-nr1h3)] zebrafish strains, we measured post-prandial lipid excursion with live imaging in larvae and physiological methods in adults. We also conducted a long-term high-cholesterol dietary challenge in adults to examine the chronic effect of modulating nr1h3 gene dose on the development of hypercholesterolemia and hepatic lipid accumulation. Over-expression of Lxrα in the intestine delays the transport of ingested lipids in larvae, while deletion of Lxrα increases the rate of lipid transport. Pre-treating wildtype larvae with the liver-sparing Lxr agonist hyodeoxycholic acid also delayed the rate of intestinal lipid transport in larvae. In adult males, deletion of Lxrα accelerates intestinal transport of ingested lipids. Adult females showed higher plasma Lipoprotein lipase (Lpl) activity compared to males, and lower post-gavage blood triacylglycerol (TAG) excursion. Despite the sexually dimorphic effect on acute intestinal lipid handling, Tg(fabp2a:EGFP-nr1h3) adults of both sexes are protected from high cholesterol diet (HCD)-induced hepatic lipid accumulation, while nr1h3−/− mutants are sensitive to the effects of HCD challenge. These data indicate that intestinal Lxr activity dampens the pace of intestinal lipid transport cell-autonomously. Selective activation of intestinal Lxrα holds therapeutic promise.
Collapse
Affiliation(s)
- Tibiábin Benítez-Santana
- University of Utah Molecular Medicine Program, School of Medicine, University of UtahSalt Lake City, UT, USA.,Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, School of Medicine, University of UtahSalt Lake City, UT, USA
| | - Sarah E Hugo
- University of Utah Molecular Medicine Program, School of Medicine, University of UtahSalt Lake City, UT, USA.,Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, School of Medicine, University of UtahSalt Lake City, UT, USA
| | - Amnon Schlegel
- University of Utah Molecular Medicine Program, School of Medicine, University of UtahSalt Lake City, UT, USA.,Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, School of Medicine, University of UtahSalt Lake City, UT, USA.,Department of Biochemistry, School of Medicine, University of UtahSalt Lake City, UT, USA.,Department of Nutrition and Integrative Physiology, College of Health, University of UtahSalt Lake City, UT, USA
| |
Collapse
|
90
|
Cedó L, Santos D, Ludwig IA, Silvennoinen R, García-León A, Kaipiainen L, Carbó JM, Valledor AF, Gylling H, Motilva MJ, Kovanen PT, Lee-Rueckert M, Blanco-Vaca F, Escolà-Gil JC. Phytosterol-mediated inhibition of intestinal cholesterol absorption in mice is independent of liver X receptor. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201700055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 01/22/2023]
Affiliation(s)
- Lídia Cedó
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau; Barcelona Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas; CIBERDEM, Hospitalet de Llobregat Spain
| | - David Santos
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau; Barcelona Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas; CIBERDEM, Hospitalet de Llobregat Spain
| | - Iziar A. Ludwig
- Food Technology Department, UTPV-XaRTA, Agrotecnio Center; University of Lleida; Lleida Spain
| | | | - Annabel García-León
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau; Barcelona Spain
- Departament de Bioquímica, Biología Molecular i Biomedicina; Universitat Autònoma de Barcelona; Barcelona Spain
| | - Leena Kaipiainen
- University of Helsinki and Helsinki University Central Hospital; Department of Internal Medicine; Helsinki Finland
| | - José M. Carbó
- Department of Cellular Biology, Physiology and Immunology; School of Biology, University of Barcelona; Barcelona Spain
| | - Annabel F. Valledor
- Department of Cellular Biology, Physiology and Immunology; School of Biology, University of Barcelona; Barcelona Spain
| | - Helena Gylling
- University of Helsinki and Helsinki University Central Hospital; Department of Internal Medicine; Helsinki Finland
| | - Maria-José Motilva
- Food Technology Department, UTPV-XaRTA, Agrotecnio Center; University of Lleida; Lleida Spain
| | | | | | - Francisco Blanco-Vaca
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau; Barcelona Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas; CIBERDEM, Hospitalet de Llobregat Spain
- Departament de Bioquímica, Biología Molecular i Biomedicina; Universitat Autònoma de Barcelona; Barcelona Spain
| | - Joan Carles Escolà-Gil
