351
|
Devisscher L, Verhelst X, Colle I, Van Vlierberghe H, Geerts A. The role of macrophages in obesity-driven chronic liver disease. J Leukoc Biol 2016; 99:693-8. [DOI: 10.1189/jlb.5ru0116-016r] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/12/2016] [Indexed: 12/21/2022] Open
|
352
|
Metformin Changes the Relationship between Blood Monocyte Toll-Like Receptor 4 Levels and Nonalcoholic Fatty Liver Disease-Ex Vivo Studies. PLoS One 2016; 11:e0150233. [PMID: 26930651 PMCID: PMC4773077 DOI: 10.1371/journal.pone.0150233] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 02/10/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Toll-like receptor 4 (TLR4) contributes to the development of NAFLD (nonalcoholic fatty liver disease) and MetS (metabolic syndrome). It is unclear whether anti-diabetic metformin affects TLR4 expression on blood monocytes, thereby protecting or improving inflammatory parameters. Therefore, we investigated TLR4 in patients with NAFLD meeting different sets of MetS criteria and linked the results with the disease burden. METHODS 70 subjects were characterized and divided into three groups: (I) healthy individuals, (II) nonobese with NAFLD and without MetS, and (III) prediabetic, obese with NAFLD and MetS. We determined the concentrations of IL-1β, IL-6, TNFα, and monocyte TLR4 levels in fresh blood as well as in blood cultures with or without metformin supplementation. RESULTS The characteristics of the study groups revealed a significant association between NAFLD and BMI, MetS and inflammatory parameters, and TLR4. In ex vivo studies, 100 μM of metformin decreased the TLR4 level by 19.9% (II group) or by 35% (III group) as well as IL-1β and TNFα production. A stepwise multiple regression analysis highlighted a strong effect of metformin on attenuation of the link between TLR4 and NAFLD, and TNFα. CONCLUSION We concluded that, by attenuation of the blood monocyte TLR4 level, metformin reduced their inflammatory potential-critical after recruitment these cells into liver. However, this finding should be confirmed after in vivo metformin administration.
Collapse
|
353
|
Lipid metabolism and signaling in cardiac lipotoxicity. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1513-24. [PMID: 26924249 DOI: 10.1016/j.bbalip.2016.02.016] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/19/2016] [Accepted: 02/19/2016] [Indexed: 01/01/2023]
Abstract
The heart balances uptake, metabolism and oxidation of fatty acids (FAs) to maintain ATP production, membrane biosynthesis and lipid signaling. Under conditions where FA uptake outpaces FA oxidation and FA sequestration as triacylglycerols in lipid droplets, toxic FA metabolites such as ceramides, diacylglycerols, long-chain acyl-CoAs, and acylcarnitines can accumulate in cardiomyocytes and cause cardiomyopathy. Moreover, studies using mutant mice have shown that dysregulation of enzymes involved in triacylglycerol, phospholipid, and sphingolipid metabolism in the heart can lead to the excess deposition of toxic lipid species that adversely affect cardiomyocyte function. This review summarizes our current understanding of lipid uptake, metabolism and signaling pathways that have been implicated in the development of lipotoxic cardiomyopathy under conditions including obesity, diabetes, aging, and myocardial ischemia-reperfusion. This article is part of a Special Issue entitled: Heart Lipid Metabolism edited by G.D. Lopaschuk.
