1
|
Barnhart S, Shimizu-Albergine M, Kedar E, Kothari V, Shao B, Krueger M, Hsu CC, Tang J, Kanter JE, Kramer F, Djukovic D, Pascua V, Loo YM, Colonna L, Van den Bogaerde SJ, An J, Gale M, Reue K, Fisher EA, Gharib SA, Elkon KB, Bornfeldt KE. Type I IFN induces long-chain acyl-CoA synthetase 1 to generate a phosphatidic acid reservoir for lipotoxic saturated fatty acids. J Lipid Res 2024:100730. [PMID: 39675509 DOI: 10.1016/j.jlr.2024.100730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/22/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024] Open
Abstract
Long-chain acyl-CoA synthetase 1 (ACSL1) catalyzes the conversion of long-chain fatty acids to acyl-CoAs. ACSL1 is required for β-oxidation in tissues that rely on fatty acids as fuel, but no consensus exists on why ACSL1 is induced by inflammatory mediators in immune cells. We used a comprehensive and unbiased approach to investigate the role of ACSL1 induction by interferon type I (IFN-I) in myeloid cells in vitro and in a mouse model of IFN-I overproduction. Our results show that IFN-I induces ACSL1 in macrophages via its interferon-α/β receptor, and consequently that expression of ACSL1 is increased in myeloid cells from individuals with systemic lupus erythematosus (SLE), an autoimmune condition characterized by increased IFN production. Taking advantage of a myeloid cell-targeted ACSL1-deficient mouse model and a series of lipidomics, proteomics, metabolomics and functional analyses, we show that IFN-I leverages induction of ACSL1 to increase accumulation of fully saturated phosphatidic acid species in macrophages. Conversely, ACSL1 induction is not needed for IFN-I's ability to induce the prototypical IFN-stimulated protein signature or to suppress proliferation or macrophage metabolism. Loss of ACSL1 in IFN-I stimulated myeloid cells enhances apoptosis and secondary necrosis in vitro, especially in the presence of increased saturated fatty acid load, and in a mouse model of atherosclerosis associated with IFN overproduction, resulting in larger lesion necrotic cores. We propose that ACSL1 induction is a mechanism used by IFN-I to increase phosphatidic acid saturation while protecting the cells from saturated fatty acid-induced cell death.
Collapse
Affiliation(s)
- Shelley Barnhart
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Masami Shimizu-Albergine
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Eyal Kedar
- Division of Rheumatology, University of Washington, Seattle WA 98109
| | - Vishal Kothari
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Baohai Shao
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Melissa Krueger
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA 98109
| | - Cheng-Chieh Hsu
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Jingjing Tang
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Jenny E Kanter
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Farah Kramer
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Danijel Djukovic
- Northwest Metabolomics Research Center and the Department of Anesthesiology and Pain Medicine, University of Washington, Seattle WA 98109
| | - Vadim Pascua
- Northwest Metabolomics Research Center and the Department of Anesthesiology and Pain Medicine, University of Washington, Seattle WA 98109
| | - Yueh-Ming Loo
- Department of Immunology, University of Washington, Seattle WA 98109
| | - Lucrezia Colonna
- Division of Rheumatology, University of Washington, Seattle WA 98109
| | | | - Jie An
- Division of Rheumatology, University of Washington, Seattle WA 98109
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle WA 98109
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016
| | - Sina A Gharib
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA 98109
| | - Keith B Elkon
- Department of Immunology, University of Washington, Seattle WA 98109
| | - Karin E Bornfeldt
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109; Department of Laboratory Medicine and Pathology, University of Washington, Seattle WA 98109.
| |
Collapse
|
2
|
Liu Q, Gu X, Liu X, Gu Y, Zhang H, Yang J, Huang Z. Long-chain fatty acids - The turning point between 'mild' and 'severe' acute pancreatitis. Heliyon 2024; 10:e31296. [PMID: 38828311 PMCID: PMC11140623 DOI: 10.1016/j.heliyon.2024.e31296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Acute pancreatitis (AP) is an inflammatory disease characterized by localized pancreatic injury and a systemic inflammatory response. Fatty acids (FAs), produced during the breakdown of triglycerides (TGs) in blood and peripancreatic fat, escalate local pancreatic inflammation to a systemic level by damaging pancreatic acinar cells (PACs) and triggering M1 macrophage polarization. This paper provides a comprehensive analysis of lipases' roles in the onset and progression of AP, as well as the effects of long-chain fatty acids (LCFAs) on the function of pancreatic acinar cells (PACs). Abnormalities in the function of PACs include Ca2+ overload, premature trypsinogen activation, protein kinase C (PKC) expression, endoplasmic reticulum (ER) stress, and mitochondrial and autophagic dysfunction. The study highlights the contribution of long-chain saturated fatty acids (LC-SFAs), especially palmitic acid (PA), to M1 macrophage polarization through the activation of the NLRP3 inflammasome and the NF-κB pathway. Furthermore, we investigated lipid lowering therapy for AP. This review establishes a theoretical foundation for pro-inflammatory mechanisms associated with FAs in AP and facilitating drug development.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310058, China
- Hangzhou Hospital & Institute of Digestive Diseases, Hangzhou, Zhejiang 310006, China
| | - Xinyi Gu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310003, China
| | - Xiaodie Liu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310003, China
| | - Ye Gu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310058, China
| | - Hongchen Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310058, China
| | - Jianfeng Yang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310058, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310003, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310058, China
- Hangzhou Hospital & Institute of Digestive Diseases, Hangzhou, Zhejiang 310006, China
| | - Zhicheng Huang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310058, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310003, China
| |
Collapse
|
3
|
Kruchinina MV, Svetlova IO, Osipenko MF, Abaltusova NV, Gromov AA, Shashkov MV, Sokolova AS, Yakovina IN, Borisova AV. Fatty Acids of Erythrocyte Membranes and Blood Serum in Differential Diagnosis of Inflammatory Bowel Diseases. RUSSIAN JOURNAL OF GASTROENTEROLOGY, HEPATOLOGY, COLOPROCTOLOGY 2022; 32:50-67. [DOI: 10.22416/1382-4376-2022-32-4-50-67] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Aim: to study fatty acid levels in erythrocyte membranes (RBC) and blood serum (BS) in patients with inflammatory bowel diseases (IBDs) to develop differential diagnostic models including fatty acids as biomarkers to distinguish between nosological entities of IBDs (ulcerative colitis — UC, Crohn's disease — CD, unclassified colitis — UCC).Materials and methods. We examined 110 patients (mean age 37,7 ± 12,1 years) with IBDs and 53 healthy patients in control group (43,3 ± 11,7 years). The IBDs group included 50 patients with UC, 41 patients with CD, 19 patients with UCC. An exacerbation of the disease was revealed in 42 patients (84 %) with UC, 34 patients with CD (82.9 %) and 11 people with UCC (57.9 %). The study of fatty acids (FA) composition of RBC membranes and BS was carried out using GC/MS system based on three Agilent 7000B quadrupoles (USA).Results. The most significant for distinguishing active UC from CD exacerbation were serum levels of elaidin (p = 0.0006); docosatetraenoic (n-6) (p = 0.004); docodienic (n-6) (p = 0.009); omega-3/omega-6 ratio (p = 0.02); docosapentaenoic (n-3) (p = 0.03); the sum of eicosapentaenoic and docosahexaenoic (p = 0.03), as well as the content of RBC lauric FA (p = 0.04) (AUC — 0.89, sensitivity — 0.91, specificity — 0.89, diagnostic accuracy — 0.91). To distinguish active UC from the same of UCC, the following serum FA were found to be significant: alpha-linolenic; saturated (pentadecanoic, palmitic, stearic, arachidic); monounsaturated (palmitoleic, oleic); omega-6 (hexadecadienic, arachidonic) (p = 0.00000011—0.03300000) (AUC — 0.995, sensitivity — 0.98, specificity — 0.96, diagnostic accuracy — 0.97). The most significant in distinguishing patients with active CD from UCC exacerbation were levels of the following FA: alpha-linolenic; palmitoleic; oleic; the amount of saturated fatty acids (SFA); total unsaturated fatty acids (UFA); stearic; monounsaturated fatty acids (MUFA) amount; SFA/UFA; SFA/PUFA (polyunsaturated fatty acids); linoleic; total PUFA n6; lauric; arachidic acid (p = 0.0000000017–0.030000000) (AUC — 0.914, sensitivity — 0.90, specificity — 0.87, diagnostic accuracy — 0.91).Conclusion. The study of FA levels in groups with different nosological forms of IBDs using complex statistical analysis, including machine learning methods, made it possible to create diagnostic models that differentiate CD, UC and UCC in the acute stage with high accuracy. The proposed approach is promising for the purposes of differential diagnosis of nosological forms of IBDs.