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau; Barcelona Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas; CIBERDEM, Hospitalet de Llobregat Spain
- Departament de Bioquímica, Biología Molecular i Biomedicina; Universitat Autònoma de Barcelona; Barcelona Spain
| |
Collapse
|
91
|
Im AR, Kim YH, Lee HW, Song KH. Water Extract of Dolichos lablab Attenuates Hepatic Lipid Accumulation in a Cellular Nonalcoholic Fatty Liver Disease Model. J Med Food 2017; 19:495-503. [PMID: 27152979 DOI: 10.1089/jmf.2015.3623] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease that is rising in prevalence worldwide. Therapeutic strategies for patients with NAFLD are limited by a lack of effective drugs. In this report, we show that Dolichos lablab water extract (DLL-Ex) protects against free fatty acid (FFA)-induced lipid accumulation and attenuates expression of genes involved in lipid droplet accumulation in cellular NAFLD models. The hepatoprotective effects and underlying mechanism of DLL-Ex were assessed using an in vitro cellular model in which NAFLD was simulated by inducing excessive FFA influx into hepatocytes. HepG2 cells were treated with DLL-Ex and FFAs for 24 h, after which intracellular lipid content was observed by using Nile Red and Oil Red O staining. Quantitative real-time polymerase chain reaction was used to measure expression levels of genes related to FFA-mediated cellular energy depletion. Western blotting was used to measure protein levels of phosphorylated c-Jun N-terminal kinase, AMP-activated protein kinase alpha (AMPKα), and peroxisome proliferator-activated receptor γ coactivator 1 alpha. In HepG2 cells, DLL-Ex inhibited expression of CD36, which regulates fatty acid uptake, as well as BODIPY-labeled fatty acid uptake. Additionally, DLL-Ex significantly attenuated FFA-mediated cellular energy depletion and mitochondrial membrane depolarization. Furthermore, DLL-Ex enhanced phosphorylation of AMPK, indicating that AMPK is a critical regulator of DLL-Ex-mediated inhibition of hepatic lipid accumulation, possibly through its antioxidative effect. These results demonstrate that DLL-Ex exerts potent anti-NAFLD activity, suggesting that it could be a potential adjuvant treatment for patients with NAFLD.
Collapse
Affiliation(s)
- A-Rang Im
- 1 KM Convergence Research Division, Korea Institute of Oriental Medicine , Daejeon, Korea
| | - Yun Hee Kim
- 1 KM Convergence Research Division, Korea Institute of Oriental Medicine , Daejeon, Korea
| | - Hye Won Lee
- 1 KM Convergence Research Division, Korea Institute of Oriental Medicine , Daejeon, Korea
| | - Kwang Hoon Song
- 2 Mibyeong Research Center, Korea Institute of Oriental Medicine , Daejeon, Korea.,3 University of Science and Technology , Daejeon, Korea
| |
Collapse
|
92
|
López-Lluch G. Mitochondrial activity and dynamics changes regarding metabolism in ageing and obesity. Mech Ageing Dev 2017; 162:108-121. [DOI: 10.1016/j.mad.2016.12.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/24/2016] [Accepted: 12/13/2016] [Indexed: 12/14/2022]
|
93
|
Hyodeoxycholic acid derivatives as liver X receptor α and G-protein-coupled bile acid receptor agonists. Sci Rep 2017; 7:43290. [PMID: 28233865 PMCID: PMC5324103 DOI: 10.1038/srep43290] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/23/2017] [Indexed: 12/22/2022] Open
Abstract
Bile acids are extensively investigated for their potential in the treatment of human disorders. The liver X receptors (LXRs), activated by oxysterols and by a secondary bile acid named hyodeoxycholic acid (HDCA), have been found essential in the regulation of lipid homeostasis in mammals. Unfortunately, LXRα activates lipogenic enzymes causing accumulation of lipid in the liver. In addition to LXRs, HDCA has been also shown to function as ligand for GPBAR1, a G protein coupled receptor for secondary bile acids whose activation represents a promising approach to liver steatosis. In the present study, we report a library of HDCA derivatives endowed with modulatory activity on the two receptors. The lead optimization of HDCA moiety was rationally driven by the structural information on the binding site of the two targets and results from pharmacological characterization allowed the identification of hyodeoxycholane derivatives with selective agonistic activity toward LXRα and GPBAR1 and notably to the identification of the first example of potent dual LXRα/GPBAR1 agonists. The new chemical entities might hold utility in the treatment of dyslipidemic disorders.