Collapse
|
354
|
Pararasa C, Ikwuobe J, Shigdar S, Boukouvalas A, Nabney IT, Brown JE, Devitt A, Bailey CJ, Bennett SJ, Griffiths HR. Age-associated changes in long-chain fatty acid profile during healthy aging promote pro-inflammatory monocyte polarization via PPARγ. Aging Cell 2016; 15:128-39. [PMID: 26522807 PMCID: PMC4717269 DOI: 10.1111/acel.12416] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2015] [Indexed: 12/22/2022] Open
Abstract
Differences in lipid metabolism associate with age‐related disease development and lifespan. Inflammation is a common link between metabolic dysregulation and aging. Saturated fatty acids (FAs) initiate pro‐inflammatory signalling from many cells including monocytes; however, no existing studies have quantified age‐associated changes in individual FAs in relation to inflammatory phenotype. Therefore, we have determined the plasma concentrations of distinct FAs by gas chromatography in 26 healthy younger individuals (age < 30 years) and 21 healthy FA individuals (age > 50 years). Linear mixed models were used to explore the association between circulating FAs, age and cytokines. We showed that plasma saturated, poly‐ and mono‐unsaturated FAs increase with age. Circulating TNF‐α and IL‐6 concentrations increased with age, whereas IL‐10 and TGF‐β1 concentrations decreased. Oxidation of MitoSOX Red was higher in leucocytes from FA adults, and plasma oxidized glutathione concentrations were higher. There was significant colinearity between plasma saturated FAs, indicative of their metabolic relationships. Higher levels of the saturated FAs C18:0 and C24:0 were associated with lower TGF‐β1 concentrations, and higher C16:0 were associated with higher TNF‐α concentrations. We further examined effects of the aging FA profile on monocyte polarization and metabolism in THP1 monocytes. Monocytes preincubated with C16:0 increased secretion of pro‐inflammatory cytokines in response to phorbol myristate acetate‐induced differentiation through ceramide‐dependent inhibition of PPARγ activity. Conversely, C18:1 primed a pro‐resolving macrophage which was PPARγ dependent and ceramide dependent and which required oxidative phosphorylation. These data suggest that a midlife adult FA profile impairs the switch from proinflammatory to lower energy, requiring anti‐inflammatory macrophages through metabolic reprogramming.
Collapse
Affiliation(s)
- Chathyan Pararasa
- Life & Health Sciences Aston University Birmingham B4 7ET UK
- Aston Research Centre for Healthy Ageing Aston University Birmingham B4 7ET UK
| | - John Ikwuobe
- Life & Health Sciences Aston University Birmingham B4 7ET UK
| | | | | | - Ian T. Nabney
- Aston Research Centre for Healthy Ageing Aston University Birmingham B4 7ET UK
| | - James E. Brown
- Life & Health Sciences Aston University Birmingham B4 7ET UK
| | - Andrew Devitt
- Life & Health Sciences Aston University Birmingham B4 7ET UK
- Aston Research Centre for Healthy Ageing Aston University Birmingham B4 7ET UK
| | - Clifford J. Bailey
- Life & Health Sciences Aston University Birmingham B4 7ET UK
- Aston Research Centre for Healthy Ageing Aston University Birmingham B4 7ET UK
| | | | - Helen R. Griffiths
- Life & Health Sciences Aston University Birmingham B4 7ET UK
- Aston Research Centre for Healthy Ageing Aston University Birmingham B4 7ET UK
| |
Collapse
|
355
|
Ruiz-Núñez B, Dijck-Brouwer DAJ, Muskiet FAJ. The relation of saturated fatty acids with low-grade inflammation and cardiovascular disease. J Nutr Biochem 2016; 36:1-20. [PMID: 27692243 DOI: 10.1016/j.jnutbio.2015.12.007] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 12/03/2015] [Accepted: 12/16/2015] [Indexed: 12/15/2022]
Abstract
The mantra that dietary (saturated) fat must be minimized to reduce cardiovascular disease (CVD) risk has dominated nutritional guidelines for decades. Parallel to decreasing intakes of fat and saturated fatty acids (SFA), there have been increases in carbohydrate and sugar intakes, overweight, obesity and type 2 diabetes mellitus. The "lipid hypothesis" coined the concept that fat, especially SFA, raises blood low-density lipoprotein-cholesterol and thereby CVD risk. In view of current controversies regarding their adequate intakes and effects, this review aims to summarize research regarding this heterogenic group of fatty acids and the mechanisms relating them to (chronic) systemic low-grade inflammation, insulin resistance, metabolic syndrome and notably CVD. The intimate relationship between inflammation and metabolism, including glucose, fat and cholesterol metabolism, revealed that the dyslipidemia in Western societies, notably increased triglycerides, "small dense" low-density lipoprotein and "dysfunctional" high-density lipoprotein, is influenced by many unfavorable lifestyle factors. Dietary SFA is only one of these, not necessarily the most important, in healthy, insulin-sensitive people. The environment provides us not only with many other proinflammatory stimuli than SFA but also with many antiinflammatory counterparts. Resolution of the conflict between our self-designed environment and ancient genome may rather rely on returning to the proinflammatory/antiinflammatory balance of the Paleolithic era in consonance with the 21st century culture. Accordingly, dietary guidelines might reconsider recommendations for SFA replacement and investigate diet in a broader context, together with nondietary lifestyle factors. This should be a clear priority, opposed to the reductionist approach of studying the effects of single nutrients, such as SFA.