Collapse
Affiliation(s)
- M. V. Kruchinina
- Research Institute of Internal and Preventive Medicine — Branch of the Institute of Cytology and Genetics, Siberian branch of Russian Academy of Sciences; Novosibirsk State Medical University
| | - I. O. Svetlova
- Research Institute of Internal and Preventive Medicine — Branch of the Institute of Cytology and Genetics, Siberian branch of Russian Academy of Sciences; Novosibirsk State Medical University
| | | | - N. V. Abaltusova
- Research Institute of Internal and Preventive Medicine — Branch of the Institute of Cytology and Genetics, Siberian branch of Russian Academy of Sciences
| | - A. A. Gromov
- Research Institute of Internal and Preventive Medicine — Branch of the Institute of Cytology and Genetics, Siberian branch of Russian Academy of Sciences
| | - M. V. Shashkov
- Boreskov Institute of Catalysis, Siberian branch of Russian Academy of Sciences
| | - A. S. Sokolova
- Novosibirsk Institute of Organic Chemistry, Siberian branch of Russian Academy of Sciences
| | | | - A. V. Borisova
- Research Institute of Internal and Preventive Medicine — Branch of the Institute of Cytology and Genetics, Siberian branch of Russian Academy of Sciences
| |
Collapse
|
4
|
Szczepańska P, Hapeta P, Lazar Z. Advances in production of high-value lipids by oleaginous yeasts. Crit Rev Biotechnol 2021; 42:1-22. [PMID: 34000935 DOI: 10.1080/07388551.2021.1922353] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The global market for high-value fatty acids production, mainly omega-3/6, hydroxy fatty-acids, waxes and their derivatives, has seen strong development in the last decade. The reason for this growth was the increasing utilization of these lipids as significant ingredients for cosmetics, food and the oleochemical industries. The large demand for these compounds resulted in a greater scientific interest in research focused on alternative sources of oil production - among which microorganisms attracted the most attention. Microbial oil production offers the possibility to engineer the pathways and store lipids enriched with the desired fatty acids. Moreover, costly chemical steps are avoided and direct commercial use of these fatty acids is available. Among all microorganisms, the oleaginous yeasts have become the most promising hosts for lipid production - their efficient lipogenesis, ability to use various (often highly affordable) carbon sources, feasible large-scale cultivations and wide range of available genetic engineering tools turns them into powerful micro-factories. This review is an in-depth description of the recent developments in the engineering of the lipid biosynthetic pathway with oleaginous yeasts. The different classes of valuable lipid compounds with their derivatives are described and their importance for human health and industry is presented. The emphasis is also placed on the optimization of culture conditions in order to improve the yield and titer of these valuable compounds. Furthermore, the important economic aspects of the current microbial oil production are discussed.
Collapse
Affiliation(s)
- Patrycja Szczepańska
- Department of Biotechnology and Food Microbiology, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Piotr Hapeta
- Department of Biotechnology and Food Microbiology, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Zbigniew Lazar
- Department of Biotechnology and Food Microbiology, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| |
Collapse
|
5
|
van Dierendonck XAMH, de la Rosa Rodriguez MA, Georgiadi A, Mattijssen F, Dijk W, van Weeghel M, Singh R, Borst JW, Stienstra R, Kersten S. HILPDA Uncouples Lipid Droplet Accumulation in Adipose Tissue Macrophages from Inflammation and Metabolic Dysregulation. Cell Rep 2021; 30:1811-1822.e6. [PMID: 32049012 DOI: 10.1016/j.celrep.2020.01.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 12/10/2019] [Accepted: 01/15/2020] [Indexed: 01/15/2023] Open
Abstract
Obesity leads to a state of chronic, low-grade inflammation that features the accumulation of lipid-laden macrophages in adipose tissue. Here, we determined the role of macrophage lipid-droplet accumulation in the development of obesity-induced adipose-tissue inflammation, using mice with myeloid-specific deficiency of the lipid-inducible HILPDA protein. HILPDA deficiency markedly reduced intracellular lipid levels and accumulation of fluorescently labeled fatty acids. Decreased lipid storage in HILPDA-deficient macrophages can be rescued by inhibition of adipose triglyceride lipase (ATGL) and is associated with increased oxidative metabolism. In diet-induced obese mice, HILPDA deficiency does not alter inflammatory and metabolic parameters, despite markedly reducing lipid accumulation in macrophages. Overall, we find that HILPDA is a lipid-inducible, physiological inhibitor of ATGL-mediated lipolysis in macrophages and uncouples lipid storage in adipose tissue macrophages from inflammation and metabolic dysregulation. Our data question the contribution of lipid droplet accumulation in adipose tissue macrophages in obesity-induced inflammation and metabolic dysregulation.
Collapse
Affiliation(s)
- Xanthe A M H van Dierendonck
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands; Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein 8, 6525 GA Nijmegen, the Netherlands
| | - Montserrat A de la Rosa Rodriguez
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Anastasia Georgiadi
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Frits Mattijssen
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Wieneke Dijk
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Rajat Singh
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer 505D, Bronx, NY 10461, USA
| | - Jan Willem Borst
- Laboratory of Biochemistry, Microspectroscopy Research Facility, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Rinke Stienstra
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands; Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein 8, 6525 GA Nijmegen, the Netherlands.
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands.
| |
Collapse
|
6
|
|
7
|
RXRs control serous macrophage neonatal expansion and identity and contribute to ovarian cancer progression. Nat Commun 2020; 11:1655. [PMID: 32246014 PMCID: PMC7125161 DOI: 10.1038/s41467-020-15371-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/28/2020] [Indexed: 12/04/2022] Open
Abstract
Tissue-resident macrophages (TRMs) populate all tissues and play key roles in homeostasis, immunity and repair. TRMs express a molecular program that is mostly shaped by tissue cues. However, TRM identity and the mechanisms that maintain TRMs in tissues remain poorly understood. We recently found that serous-cavity TRMs (LPMs) are highly enriched in RXR transcripts and RXR-response elements. Here, we show that RXRs control mouse serous-macrophage identity by regulating chromatin accessibility and the transcriptional regulation of canonical macrophage genes. RXR deficiency impairs neonatal expansion of the LPM pool and reduces the survival of adult LPMs through excess lipid accumulation. We also find that peritoneal LPMs infiltrate early ovarian tumours and that RXR deletion diminishes LPM accumulation in tumours and strongly reduces ovarian tumour progression in mice. Our study reveals that RXR signalling controls the maintenance of the serous macrophage pool and that targeting peritoneal LPMs may improve ovarian cancer outcomes. Macrophages can differentiate to perform homeostatic tissue-specific functions. Here the authors show that RXR signalling is critical for large peritoneal macrophage (LPM) expansion during neonatal life and LPM lipid metabolism and survival during adult homeostasis, and that ovarian cancer growth relies on RXR-dependent LPMs.
Collapse
|
8
|
Roelands J, Garand M, Hinchcliff E, Ma Y, Shah P, Toufiq M, Alfaki M, Hendrickx W, Boughorbel S, Rinchai D, Jazaeri A, Bedognetti D, Chaussabel D. Long-Chain Acyl-CoA Synthetase 1 Role in Sepsis and Immunity: Perspectives From a Parallel Review of Public Transcriptome Datasets and of the Literature. Front Immunol 2019; 10:2410. [PMID: 31681299 PMCID: PMC6813721 DOI: 10.3389/fimmu.2019.02410] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 09/26/2019] [Indexed: 12/21/2022] Open
Abstract
A potential role for the long-chain acyl-CoA synthetase family member 1 (ACSL1) in the immunobiology of sepsis was explored during a hands-on training workshop. Participants first assessed the robustness of the potential gap in biomedical knowledge identified via an initial screen of public transcriptome data and of the literature associated with ACSL1. Increase in ACSL1 transcript abundance during sepsis was confirmed in several independent datasets. Querying the ACSL1 literature also confirmed the absence of reports associating ACSL1 with sepsis. Inferences drawn from both the literature (via indirect associations) and public transcriptome data (via correlation) point to the likely participation of ACSL1 and ACSL4, another family member, in inflammasome activation in neutrophils during sepsis. Furthermore, available clinical data indicate that levels of ACSL1 and ACSL4 induction was significantly higher in fatal cases of sepsis. This denotes potential translational relevance and is consistent with involvement in pathways driving potentially deleterious systemic inflammation. Finally, while ACSL1 expression was induced in blood in vitro by a wide range of pathogen-derived factors as well as TNF, induction of ACSL4 appeared restricted to flagellated bacteria and pathogen-derived TLR5 agonists and IFNG. Taken together, this joint review of public literature and omics data records points to two members of the acyl-CoA synthetase family potentially playing a role in inflammasome activation in neutrophils. Translational relevance of these observations in the context of sepsis and other inflammatory conditions remain to be investigated.