Collapse
|
94
|
Downing LE, Edgar D, Ellison PA, Ricketts ML. Mechanistic insight into nuclear receptor-mediated regulation of bile acid metabolism and lipid homeostasis by grape seed procyanidin extract (GSPE). Cell Biochem Funct 2017; 35:12-32. [DOI: 10.1002/cbf.3247] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/13/2016] [Accepted: 12/16/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Laura E. Downing
- Department of Agriculture, Nutrition and Veterinary Sciences; University of Nevada Reno; Reno Nevada USA
| | - Daniel Edgar
- Department of Biochemistry and Molecular Biology; University of Nevada Reno; Reno Nevada USA
| | - Patricia A. Ellison
- Department of Biochemistry and Molecular Biology; University of Nevada Reno; Reno Nevada USA
| | - Marie-Louise Ricketts
- Department of Agriculture, Nutrition and Veterinary Sciences; University of Nevada Reno; Reno Nevada USA
| |
Collapse
|
95
|
Cui X, Chopp M, Zhang Z, Li R, Zacharek A, Landschoot-Ward J, Venkat P, Chen J. ABCA1/ApoE/HDL Pathway Mediates GW3965-Induced Neurorestoration After Stroke. Stroke 2016; 48:459-467. [PMID: 28028143 DOI: 10.1161/strokeaha.116.015592] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/09/2016] [Accepted: 11/23/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND PURPOSE ATP-binding cassette transporter A1 (ABCA1) is a major reverse cholesterol transporter and plays critical role in the formation of brain high-density lipoprotein (HDL) cholesterol. Apolipoprotein E (ApoE) is the most abundant apolipoprotein and transports cholesterol into cells in brain. ABCA1 and ApoE are upregulated by liver-X receptors. Activation of liver-X receptors has neurorestorative benefit for stroke. The current study investigates whether ABCA1/ApoE/HDL pathway mediates GW3965, a synthetic dual liver-X receptor agonist, induced neurorestoration after stroke. METHODS Middle-aged male specific brain ABCA1-deficient (ABCA1-B/-B) and floxed-control (ABCA1fl/fl) mice were subjected to distal middle-cerebral artery occlusion (dMCAo) and gavaged with saline or GW3965 (10 mg/kg) or intracerebral infusion of artificial cerebrospinal fluid or human plasma HDL3 in ABCA1-B/-B stroke mice, starting 24 hours after dMCAo and daily until euthanization 14 days after dMCAo. RESULTS No differences in the blood level of total cholesterol and triglyceride and lesion volume were found among the groups. Compared with ABCA1fl/fl ischemic mice, ABCA1-B/-B ischemic mice exhibited impairment functional outcome and decreased ABCA1/ApoE expression and decreased gray/white matter densities in the ischemic boundary zone 14 days after dMCAo. GW3965 treatment of ABCA1fl/fl ischemic mice led to increased brain ABCA1/ApoE expression, concomitantly to increased blood HDL, gray/white matter densities and oligodendrocyte progenitor cell numbers in the ischemic boundary zone, as well as improved functional outcome 14 days after dMCAo. GW3965 treatment had negligible beneficial effects in ABCA1-B/-B ischemic mice. However, intracerebral infusion of human plasma HDL3 significantly attenuated ABCA1-B/-B-induced deficits. In vitro, GW3965 treatment (5 μM) increased ABCA1/synaptophysin level and neurite/axonal outgrowth in primary cortical neurons derived from ABCA1fl/fl embryos, but not in neurons derived from ABCA1-B/-B embryos. HDL treatment (80 μg/mL) attenuated the reduction of neurite/axonal outgrowth in neurons derived from ABCA1-B/-B embryos. CONCLUSIONS ABCA1/ApoE/HDL pathway, at least partially, contributes to GW3965-induced neurorestoration after stroke.
Collapse
Affiliation(s)
- Xu Cui
- From the Department of Neurology, Henry Ford Health System, Detroit, MI (X.C., M.C., Z.Z., R.L., A.Z., J.L.-W., P.V., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.).
| | - Michael Chopp
- From the Department of Neurology, Henry Ford Health System, Detroit, MI (X.C., M.C., Z.Z., R.L., A.Z., J.L.-W., P.V., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Zhenggang Zhang
- From the Department of Neurology, Henry Ford Health System, Detroit, MI (X.C., M.C., Z.Z., R.L., A.Z., J.L.-W., P.V., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Rongwen Li
- From the Department of Neurology, Henry Ford Health System, Detroit, MI (X.C., M.C., Z.Z., R.L., A.Z., J.L.-W., P.V., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Alex Zacharek
- From the Department of Neurology, Henry Ford Health System, Detroit, MI (X.C., M.C., Z.Z., R.L., A.Z., J.L.-W., P.V., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Julie Landschoot-Ward
- From the Department of Neurology, Henry Ford Health System, Detroit, MI (X.C., M.C., Z.Z., R.L., A.Z., J.L.-W., P.V., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Poornima Venkat
- From the Department of Neurology, Henry Ford Health System, Detroit, MI (X.C., M.C., Z.Z., R.L., A.Z., J.L.-W., P.V., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Jieli Chen
- From the Department of Neurology, Henry Ford Health System, Detroit, MI (X.C., M.C., Z.Z., R.L., A.Z., J.L.-W., P.V., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| |
Collapse
|
96
|
Maithilikarpagaselvi N, Sridhar MG, Swaminathan RP, Sripradha R, Badhe B. Curcumin inhibits hyperlipidemia and hepatic fat accumulation in high-fructose-fed male Wistar rats. PHARMACEUTICAL BIOLOGY 2016; 54:2857-2863. [PMID: 27241764 DOI: 10.1080/13880209.2016.1187179] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 04/29/2016] [Accepted: 05/03/2016] [Indexed: 06/05/2023]
Abstract
CONTEXT Curcumin, an active principal of Curcuma longa Linn. (Zingiberaceae), has potent antioxidant and anti-inflammatory properties. OBJECTIVES This study investigated the effects of curcumin on hyperlipidemia and hepatic steatosis in high-fructose-fed Wistar rats. MATERIALS AND METHODS Forty male Wistar rats were divided into four groups with 10 rats in each. Two groups were fed with standard rodent diet and the other two with 60% high-fructose diet for 10 weeks. Curcumin (200 mg/kg body weight) was administered along with the diets simultaneously to each of the aforementioned diet groups. After 10 weeks of experiment, blood samples were collected from tail vein. Liver, adipose and epididymal tissues were collected after sacrifice of the animals and stored for further analyses. RESULTS Administration of curcumin reduced body weight (280.6 ± 7.4 g), liver weight (2.5 ± 0.2 g/100 g BW), adipose weight (1.4 ± 0.3 g/100 g BW), plasma levels of TAG (86.1 ± 13.5 mg/dL), VLDL-C (17.2 ± 2.7 mg/dL), lipid ratios and increased HDL-C (28.4 ± 4.5 mg/dL) in fructose-fed rats. Curcumin supplementation significantly lowered TAG content and decreased the protein expression of LXR-α (43%) and SREBP1c (59%) in the liver. Furthermore, curcumin suppressed the expression of lipogenic enzymes, ACLY (95%), ACC (50%) and FAS (77%) in rats fed with high-fructose diet. No significant change was found in the expression of PPAR-α. DISCUSSION AND CONCLUSION Curcumin prevented the high-fructose induced hyperlipidemia and hepatic steatosis.