Collapse
Affiliation(s)
- Jessica Roelands
- Sidra Medicine, Doha, Qatar.,Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | | | - Emily Hinchcliff
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ying Ma
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Parin Shah
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | | | | | | | - Amir Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | |
Collapse
|
9
|
Natarajan G, Perriotte-Olson C, Casey CA, Donohue TM, Talmon GA, Harris EN, Kabanov AV, Saraswathi V. Effect of nanoformulated copper/zinc superoxide dismutase on chronic ethanol-induced alterations in liver and adipose tissue. Alcohol 2019; 79:71-79. [PMID: 30611703 DOI: 10.1016/j.alcohol.2018.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/22/2018] [Accepted: 12/28/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND We previously reported that nanoformulated copper/zinc superoxide dismutase (Nano) attenuates non-alcoholic fatty liver disease and adipose tissue (AT) inflammation in obese animals. Here, we sought to determine whether Nano treatment attenuates alcohol-associated liver disease (AALD) and AT inflammation in alcohol-fed mice. METHODS We pre-treated E-47 cells (HepG2 cells that over-express CYP2E1) with native- or nano-superoxide dismutase (SOD) for 6 h, followed by treatment with ethanol and/or linoleic acid (LA), a free fatty acid. For in vivo studies, male C57BL/6 mice were fed the Lieber-DeCarli control or ethanol liquid diet for 4 weeks. The mice received Nano once every 2 days during the last 2 weeks of ethanol feeding. RESULTS Our in vitro studies revealed that Nano pretreatment reduced LA + ethanol-induced oxidative stress in E-47 cells. Our in vivo experiments showed that ethanol-fed Nano-treated mice had 22% lower hepatic triglyceride levels than mice fed ethanol alone. Nano-treated ethanol-fed mice also had 2-fold lower levels of Cd68 and similarly reduced levels of Ccl2 and Mmp12 mRNAs, than in untreated ethanol-fed mice. We also noted that ethanol feeding caused a remarkable increase in hepatic and/or plasma MCP-1 and CCR2 protein, which was blunted in ethanol + Nano-treated animals. The hepatic content of SREBP-1c, a transcription factor that promotes lipogenesis, was higher in ethanol-fed mice than controls but was attenuated in ethanol + Nano-treated animals. Further, livers of ethanol + Nano-treated mice had significantly higher levels of phosphorylated adenosine monophosphate-activated protein kinase (AMPK) than both control and ethanol-fed mice. In AT, the levels of Il6 mRNA, a hepatoprotective cytokine, and that of Arg1, a marker of anti-inflammatory macrophages, were significantly increased in ethanol + Nano-treated mice compared with control mice. CONCLUSION Our data indicate that Nano treatment attenuates ethanol-induced steatohepatitis and that this effect is associated with an apparent activation of AMPK signaling. Our data also suggest that Nano induces Arg1 and Il6 expression in AT, suggesting anti-inflammatory effects in this tissue.
Collapse
Affiliation(s)
- Gopalakrishnan Natarajan
- Department of Internal Medicine, Divisions of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, United States
| | - Curtis Perriotte-Olson
- Department of Internal Medicine, Divisions of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, United States
| | - Carol A Casey
- Department of Gastroenterology and Hepatology, University of Nebraska Medical Center, Omaha, NE, United States; VA Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Terrence M Donohue
- Department of Gastroenterology and Hepatology, University of Nebraska Medical Center, Omaha, NE, United States; VA Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Geoffrey A Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Edward N Harris
- Department of Biochemistry, University of Nebraska at Lincoln, Lincoln, NE, United States
| | - Alexander V Kabanov
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Viswanathan Saraswathi
- Department of Internal Medicine, Divisions of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, United States; VA Nebraska-Western Iowa Health Care System, Omaha, NE, United States.
| |
Collapse
|
10
|
Kang R, Kroemer G, Tang D. The tumor suppressor protein p53 and the ferroptosis network. Free Radic Biol Med 2019; 133:162-168. [PMID: 29800655 PMCID: PMC6251771 DOI: 10.1016/j.freeradbiomed.2018.05.074] [Citation(s) in RCA: 424] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/16/2018] [Accepted: 05/19/2018] [Indexed: 12/14/2022]
Abstract
Ferroptosis is a form of lipid peroxidation-induced cell death that can be regulated in many ways, from altering the activity of antioxidant enzymes to the level of transcription factors. The p53 tumor suppressor is 'the guardian of the genome' that participates in the control of cell survival and division under various stresses. Beyond its effects on apoptosis, autophagy, and cell cycle, p53 also regulates ferroptosis either through a transcriptional or posttranslational mechanism. On one hand, p53 can enhance ferroptosis by inhibiting the expression of SLC7A11 (solute carrier family 7 member 11) or by enhancing that of SAT1 (spermidine/spermine N1-acetyltransferase 1) and GLS2 (glutaminase 2). On the other hand, p53 suppresses ferroptosis through the direct inhibition of DPP4 (dipeptidyl peptidase 4) activity or by the induction of CDKN1A/p21 (cyclin dependent kinase inhibitor 1 A) expression. Here, we review recent discoveries and emerging trends in the study of the ferroptosis network and highlight the context-dependent impact of p53 on ferroptosis and oxidative stress.
Collapse
Affiliation(s)
- Rui Kang
- The Third Affiliated Hospital, Central of DAMP Biology, Guangzhou Medical University, Guangzhou, Guangdong 510150, China; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Guido Kroemer
- Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France; Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France; Université Pierre et Marie Curie, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, 94800 Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France; Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Daolin Tang
- The Third Affiliated Hospital, Central of DAMP Biology, Guangzhou Medical University, Guangzhou, Guangdong 510150, China; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
11
|
Schmölz L, Schubert M, Kirschner J, Kluge S, Galli F, Birringer M, Wallert M, Lorkowski S. Long-chain metabolites of vitamin E: Interference with lipotoxicity via lipid droplet associated protein PLIN2. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:919-927. [DOI: 10.1016/j.bbalip.2018.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 04/17/2018] [Accepted: 05/03/2018] [Indexed: 01/25/2023]
|
12
|
Zhang X, Chen W, Shao S, Xu G, Song Y, Xu C, Gao L, Hu C, Zhao J. A High-Fat Diet Rich in Saturated and Mono-Unsaturated Fatty Acids Induces Disturbance of Thyroid Lipid Profile and Hypothyroxinemia in Male Rats. Mol Nutr Food Res 2018; 62:e1700599. [PMID: 29363248 DOI: 10.1002/mnfr.201700599] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/12/2018] [Indexed: 01/09/2023]
Abstract
SCOPE Increasing evidence has shown that the disturbance of lipid metabolism might make a possible contribution to the pathogenesis of organ dysfunction, including thyroid, yet it is unknown whether excess intake of dietary fat interferes in thyroid lipid profile. We investigate the effects of dietary fat toward the thyroid lipid profile and thyroid function. METHODS AND RESULTS Male Sprague-Dawley rats are fed with high-fat diet (HFD) rich in saturated and mono-unsaturated fatty acids or chow diet for 18 weeks. LC-MS analysis of thyroid shows that total free fatty acids (FFAs) content is significantly higher in HFD rats. The concentration of highly saturated triglycerides significantly increases in HFD rats, whereas the polyunsaturated triglyceride significantly decreases, indicating the decrease in unsaturation in the HFD group. Significant increase of lysophosphatidylcholines (LPC) is observed in HFD rats. Thyroid function tests show hypothyroxinemia (total thyroxine [TT4 ] and free thyroxine [FT4 ]) in HFD rats, and elevated thyrotropin (TSH) concentration. The HFD rats also show decreased thyroid uptake of iodine. CONCLUSION Excess intake of dietary fat induces disturbance of thyroid lipid profile and hypothyroxinemia, indicating thyroid dysfunction. We speculate that it may provide a new prospect in understanding the pathogenesis of hypothyroidism.
Collapse
Affiliation(s)
- Xiaohan Zhang
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Ji-nan, China.,Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, China.,Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, China
| | - Wenbin Chen
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Ji-nan, China
| | - Shanshan Shao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Ji-nan, China.,Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, China.,Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yongfeng Song
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Ji-nan, China.,Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, China.,Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, China
| | - Chao Xu
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Ji-nan, China.,Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, China.,Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, China
| | - Ling Gao
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, China.,Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, China.,Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Ji-nan, China
| | - Chunxiu Hu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Ji-nan, China.,Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, China.,Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, China
| |
Collapse
|
13
|
Karasawa T, Kawashima A, Usui-Kawanishi F, Watanabe S, Kimura H, Kamata R, Shirasuna K, Koyama Y, Sato-Tomita A, Matsuzaka T, Tomoda H, Park SY, Shibayama N, Shimano H, Kasahara T, Takahashi M. Saturated Fatty Acids Undergo Intracellular Crystallization and Activate the NLRP3 Inflammasome in Macrophages. Arterioscler Thromb Vasc Biol 2018; 38:744-756. [PMID: 29437575 DOI: 10.1161/atvbaha.117.310581] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 01/25/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Inflammation provoked by the imbalance of fatty acid composition, such as excess saturated fatty acids (SFAs), is implicated in the development of metabolic diseases. Recent investigations suggest the possible role of the NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain containing 3) inflammasome, which regulates IL-1β (interleukin 1β) release and leads to inflammation, in this process. Therefore, we investigated the underlying mechanism by which SFAs trigger NLRP3 inflammasome activation. APPROACH AND RESULTS The treatment with SFAs, such as palmitic acid and stearic acid, promoted IL-1β release in murine primary macrophages while treatment with oleic acid inhibited SFA-induced IL-1β release in a dose-dependent manner. Analyses using polarized light microscopy revealed that intracellular crystallization was provoked in SFA-treated macrophages. As well as IL-1β release, the intracellular crystallization and lysosomal dysfunction were inhibited in the presence of oleic acid. These results suggest that SFAs activate NLRP3 inflammasome through intracellular crystallization. Indeed, SFA-derived crystals activated NLRP3 inflammasome and subsequent IL-1β release via lysosomal dysfunction. Excess SFAs also induced crystallization and IL-1β release in vivo. Furthermore, SFA-derived crystals provoked acute inflammation, which was impaired in IL-1β-deficient mice. CONCLUSIONS These findings demonstrate that excess SFAs cause intracellular crystallization and subsequent lysosomal dysfunction, leading to the activation of the NLRP3 inflammasome, and provide novel insights into the pathogenesis of metabolic diseases.