Collapse
Affiliation(s)
| | - Magadi Gopalakrishna Sridhar
- a Department of Biochemistry , Jawaharlal Institute of Postgraduate Medical Education and Research , Pondicherry , India
| | | | - Ramalingam Sripradha
- a Department of Biochemistry , Jawaharlal Institute of Postgraduate Medical Education and Research , Pondicherry , India
| | - Bhawana Badhe
- c Department of Pathology , Jawaharlal Institute of Postgraduate Medical Education and Research , Pondicherry , India
| |
Collapse
|
97
|
Panneerselvam S, Packirisamy RM, Bobby Z, Elizabeth Jacob S, Sridhar MG. Soy isoflavones ( Glycine max ) ameliorate hypertriglyceridemia and hepatic steatosis in high fat-fed ovariectomized Wistar rats (an experimental model of postmenopausal obesity). J Nutr Biochem 2016; 38:57-69. [DOI: 10.1016/j.jnutbio.2016.08.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/23/2016] [Accepted: 08/10/2016] [Indexed: 10/21/2022]
|
98
|
Kong Q, Zhang H, Zhao T, Zhang W, Yan M, Dong X, Li P. Tangshen formula attenuates hepatic steatosis by inhibiting hepatic lipogenesis and augmenting fatty acid oxidation in db/db mice. Int J Mol Med 2016; 38:1715-1726. [PMID: 27840945 PMCID: PMC5117754 DOI: 10.3892/ijmm.2016.2799] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 11/03/2016] [Indexed: 12/24/2022] Open
Abstract
Tangshen formula (TSF), a well-prescribed traditional Chinese formula, has been used in the treatment of diabetic nephropathy. However, whether TSF ameliorates dyslipidemia and liver injury associated with diabetes remains unclear. In this study, we examined the effects of TSF on lipid profiles and hepatic steatosis in db/db mice. For this purpose, 8‑week-old db/db mice were treated with TSF or saline for 12 weeks via gavage and db/m mice were used as controls. Body weight and blood glucose levels were monitored weekly and bi-weekly, respectively. Blood samples were obtained for the analysis of lipids and enzymes related to hepatic function, and liver tissues were analyzed by histology, immunohistochemistry and molecular examination. The results revealed that TSF markedly reduced body weight, liver index [liver/body weight (LW/BW)] and improved lipid profiles, hepatic function and steatosis in db/db mice. TSF induced the phosphoralation of AMP-activated protein kinase and inhibited the activity of sterol regulatory element-binding protein 1 together with the inhibition of the expression of genes involved in de novo lipogenesis (DNL) and gluconeogenesis, such as fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), stearoyl CoA desaturase 1 (SCD1), glucose-6-phosphatase (G6pc) and phosphoenolpyruvate carboxykinase 1 (Pck1). Additionally, the silent mating type information regulation 2 homolog 1 (Sirt1)/peroxisome proliferator-activated receptor α (PPARα)/malonyl-CoA decarboxylase (MLYCD) cascade was potently activated by TSF in the liver and skeletal muscle of db/db mice, which led to enhanced fatty acid oxidation. These findings demonstrated that TSF attenuated hepatic fat accumulation and steatosis in db/db mice by inhibiting lipogenesis and augmenting fatty acid oxidation.