Collapse
Affiliation(s)
- Tadayoshi Karasawa
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.).
| | - Akira Kawashima
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Fumitake Usui-Kawanishi
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Sachiko Watanabe
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Hiroaki Kimura
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Ryo Kamata
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Koumei Shirasuna
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Yutaro Koyama
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Ayana Sato-Tomita
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Takashi Matsuzaka
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Hiroshi Tomoda
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Sam-Yong Park
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Naoya Shibayama
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Hitoshi Shimano
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Tadashi Kasahara
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.)
| | - Masafumi Takahashi
- From the Division of Inflammation Research, Center for Molecular Medicine (T. Karasawa, A.K., F.U.-K., S.W., H.K., R.K., K.S., Y.K., T. Kasahara, M.T.) and Division of Biophysics, Department of Physiology (A.S.-T., N.S.), Jichi Medical University, Tochigi, Japan; Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (T.M., H.S.); Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (H.T.); and Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan (S.-Y.P.).
| |
Collapse
|
14
|
Ito Z, Uchiyama K, Odahara S, Takami S, Saito K, Kobayashi H, Koido S, Kubota T, Ohkusa T, Saruta M. Fatty Acids as Useful Serological Markers for Crohn's Disease. Dig Dis 2017; 36:209-217. [PMID: 29275413 DOI: 10.1159/000485096] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/06/2017] [Indexed: 02/02/2023]
Abstract
BACKGROUND We have previously reported that patients with Crohn's disease (CD) have a very specific erythrocyte membrane phospholipid fatty acid profile. The findings of this study suggest that the activities of enzymes involved in the metabolism of linoleic acid (LA), that is, delta-6 desaturase, are higher in CD patients than in healthy individuals. METHODS We evaluated the utilities of various fatty acid compositions of the plasma (p-) as new serological markers for CD compared to those of erythrocyte membranes (e-). RESULTS Fifty CD patients and 50 healthy individuals were enrolled. In both plasma and erythrocyte membranes, the weight percentages of palmitic acid (PA) were significantly higher, while those of LA were significantly lower in CD patients than in controls. Fatty acids with high sensitivity and specificity were p-PA (0.86 and 0.74) and e-PA (0.80 and 0.74). With PA and LA as a CD fatty acid index (CDFAi), that is, CDFAi = (PA/LA), the sensitivity and specificity of plasma CDFAi (p-CDFAi) and e-CDFAi were 0.80 and 0.80; and 0.82 and 0.88 respectively. CONCLUSION In CD patients, various fatty acids were specifically altered in both plasma and erythrocytes, and p-PA and p-CDFAi are potentially useful as new serological markers for CD.
Collapse
Affiliation(s)
- Zensho Ito
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Jikei University School of Medicine (Kashiwa Hospital), Chiba, Japan
| | - Kan Uchiyama
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Jikei University School of Medicine (Kashiwa Hospital), Chiba, Japan
| | - Shunichi Odahara
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Jikei University School of Medicine (Kashiwa Hospital), Chiba, Japan
| | - Shinichiro Takami
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Jikei University School of Medicine (Kashiwa Hospital), Chiba, Japan
| | - Keisuke Saito
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Jikei University School of Medicine (Kashiwa Hospital), Chiba, Japan
| | - Hiroko Kobayashi
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Jikei University School of Medicine (Kashiwa Hospital), Chiba, Japan
| | - Shigeo Koido
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Jikei University School of Medicine (Kashiwa Hospital), Chiba, Japan
| | - Takahiro Kubota
- Department of Biopharmaceutics, Faculty of Pharmaceutical Science, Niigata University of Pharmacy and Applied Life Sciences, Niigata, Japan
| | - Toshifumi Ohkusa
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Jikei University School of Medicine (Kashiwa Hospital), Chiba, Japan
| | - Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Natarajan G, Perriotte-Olson C, Bhinderwala F, Powers R, Desouza CV, Talmon GA, Yuhang J, Zimmerman MC, Kabanov AV, Saraswathi V. Nanoformulated copper/zinc superoxide dismutase exerts differential effects on glucose vs lipid homeostasis depending on the diet composition possibly via altered AMPK signaling. Transl Res 2017; 188:10-26. [PMID: 28867395 PMCID: PMC5819896 DOI: 10.1016/j.trsl.2017.08.002] [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: 12/23/2016] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 12/17/2022]
Abstract
Evidence suggests that superoxide dismutase 1 (SOD1) promotes glucose vs lipid metabolism depending on the diet type. We recently reported that nanoformulated SOD1 (Nano) improved lipid metabolism without altering glucose homeostasis in high-fat (HF) diet-fed mice. Here, we sought to determine the effects and potential mechanisms of Nano in modulating glucose and lipid homeostasis in mice fed a normal chow diet (CD) vs HF diet. Mice were fed a CD or a HF diet (45%) for 10 wk and injected with Nano once every 2 days for 15 days. The fasting glucose level was lower (P < 0.05) in CD + Nano-treated mice compared to control. Conversely, blood glucose was not altered but serum triglycerides were lower in HF + Nano-treated mice. Genes involved in fatty acid synthesis were reduced by Nano in the skeletal muscle of CD but not of HF diet-fed mice. Adenosine monophosphate-activated protein kinase (AMPK), which promotes both glucose and lipid metabolism depending on the fuel availability, is activated by Nano in CD-fed mice. Moreover, Nano increased phosphorylation of ACC, a downstream target of AMPK, in both CD and HF diet-fed mice. Nano increased mitochondrial respiration in C2C12 myocytes in the presence of glucose or fatty acid, and this effect is inhibited by Compound C, an AMPK inhibitor. Our data suggest that Nano promotes glucose and lipid metabolism in CD and HF diet-fed mice, respectively, and this effect is mediated partly via AMPK signaling.
Collapse
Affiliation(s)
- Gopalakrishnan Natarajan
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Neb
| | - Curtis Perriotte-Olson
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Neb
| | - Fatema Bhinderwala
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Neb; Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Neb
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Neb; Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Neb
| | - Cyrus V Desouza
- VA Nebraska-Western Iowa Health Care System, Omaha, Neb; Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Neb
| | - Geoffrey A Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Neb
| | - Jiang Yuhang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Matthew C Zimmerman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Neb
| | - Alexander V Kabanov
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Viswanathan Saraswathi
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Neb; VA Nebraska-Western Iowa Health Care System, Omaha, Neb.
| |
Collapse
|
16
|
Li Z, Kim HJ, Park MS, Ji GE. Effects of fermented ginseng root and ginseng berry on obesity and lipid metabolism in mice fed a high-fat diet. J Ginseng Res 2017; 42:312-319. [PMID: 29983612 PMCID: PMC6026359 DOI: 10.1016/j.jgr.2017.04.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/07/2017] [Indexed: 12/30/2022] Open
Abstract
Background Previous studies have shown that both ginseng root and ginseng berry exhibit antiobesity and antidiabetic effects. However, a direct comparison of the efficacy and mechanisms between the root and the berry after oral administration remains to be illuminated. Methods In this study, we observed the effects of fermented ginseng root (FGR) and fermented ginseng berry (FGB) on obesity and lipid metabolism in high-fat diet induced obese mice. Results FGR and FGB significantly inhibited the activity of pancreatic lipase in vitro. Both FGR and FGB significantly suppressed weight gain and excess food intake and improved hypercholesterolemia and fatty liver, while only FGR significantly attenuated hyperglycemia and insulin resistance. Both FGR and FGB significantly inhibited the mRNA expression of Ldlr and Acsl1 while FGR also significantly inhibited expression of Cebpa and Dgat2 in liver. FGR significantly decreased the epididymal fat weight of mice while FGB significantly inhibited the mRNA expression of genes Cebpa, Fas, Hsl, Il1b, and Il6 in adipose tissue. Conclusion Saponin from both FGR and FGB had a beneficial effect on high-fat diet-induced obesity. Compared to FGB, FGR exhibited more potent antihyperglycemic and antiobesity effect. However, only FGB significantly inhibited mRNA expression of inflammatory markers such as interleukins 1β and 6 in adipose tissue.
Collapse
Affiliation(s)
- Zhipeng Li
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea
| | - Hee Jung Kim
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea
| | - Myeong Soo Park
- Department of Hotel Culinary Arts, Yeonsung University, Anyang, Republic of Korea
| | - Geun Eog Ji
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea.,Research Institute, Bifido Co., Ltd., Hongchun, Republic of Korea
| |
Collapse
|
17
|
Saraswathi V, Perriotte-Olson C, Ganesan M, Desouza CV, Alnouti Y, Duryee MJ, Thiele GM, Nordgren TM, Clemens DL. A combination of dietary N-3 fatty acids and a cyclooxygenase-1 inhibitor attenuates nonalcoholic fatty liver disease in mice. J Nutr Biochem 2017; 42:149-159. [PMID: 28187366 DOI: 10.1016/j.jnutbio.2017.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/13/2016] [Accepted: 01/19/2017] [Indexed: 12/12/2022]
Abstract
We sought to determine whether a combination of purified n-3 fatty acids (n-3) and SC-560 (SC), a cyclooxygenase-1-specific inhibitor, is effective in ameliorating nonalcoholic fatty liver disease in obesity. Female wild-type mice were fed a high-fat and high-cholesterol diet (HF) supplemented with n-3 in the presence or absence of SC. Mice treated with SC alone exhibited no change in liver lipids, whereas n-3-fed mice tended to have lower hepatic lipids. Mice given n-3+SC had significantly lower liver lipids compared with HF controls indicating enhanced lipid clearance. Total and sulfated bile acids were significantly higher only in n-3+SC-treated mice compared with chow diet (CD) controls. Regarding mechanisms, the level of pregnane X receptor (PXR), a nuclear receptor regulating drug/bile detoxification, was significantly higher in mice given n-3 or n-3+SC. Studies in precision-cut liver slices and in cultured hepatoma cells showed that n-3+SC enhanced not only the expression/activation of PXR and its target genes but also the expression of farnesoid X receptor (FXR), another regulator of bile synthesis/clearance, indicating that n-3+SC can induce both PXR and FXR. The mRNA level of FGFR4 which inhibits bile formation showed a significant reduction in Huh 7 cells upon n-3 and n-3+SC treatment. PXR overexpression in hepatoma cells confirmed that n-3 or SC each induced the expression of PXR target genes and in combination had an enhanced effect. Our findings suggest that combining SC with n-3 potentiates its lipid-lowering effect, in part, by enhanced PXR and/or altered FXR/FGFR4 signaling.
Collapse
Affiliation(s)
- Viswanathan Saraswathi
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Curtis Perriotte-Olson
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA
| | - Murali Ganesan
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cyrus V Desouza
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Yazen Alnouti
- Department of Pharmaceutical Science, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael J Duryee
- Department of Internal Medicine, Division of Rheumatology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Pharmaceutical Science, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Tara M Nordgren
- Department of Internal Medicine, Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dahn L Clemens
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
18
|
Identification of ACSL4 as a biomarker and contributor of ferroptosis. Biochem Biophys Res Commun 2016; 478:1338-43. [PMID: 27565726 DOI: 10.1016/j.bbrc.2016.08.124] [Citation(s) in RCA: 690] [Impact Index Per Article: 76.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 08/22/2016] [Indexed: 02/07/2023]
Abstract
Ferroptosis, a recently identified form of non-apoptotic cell death, is involved in several physiological and pathological processes. Although lipid peroxidation plays a central role in triggering ferroptosis, the essential regulator of lipid metabolism in ferroptosis remains poorly defined. Here, we show that acyl-CoA synthetase long-chain family member 4 (ACSL4) is required for ferroptotic cancer cell death. Compared with ferroptosis-sensitive cells (e.g., HepG2 and HL60), the expression of ACSL4 was remarkably downregulated in ferroptosis-resistant cells (e.g., LNCaP and K562). In contrast, the expression of other ACSLs, including ACSL1, ACSL3, ACSL5, and ACSL6, did not correlate with ferroptosis sensitivity. Moreover, knockdown of ACSL4 by specific shRNA inhibited erastin-induced ferroptosis in HepG2 and HL60 cells, whereas overexpression of ACSL4 by gene transfection restored sensitivity of LNCaP and K562 cells to erastin. Mechanically, ACSL4-mediated production of 5-hydroxyeicosatetraenoic acid (5-HETE) contributed to ferroptosis. Pharmacological inhibition of 5-HETE production by zileuton limited ACSL4 overexpression-induced ferroptosis. Collectively, these results indicate that ACSL4 is not only a sensitive monitor of ferroptosis, but also an important contributor of ferroptosis.
Collapse
|
19
|
Impact of high-fat diet on the proteome of mouse liver. J Nutr Biochem 2016; 31:10-9. [DOI: 10.1016/j.jnutbio.2015.12.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 10/06/2015] [Accepted: 12/22/2015] [Indexed: 11/22/2022]
|
20
|
Perriotte-Olson C, Adi N, Manickam DS, Westwood RA, Desouza CV, Natarajan G, Crook A, Kabanov AV, Saraswathi V. Nanoformulated copper/zinc superoxide dismutase reduces adipose inflammation in obesity. Obesity (Silver Spring) 2016; 24:148-56. [PMID: 26612356 PMCID: PMC6699510 DOI: 10.1002/oby.21348] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 08/20/2015] [Accepted: 08/26/2015] [Indexed: 12/17/2022]
Abstract
OBJECTIVE An intimate association exists between oxidative stress and inflammation. Because adipose tissue (AT) inflammation is intricately linked to metabolic disorders, it was hypothesized that reducing oxidative stress would be effective in ameliorating AT inflammation in obesity. METHODS Wild-type mice were fed a high-fat diet (HF) for 8 weeks followed by a 2-week treatment with nanoformulated copper/zinc superoxide dismutase (NanoSOD). The mice were divided into: 1) chow diet, 2) HF, and 3) HF + NanoSOD. RESULTS The HF + NanoSOD-treated mice showed a significant decrease in plasma and liver triglycerides when compared with HF-fed mice. Interestingly, NanoSOD reduced the expression of macrophage and inflammatory markers in visceral AT (VAT) and stromal cells derived from VAT. Moreover, the activation of proinflammatory signaling pathways, in particular, the extracellular signal-regulated kinases, was blunted in VAT on NanoSOD treatment. However, markers of oxidative stress were not altered significantly in the HF + NanoSOD group in the experimental conditions. Pretreatment of either macrophages or adipocytes significantly reduced the inflammatory response invoked in an in vitro coculture system, further supporting the role of NanoSOD in inhibiting obesity-linked inflammation. CONCLUSIONS This data suggest that NanoSOD is effective not only in reducing AT macrophage accumulation and AT inflammation but also in promoting triglyceride metabolism in obesity.
Collapse
Affiliation(s)
- Curtis Perriotte-Olson
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Research Service, Veterans Administration Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Nikhil Adi
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Research Service, Veterans Administration Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Devika S Manickam
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rachel A Westwood
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Research Service, Veterans Administration Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Cyrus V Desouza
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Research Service, Veterans Administration Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Gopalakrishnan Natarajan
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Research Service, Veterans Administration Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Alexandra Crook
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Research Service, Veterans Administration Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Alexander V Kabanov
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Viswanathan Saraswathi
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Research Service, Veterans Administration Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| |
Collapse
|
21
|
Production of conjugated linoleic acid by heterologous expression of linoleic acid isomerase in oleaginous fungus Mortierella alpina. Biotechnol Lett 2015; 37:1983-92. [PMID: 26109146 DOI: 10.1007/s10529-015-1871-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/22/2015] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To increase the commercial value of oleaginous fungus Mortierella alpina by incorporation of trans-10,cis-12 conjugated linoleic acid (CLA) into the polyunsaturated fatty acids (PUFAs) of M. alpina via Propionibacterium acnes isomerase (PAI) conversion. RESULTS The PAI gene and the codon optimization version were heterologously expressed in M. alpina via Agrobacterium tumefaciens-mediated transformation (ATMT). Coding usage modification significantly improved the translation of PAI transcripts and trans-10,cis-12 CLA was produced up to 1.2 mg l(-1), which corresponds to approx. 0.05% of the total fatty acid (TFA). Since PAI prefers free linoleic acid as a substrate rather than any other forms, 5 μM long-chain acyl CoA synthetase inhibitor was added and the trans-10,cis-12 CLA content increased approx. 24-fold to 29 mg l(-1), reaching up to 1.2% (w/w) of the TFA in M. alpina. CONCLUSION Heterologous expression of PAI in M. alpina by ATMT methods is a practicable way in biosynthesis of CLA and this system may be a feasible platform for industrial production of CLA.
Collapse
|
22
|
Hill AA, Reid Bolus W, Hasty AH. A decade of progress in adipose tissue macrophage biology. Immunol Rev 2015; 262:134-52. [PMID: 25319332 DOI: 10.1111/imr.12216] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
One decade has passed since seminal publications described macrophage infiltration into adipose tissue (AT) as a key contributor to inflammation and obesity-related insulin resistance. Currently, a PubMed search for 'adipose tissue inflammation' reveals over 3500 entries since these original reports. We now know that resident macrophages in lean AT are alternatively activated, M2-like, and play a role in AT homeostasis. In contrast, the macrophages in obese AT are dramatically increased in number and are predominantly classically activated, M1-like, and promote inflammation and insulin resistance. Mediators of AT macrophage (ATM) phenotype include adipokines and fatty acids secreted from adipocytes as well as cytokines secreted from other immune cells in AT. There are several mechanisms that could explain the large increase in ATMs in obesity. These include recruitment-dependent mechanisms such as adipocyte death, chemokine release, and lipolysis of fatty acids. Newer evidence also points to recruitment-independent mechanisms such as impaired apoptosis, increased proliferation, and decreased egress. Although less is known about the homeostatic function of M2-like resident ATMs, recent evidence suggests roles in AT expansion, thermoregulation, antigen presentation, and iron homeostasis. The field of immunometabolism has come a long way in the past decade, and many exciting new discoveries are bound to be made in the coming years that will expand our understanding of how AT stands at the junction of immune and metabolic co-regulation.