Collapse
Affiliation(s)
- Qin Kong
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P.R. China
| | - Haojun Zhang
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Tingting Zhao
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Weiku Zhang
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Meihua Yan
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Xi Dong
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Ping Li
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P.R. China
| |
Collapse
|
99
|
Hoekstra M, Van Berkel TJ. Functionality of High-Density Lipoprotein as Antiatherosclerotic Therapeutic Target. Arterioscler Thromb Vasc Biol 2016; 36:e87-e94. [DOI: 10.1161/atvbaha.116.308262] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Menno Hoekstra
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, The Netherlands
| | - Theo J.C. Van Berkel
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, The Netherlands
| |
Collapse
|
100
|
Hu W, Zhang W, Chen Y, Rana U, Teng RJ, Duan Y, Liu Z, Zhao B, Foeckler J, Weiler H, Kallinger RE, Thomas MJ, Zhang K, Han J, Miao QR. Nogo-B receptor deficiency increases liver X receptor alpha nuclear translocation and hepatic lipogenesis through an adenosine monophosphate-activated protein kinase alpha-dependent pathway. Hepatology 2016; 64:1559-1576. [PMID: 27480224 PMCID: PMC5074877 DOI: 10.1002/hep.28747] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 06/30/2016] [Accepted: 07/14/2016] [Indexed: 01/01/2023]
Abstract
UNLABELLED Nogo-B receptor (NgBR) was identified as a specific receptor for binding Nogo-B and is essential for the stability of Niemann-Pick type C2 protein (NPC2) and NPC2-dependent cholesterol trafficking. Here, we report that NgBR expression levels decrease in the fatty liver and that NgBR plays previously unrecognized roles in regulating hepatic lipogenesis through NPC2-independent pathways. To further elucidate the pathophysiological role of NgBR in mammals, we generated NgBR liver-specific knockout mice and investigated the roles of NgBR in hepatic lipid homeostasis. The results showed that NgBR knockout in mouse liver did not decrease NPC2 levels or increase NPC2-dependent intracellular cholesterol levels. However, NgBR deficiency still resulted in remarkable cellular lipid accumulation that was associated with increased free fatty acids and triglycerides in hepatocytes in vitro and in mouse livers in vivo. Mechanistically, NgBR deficiency specifically promotes the nuclear translocation of the liver X receptor alpha (LXRα) and increases the expression of LXRα-targeted lipogenic genes. LXRα knockout attenuates the accumulation of free fatty acids and triglycerides caused by NgBR deficiency. In addition, we elucidated the mechanisms by which NgBR bridges the adenosine monophosphate-activated protein kinase alpha signaling pathway with LXRα nuclear translocation and LXRα-mediated lipogenesis. CONCLUSION NgBR is a specific negative regulator for LXRα-dependent hepatic lipogenesis. Loss of NgBR may be a potential trigger for inducing hepatic steatosis. (Hepatology 2016;64:1559-1576).
Collapse
Affiliation(s)
- Wenquan Hu
- Department of Surgery and Pathology, Medical College of Wisconsin
,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Wenwen Zhang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Yuanli Chen
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Ujala Rana
- Department of Surgery and Pathology, Medical College of Wisconsin
| | - Ru-jeng Teng
- Department of Pediatrics, Children’s Research Institute, Medical College of Wisconsin
| | - Yajun Duan
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Zhong Liu
- Department of Surgery and Pathology, Medical College of Wisconsin
| | - Baofeng Zhao
- Department of Surgery and Pathology, Medical College of Wisconsin
| | | | | | | | - Michael J. Thomas
- Department of Pharmacology and Toxicology, Medical College of Wisconsin
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Jihong Han
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China. .,College of Biomedical Engineering, Hefei University of Technology, Hefei, China.
| | - Qing Robert Miao
- Departments of Surgery and Pathology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|