Collapse
Affiliation(s)
- Andrea A Hill
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | |
Collapse
|
23
|
Lu M, Kho T, Munford RS. Prolonged triglyceride storage in macrophages: pHo trumps pO2 and TLR4. THE JOURNAL OF IMMUNOLOGY 2014; 193:1392-7. [PMID: 24973452 DOI: 10.4049/jimmunol.1400886] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Lipid-laden macrophages contribute to pathologies as diverse as atherosclerosis and tuberculosis. Three common stimuli are known to promote macrophage lipid storage: low tissue oxygen tension (pO2), low extracellular pH (pHo), and exposure to agonists such as bacterial LPS. Noting that cells responding to low pO2 or agonistic bacterial molecules often decrease pHo by secreting lactic and other carboxylic acids, we studied how pHo influences the stimulation of triacylglycerol (TAG) storage by low pO2 and LPS. We found that TAG retention after incubation for 48-72 h was inversely related to pHo when primary macrophages were cultured in 21% oxygen, 4% oxygen, or with LPS at either oxygen concentration. Maintaining pHo at ~7.4 was sufficient to prevent the increase in prolonged TAG storage induced by either low pO2 or LPS. The strong influence of pHo on TAG retention may explain why lipid-laden macrophages are found in some tissue environments and not in others. It is also possible that other long-term cellular changes currently attributed to low pO2 or bacterial agonists may be promoted, at least in part, by the decrease in pHo that these stimuli induce.
Collapse
Affiliation(s)
- Mingfang Lu
- Antibacterial Host Defense Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Terry Kho
- Antibacterial Host Defense Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Robert S Munford
- Antibacterial Host Defense Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
24
|
Namgaladze D, Lips S, Leiker TJ, Murphy RC, Ekroos K, Ferreiros N, Geisslinger G, Brüne B. Inhibition of macrophage fatty acid β-oxidation exacerbates palmitate-induced inflammatory and endoplasmic reticulum stress responses. Diabetologia 2014; 57:1067-77. [PMID: 24488024 DOI: 10.1007/s00125-014-3173-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 01/02/2014] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS Saturated fatty acids (SFAs) such as palmitate activate inflammatory pathways and elicit an endoplasmic reticulum (ER) stress response in macrophages, thereby contributing to the development of insulin resistance linked to the metabolic syndrome. This study addressed the question of whether or not mitochondrial fatty acid β-oxidation (FAO) affects macrophage responses to SFA. METHODS We modulated the activity of carnitine palmitoyl transferase 1A (CPT1A) in macrophage-differentiated THP-1 monocytic cells using genetic or pharmacological approaches, treated the cells with palmitate and analysed the proinflammatory and ER stress signatures. RESULTS To inhibit FAO, we created THP-1 cells with a stable knockdown (KD) of CPT1A and differentiated them to macrophages. Consequently, in CPT1A-silenced cells FAO was reduced. CPT1A KD in THP-1 macrophages increased proinflammatory signalling, cytokine expression and ER stress responses after palmitate treatment. In addition, in human primary macrophages CPT1A KD elevated palmitate-induced inflammatory gene expression. Pharmacological inhibition of FAO with etomoxir recapitulated the CPT1A KD phenotype. Conversely, overexpression of a malonyl-CoA-insensitive CPT1A M593S mutant reduced inflammatory and ER stress responses to palmitate in THP-1 macrophages. Macrophages with a CPT1A KD accumulated diacylglycerols and triacylglycerols after palmitate treatment, while ceramide accumulation remained unaltered. Moreover, lipidomic analysis of ER phospholipids revealed increased palmitate incorporation into phosphatidylethanolamine and phosphatidylserine classes associated with the CPT1A KD. CONCLUSIONS/INTERPRETATION Our data indicate that FAO attenuates inflammatory and ER stress responses in SFA-exposed macrophages, suggesting an anti-inflammatory impact of drugs that activate FAO.
Collapse
Affiliation(s)
- Dmitry Namgaladze
- Faculty of Medicine, Institute of Biochemistry I/ZAFES, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Afonso MDS, Castilho G, Lavrador MSF, Passarelli M, Nakandakare ER, Lottenberg SA, Lottenberg AM. The impact of dietary fatty acids on macrophage cholesterol homeostasis. J Nutr Biochem 2014; 25:95-103. [DOI: 10.1016/j.jnutbio.2013.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 09/11/2013] [Accepted: 10/03/2013] [Indexed: 11/16/2022]
|
26
|
Kratky D, Obrowsky S, Kolb D, Radovic B. Pleiotropic regulation of mitochondrial function by adipose triglyceride lipase-mediated lipolysis. Biochimie 2014; 96:106-12. [PMID: 23827855 PMCID: PMC3859496 DOI: 10.1016/j.biochi.2013.06.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 06/20/2013] [Indexed: 12/12/2022]
Abstract
Lipolysis is defined as the catabolism of triacylglycerols (TGs) stored in cellular lipid droplets. Recent discoveries of essential lipolytic enzymes and characterization of numerous regulatory proteins and mechanisms have fundamentally changed our perception of lipolysis and its impact on cellular metabolism. Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme for TG catabolism in most cells and tissues. This review focuses on recent advances in understanding the (patho)physiological impact due to defective lipolysis by ATGL deficiency on mitochondrial (dys)function. Depending on the type of cells and tissues investigated, absence of ATGL has pleiotropic roles in mitochondrial function.
Collapse
Affiliation(s)
- Dagmar Kratky
- Institute of Molecular Biology and Biochemistry, Center for Molecular Medicine, Medical University of Graz, Harrachgasse 21, 8010 Graz, Austria.
| | | | | | | |
Collapse
|
27
|
Murali G, Desouza CV, Clevenger ME, Ramalingam R, Saraswathi V. Differential effects of eicosapentaenoic acid and docosahexaenoic acid in promoting the differentiation of 3T3-L1 preadipocytes. Prostaglandins Leukot Essent Fatty Acids 2014; 90:13-21. [PMID: 24332315 DOI: 10.1016/j.plefa.2013.10.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/01/2013] [Accepted: 10/30/2013] [Indexed: 11/24/2022]
Abstract
The objective of this study was to determine the effects of enrichment with n-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), on the differentiation of 3T3-L1 preadipocytes. Enrichment with DHA but not EPA significantly increased the differentiation markers compared to control differentiated cells. DHA compared to EPA treatment led to a greater increase in adiponectin secretion and, conditioned media collected from DHA treated cells inhibited monocyte migration. Moreover, DHA treatment resulted in inhibition of pro-inflammatory signaling pathways. DHA treated cells predominantly accumulated DHA in phospholipids whereas EPA treatment led to accumulation of both EPA and its elongation product docosapentaenoic acid (DPA), an n-3 fatty acid. Of note, adding DPA to DHA inhibited DHA-induced differentiation. The differential effects of EPA and DHA on preadipocyte differentiation may be due, in part, to differences in their intracellular modification which could impact the type of n-3 fatty acids incorporated into the cells.
Collapse
Affiliation(s)
- Ganesan Murali
- Departments of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, Omaha, NE, United States; Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States; Research Services, VA Nebraska Western Iowa Health Care System, Omaha, NE, United States
| | - Cyrus V Desouza
- Research Services, VA Nebraska Western Iowa Health Care System, Omaha, NE, United States; Departments of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, Omaha, NE, United States
| | - Michelle E Clevenger
- Departments of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, Omaha, NE, United States; Research Services, VA Nebraska Western Iowa Health Care System, Omaha, NE, United States
| | - Ramesh Ramalingam
- Departments of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, Omaha, NE, United States; Research Services, VA Nebraska Western Iowa Health Care System, Omaha, NE, United States
| | - Viswanathan Saraswathi
- Departments of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, Omaha, NE, United States; Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States; Research Services, VA Nebraska Western Iowa Health Care System, Omaha, NE, United States.
| |
Collapse
|
28
|
Huang YL, Morales-Rosado J, Ray J, Myers TG, Kho T, Lu M, Munford RS. Toll-like receptor agonists promote prolonged triglyceride storage in macrophages. J Biol Chem 2013; 289:3001-12. [PMID: 24337578 DOI: 10.1074/jbc.m113.524587] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Macrophages in infected tissues may sense microbial molecules that significantly alter their metabolism. In a seeming paradox, these critical host defense cells often respond by increasing glucose catabolism while simultaneously storing fatty acids (FA) as triglycerides (TAG) in lipid droplets. We used a load-chase strategy to study the mechanisms that promote long term retention of TAG in murine and human macrophages. Toll-like receptor (TLR)1/2, TLR3, and TLR4 agonists all induced the cells to retain TAG for ≥3 days. Prolonged TAG retention was accompanied by the following: (a) enhanced FA uptake and FA incorporation into TAG, with long lasting increases in acyl-CoA synthetase long 1 (ACSL1) and diacylglycerol acyltransferase-2 (DGAT2), and (b) decreases in lipolysis and FA β-oxidation that paralleled a prolonged drop in adipose triglyceride lipase (ATGL). TLR agonist-induced TAG storage is a multifaceted process that persists long after most early pro-inflammatory responses have subsided and may contribute to the formation of "lipid-laden" macrophages in infected tissues.
Collapse
Affiliation(s)
- Ying-ling Huang
- From the Antibacterial Host Defense Section, Laboratory of Clinical Infectious Diseases
| | | | | | | | | | | | | |
Collapse
|
29
|
Orr JS, Puglisi MJ, Ellacott KL, Lumeng CN, Wasserman DH, Hasty AH. Toll-like receptor 4 deficiency promotes the alternative activation of adipose tissue macrophages. Diabetes 2012; 61:2718-27. [PMID: 22751700 PMCID: PMC3478520 DOI: 10.2337/db11-1595] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obesity is characterized by adipose tissue (AT) macrophage (ATM) accumulation, which promotes AT inflammation and dysfunction. Toll-like receptor 4 (TLR4) deficiency attenuates AT inflammation in obesity but does not impede the accumulation of ATMs. The purpose of the current study was to determine whether TLR4 deficiency alters ATM polarization. TLR4(-/-) and wild-type mice were fed a low-fat, high-monounsaturated fat (HF(MUFA)), or a high-saturated fat (HF(SFA)) diet for 16 weeks. Further, we used a bone marrow transplant model to determine the influence of hematopoietic cell TLR4 signaling. The metabolic and inflammatory responses to high-fat feeding and ATM phenotype were assessed. Global and hematopoietic cell TLR4 deficiency, irrespective of recipient genotype, produced a shift in ATM phenotype toward an alternatively activated state, which was accompanied by reduced AT inflammation. Despite the observed shift in ATM phenotype, neither global nor hematopoietic cell TLR4 deficiency influenced systemic insulin sensitivity after high-fat feeding. Results of the current study suggest that TLR4 directly influences ATM polarization but question the relevance of TLR4 signaling to systemic glucose homeostasis in obesity.
Collapse
|
30
|
Wang HW, Fang JS, Kuang X, Miao LY, Wang C, Xia GL, King ML, Zhang J. Activity of long-chain acyl-CoA synthetase is required for maintaining meiotic arrest in Xenopus laevis. Biol Reprod 2012; 87:74. [PMID: 22786823 DOI: 10.1095/biolreprod.112.100511] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In most vertebrates, fully grown oocytes are arrested in meiotic prophase I and only resume the cell cycle upon external stimuli, such as hormones. The proper arrest and resumption of the meiotic cycle is critical for reproduction. A Galpha(S) signaling pathway essential for the arrest is conserved in organisms from Xenopus to mouse and human. A previous gene association study implicated that mutations of human ACSL6 may be related to premature ovarian failure. However, functional roles of ACSL6 in human infertility have yet to be reported. In the present study, we found that triacsin C, a potent and specific inhibitor for ACSL, triggers maturation in Xenopus and mouse oocytes in the absence of hormone, suggesting ACSL activity is required for the oocyte arrest. In Xenopus, acsl1b may fulfill a major role in the process, because inhibition of acsl1b by knocking down its RNA results in abnormal acceleration of oocyte maturation. Such abnormally matured eggs cannot support early embryonic development. Moreover, direct inhibition of protein palmitoylation, which lies downstream of ACSLs, also causes oocyte maturation. Furthermore, palmitoylation of Galpha(s), which is essential for its function, is inhibited when the ACSL activity is blocked by triacsin C in Xenopus. Thus, disruption of ACSL activity causes inhibition of the Galpha(s) signaling pathway in the oocytes, which may result in premature ovarian failure in human.
Collapse
Affiliation(s)
- Hua-wei Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Radovic B, Aflaki E, Kratky D. Adipose triglyceride lipase in immune response, inflammation, and atherosclerosis. Biol Chem 2012; 393:1005-11. [PMID: 22944699 PMCID: PMC3520003 DOI: 10.1515/hsz-2012-0192] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 05/23/2012] [Indexed: 12/15/2022]
Abstract
Consistent with its central importance in lipid and energy homeostasis, lipolysis occurs in essentially all tissues and cell types, including macrophages. The hydrolytic cleavage of triacylglycerol by adipose triglyceride lipase (ATGL) generates non-esterified fatty acids, which are subsequently used as essential precursors for lipid and membrane synthesis, mediators in cell signaling processes or as energy substrate in mitochondria. This review summarizes the current knowledge concerning the consequences of ATGL deficiency in macrophages with particular emphasis on macrophage (dys)-function, apoptosis, and atherosclerosis.
Collapse
Affiliation(s)
- Branislav Radovic
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Harrachgasse 21, A-8010 Graz, Austria
| | | | - Dagmar Kratky
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Harrachgasse 21, A-8010 Graz, Austria
| |
Collapse
|
32
|
Anderson EK, Hill AA, Hasty AH. Stearic acid accumulation in macrophages induces toll-like receptor 4/2-independent inflammation leading to endoplasmic reticulum stress-mediated apoptosis. Arterioscler Thromb Vasc Biol 2012; 32:1687-95. [PMID: 22556332 DOI: 10.1161/atvbaha.112.250142] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Elevated serum free fatty acid levels are associated with an increased risk of cardiovascular disease and type 2 diabetes mellitus. Macrophages are recruited to atherosclerotic plaques and metabolic tissues during obesity and accumulate lipids, including free fatty acids. We investigated the molecular consequences of intracellular saturated free fatty acid accumulation in macrophages. METHODS AND RESULTS Previously, we demonstrated that cotreatment of mouse peritoneal macrophages (MPMs) with stearic acid and triacsin C (an inhibitor of long-chain acyl coenzyme A synthetases) results in intracellular free fatty acid accumulation and apoptosis. Here, we used Western blotting analysis, real-time reverse transcription polymerase chain reaction, and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining to assess endoplasmic reticulum (ER) stress, inflammation, and apoptosis in MPMs. Intracellular stearic acid accumulation induces Toll-like receptor 4/2-independent inflammation that results in ER stress-mediated apoptosis of MPMs. Polarization of MPMs to a proinflammatory M1 phenotype increases their susceptibility to inflammation and ER stress, but not apoptosis, in response to cotreatment with stearic acid and triacsin C. CONCLUSIONS Intracellular accumulation of stearic acid in MPMs activates inflammatory signaling, leading to ER stress-mediated apoptosis. M1 macrophages are more prone to stearic acid-induced inflammation and ER stress. These same pathways may be activated in macrophages residing in atherosclerotic plaques and metabolic tissues during conditions of obesity and hyperlipidemia.
Collapse
Affiliation(s)
- Emily K Anderson
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232-0615, USA
| | | | | |
Collapse
|
33
|
Koo MS, Subbian S, Kaplan G. Strain specific transcriptional response in Mycobacterium tuberculosis infected macrophages. Cell Commun Signal 2012; 10:2. [PMID: 22280836 PMCID: PMC3317440 DOI: 10.1186/1478-811x-10-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 01/26/2012] [Indexed: 11/10/2022] Open
Abstract
Background Tuberculosis (TB), a bacterial infection caused by Mycobacterium tuberculosis (Mtb) remains a significant health problem worldwide with a third of the world population infected and nearly nine million new cases claiming 1.1 million deaths every year. The outcome following infection by Mtb is determined by a complex and dynamic host-pathogen interaction in which the phenotype of the pathogen and the immune status of the host play a role. However, the molecular mechanism by which Mtb strains induce different responses during intracellular infection of the host macrophage is not fully understood. To explore the early molecular events triggered upon Mtb infection of macrophages, we studied the transcriptional responses of murine bone marrow-derived macrophages (BMM) to infection with two clinical Mtb strains, CDC1551 and HN878. These strains have previously been shown to differ in their virulence/immunogenicity in the mouse and rabbit models of pulmonary TB. Results In spite of similar intracellular growth rates, we observed that compared to HN878, infection by CDC1551 of BMM was associated with an increased global transcriptome, up-regulation of a specific early (6 hours) immune response network and significantly elevated nitric oxide production. In contrast, at 24 hours post-infection of BMM by HN878, more host genes involved in lipid metabolism, including cholesterol metabolism and prostaglandin synthesis were up-regulated, compared to infection with CDC1551. In association with the differences in the macrophage responses to infection with the 2 Mtb strains, intracellular CDC1551 expressed higher levels of stress response genes than did HN878. Conclusions In association with the early and more robust macrophage activation, intracellular CDC1551 cells were exposed to a higher level of stress leading to increased up-regulation of the bacterial stress response genes. In contrast, sub-optimal activation of macrophages and induction of a dysregulated host cell lipid metabolism favored a less stressful intracellular environment for HN878. Our findings suggest that the ability of CDC1551 and HN878 to differentially activate macrophages during infection probably determines their ability to either resist host cell immunity and progress to active disease or to succumb to the host protective responses and be driven into a non-replicating latent state in rabbit lungs.
Collapse
Affiliation(s)
- Mi-Sun Koo
- Laboratory of Mycobacterial Immunity and Pathogenesis, The Public Health Research Institute (PHRI) at the University of Medicine and Dentistry of New Jersey (UNDNJ), 225 Warren Street, Newark, New Jersey 07103, USA.
| | | | | |
Collapse
|
34
|
Diabetes promotes an inflammatory macrophage phenotype and atherosclerosis through acyl-CoA synthetase 1. Proc Natl Acad Sci U S A 2012; 109:E715-24. [PMID: 22308341 DOI: 10.1073/pnas.1111600109] [Citation(s) in RCA: 225] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The mechanisms that promote an inflammatory environment and accelerated atherosclerosis in diabetes are poorly understood. We show that macrophages isolated from two different mouse models of type 1 diabetes exhibit an inflammatory phenotype. This inflammatory phenotype associates with increased expression of long-chain acyl-CoA synthetase 1 (ACSL1), an enzyme that catalyzes the thioesterification of fatty acids. Monocytes from humans and mice with type 1 diabetes also exhibit increased ACSL1. Furthermore, myeloid-selective deletion of ACSL1 protects monocytes and macrophages from the inflammatory effects of diabetes. Strikingly, myeloid-selective deletion of ACSL1 also prevents accelerated atherosclerosis in diabetic mice without affecting lesions in nondiabetic mice. Our observations indicate that ACSL1 plays a critical role by promoting the inflammatory phenotype of macrophages associated with type 1 diabetes; they also raise the possibilities that diabetic atherosclerosis has an etiology that is, at least in part, distinct from the etiology of nondiabetic vascular disease and that this difference is because of increased monocyte and macrophage ACSL1 expression.
Collapse
|
35
|
Kanter JE, Tang C, Oram JF, Bornfeldt KE. Acyl-CoA synthetase 1 is required for oleate and linoleate mediated inhibition of cholesterol efflux through ATP-binding cassette transporter A1 in macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:358-64. [PMID: 22020260 DOI: 10.1016/j.bbalip.2011.10.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 09/30/2011] [Accepted: 10/01/2011] [Indexed: 12/31/2022]
Abstract
Diabetes and insulin resistance increase the risk of cardiovascular disease caused by atherosclerosis through mechanisms that are poorly understood. Lipid-loaded macrophages are key contributors to all stages of atherosclerosis. We have recently shown that diabetes associated with increased plasma lipids reduces cholesterol efflux and levels of the reverse cholesterol transporter ABCA1 (ATP-binding cassette transporter A1) in mouse macrophages, which likely contributes to macrophage lipid accumulation in diabetes. Furthermore, we and others have shown that unsaturated fatty acids reduce ABCA1-mediated cholesterol efflux, and that this effect is mediated by the acyl-CoA derivatives of the fatty acids. We therefore investigated whether acyl-CoA synthetase 1 (ACSL1), a key enzyme mediating acyl-CoA synthesis in macrophages, could directly influence ABCA1 levels and cholesterol efflux in these cells. Mouse macrophages deficient in ACSL1 exhibited reduced sensitivity to oleate- and linoleate-mediated ABCA1 degradation, which resulted in increased ABCA1 levels and increased apolipoprotein A-I-dependent cholesterol efflux in the presence of these fatty acids, as compared with wildtype mouse macrophages. Conversely, overexpression of ACSL1 resulted in reduced ABCA1 levels and reduced cholesterol efflux in the presence of unsaturated fatty acids. Thus, the reduced ABCA1 and cholesterol efflux in macrophages subjected to conditions of diabetes and elevated fatty load may, at least in part, be mediated by ACSL1. These observations raise the possibility that ABCA1 levels could be increased by inhibition of acyl-CoA synthetase activity in vivo. This article is part of a Special Issue entitled Advances in High Density Lipoprotein Formation and Metabolism: A Tribute to John F. Oram (1945-2010).
Collapse
Affiliation(s)
- Jenny E Kanter
- Department of Pathology, Diabetes and Obesity Center of Excellence, University of Washington School of Medicine, Seattle, WA 98109, USA
| | | | | | | |
Collapse
|
36
|
Lammers B, Chandak PG, Aflaki E, Van Puijvelde GHM, Radovic B, Hildebrand RB, Meurs I, Out R, Kuiper J, Van Berkel TJC, Kolb D, Haemmerle G, Zechner R, Levak-Frank S, Van Eck M, Kratky D. Macrophage adipose triglyceride lipase deficiency attenuates atherosclerotic lesion development in low-density lipoprotein receptor knockout mice. Arterioscler Thromb Vasc Biol 2010; 31:67-73. [PMID: 21030715 DOI: 10.1161/atvbaha.110.215814] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The consequences of macrophage triglyceride (TG) accumulation on atherosclerosis have not been studied in detail so far. Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme for the initial step in TG hydrolysis. Because ATGL knockout (KO) mice exhibit massive TG accumulation in macrophages, we used ATGL KO mice to study the effects of macrophage TG accumulation on atherogenesis. METHODS AND RESULTS Low-density lipoprotein receptor (LDLr) KO mice were transplanted with bone marrow from ATGL KO (ATGL KO→LDLr KO) or wild-type (WT→LDLr KO) mice and challenged with a Western-type diet for 9 weeks. Despite TG accumulation in ATGL KO macrophages, atherosclerosis in ATGL KO→LDLr KO mice was 43% reduced associated with decreased plasma monocyte chemoattractant protein-1 (MCP-1) and macrophage interleukin-6 concentrations. This coincided with a reduced amount of macrophages, possibly because of a 39% increase in intraplaque apoptosis and a decreased migratory capacity of ATGL KO macrophages. The reduced number of white blood cells might be due to a 36% decreased Lin(-)Sca-1(+)cKit(+) hematopoietic stem cell population. CONCLUSIONS We conclude that the attenuation of atherogenesis in ATGL KO→LDLr KO mice is due to decreased infiltration of less inflammatory macrophages into the arterial wall and increased macrophage apoptosis.
Collapse
Affiliation(s)
- Bart Lammers
- Division of Biopharmaceutics, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Phillips CM, Goumidi L, Bertrais S, Field MR, Cupples LA, Ordovas JM, Defoort C, Lovegrove JA, Drevon CA, Gibney MJ, Blaak EE, Kiec-Wilk B, Karlstrom B, Lopez-Miranda J, McManus R, Hercberg S, Lairon D, Planells R, Roche HM. Gene-nutrient interactions with dietary fat modulate the association between genetic variation of the ACSL1 gene and metabolic syndrome. J Lipid Res 2010; 51:1793-800. [PMID: 20176858 PMCID: PMC2882737 DOI: 10.1194/jlr.m003046] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 02/17/2010] [Indexed: 12/14/2022] Open
Abstract
Long-chain acyl CoA synthetase 1 (ACSL1) plays an important role in fatty acid metabolism and triacylglycerol (TAG) synthesis. Disturbance of these pathways may result in dyslipidemia and insulin resistance, hallmarks of the metabolic syndrome (MetS). Dietary fat is a key environmental factor that may interact with genetic determinants of lipid metabolism to affect MetS risk. We investigated the relationship between ACSL1 polymorphisms (rs4862417, rs6552828, rs13120078, rs9997745, and rs12503643) and MetS risk and determined potential interactions with dietary fat in the LIPGENE-SU.VI.MAX study of MetS cases and matched controls (n = 1,754). GG homozygotes for rs9997745 had increased MetS risk {odds ratio (OR) 1.90 [confidence interval (CI) 1.15, 3.13]; P = 0.01}, displayed elevated fasting glucose (P = 0.001) and insulin concentrations (P = 0.002) and increased insulin resistance (P = 0.03) relative to the A allele carriers. MetS risk was modulated by dietary fat, whereby the risk conferred by GG homozygosity was abolished among individuals consuming either a low-fat (<35% energy) or a high-PUFA diet (>5.5% energy). In conclusion, ACSL1 rs9997745 influences MetS risk, most likely via disturbances in fatty acid metabolism, which was modulated by dietary fat consumption, particularly PUFA intake, suggesting novel gene-nutrient interactions.
Collapse
Affiliation(s)
- Catherine M. Phillips
- Nutrigenomics Research Group, UCD School of Public Health and Population Science, UCD Conway Institute, and Institute of Food and Health, University College Dublin, Ireland
| | - Louisa Goumidi
- INSERM 476, Lipid nutrients and prevention of metabolic diseases, INRA, 1260, Université de la Méditerranée, Faculté de Médecine, 27 Bd Jean Moulin, Marseille, France
| | | | | | | | - Jose M. Ordovas
- Nutrition and Genomics Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Catherine Defoort
- INSERM 476, Lipid nutrients and prevention of metabolic diseases, INRA, 1260, Université de la Méditerranée, Faculté de Médecine, 27 Bd Jean Moulin, Marseille, France
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food Biosciences, Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Christian A. Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway
| | | | - Ellen E. Blaak
- Department of Human Biology, Nutrition and Toxicology Research Institute Maastricht, Maastricht, The Netherlands
| | - Beata Kiec-Wilk
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Kopernika 15A, Krakow, Poland
| | - Britta Karlstrom
- Department of Public Health and Caring Sciences/Clinical Nutrition and Metabolism, Uppsala University, Uppsala Science Park, 751 85 Uppsala, Sweden
| | - Jose Lopez-Miranda
- Lipid and Atherosclerosis Unit, Department of Medicine, Reina Sofia University Hospital, School of Medicine, University of Cordoba, Spain
| | - Ross McManus
- Institute of Molecular Medicine, Trinity College Dublin, Ireland
| | - Serge Hercberg
- INSERM U557, INRA:CNAM, Université Paris 13, Bobigny, France
| | - Denis Lairon
- INSERM 476, Lipid nutrients and prevention of metabolic diseases, INRA, 1260, Université de la Méditerranée, Faculté de Médecine, 27 Bd Jean Moulin, Marseille, France
| | - Richard Planells
- INSERM 476, Lipid nutrients and prevention of metabolic diseases, INRA, 1260, Université de la Méditerranée, Faculté de Médecine, 27 Bd Jean Moulin, Marseille, France
| | - Helen M. Roche
- Nutrigenomics Research Group, UCD School of Public Health and Population Science, UCD Conway Institute, and Institute of Food and Health, University College Dublin, Ireland
| |
Collapse
|