Collapse
Affiliation(s)
- Begoña Ruiz-Núñez
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - D A Janneke Dijck-Brouwer
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Frits A J Muskiet
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
356
|
Anitha M, Reichardt F, Tabatabavakili S, Nezami BG, Chassaing B, Mwangi S, Vijay-Kumar M, Gewirtz A, Srinivasan S. Intestinal dysbiosis contributes to the delayed gastrointestinal transit in high-fat diet fed mice. Cell Mol Gastroenterol Hepatol 2016; 2:328-339. [PMID: 27446985 PMCID: PMC4945127 DOI: 10.1016/j.jcmgh.2015.12.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS High-fat diet (HFD) feeding is associated with gastrointestinal motility disorders. We recently reported delayed colonic motility in mice fed a HFD mice for 11 weeks. In this study, we investigated the contributing role of gut microbiota in HFD-induced gut dysmotility. METHODS Male C57BL/6 mice were fed a HFD (60% kcal fat) or a regular/control diet (RD) (18% kcal fat) for 13 weeks. Serum and fecal endotoxin levels were measured, and relative amounts of specific gut bacteria in the feces assessed by real time PCR. Intestinal transit was measured by fluorescent-labeled marker and bead expulsion test. Enteric neurons were assessed by immunostaining. Oligofructose (OFS) supplementation with RD or HFD for 5 weeks was also studied. In vitro studies were performed using primary enteric neurons and an enteric neuronal cell line. RESULTS HFD-fed mice had reduced numbers of enteric nitrergic neurons and exhibited delayed gastrointestinal transit compared to RD-fed mice. HFD-fed mice had higher fecal Firmicutes and Escherichia coli and lower Bacteroidetes compared to RD-fed mice. OFS supplementation protected against enteric nitrergic neurons loss in HFD-fed mice, and improved intestinal transit time. OFS supplementation resulted in a reductions in fecal Firmicutes and Escherichia coli and serum endotoxin levels. In vitro, palmitate activation of TLR4 induced enteric neuronal apoptosis in a p-JNK1 dependent pathway. This apoptosis was prevented by a JNK inhibitor and in neurons from TLR4-/- mice. CONCLUSIONS Together our data suggest that intestinal dysbiosis in HFD fed mice contribute to the delayed intestinal motility by inducing a TLR4-dependant neuronal loss. Manipulation of gut microbiota with OFS improved intestinal motility in HFD mice.
Collapse
Affiliation(s)
- Mallappa Anitha
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - François Reichardt
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Sahar Tabatabavakili
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Behtash Ghazi Nezami
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Benoit Chassaing
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Simon Mwangi
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Matam Vijay-Kumar
- Department of Nutritional Sciences & Medicine, The Pennsylvania State University, University Park, Pennsylvania
| | - Andrew Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Shanthi Srinivasan
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia,Correspondence Address correspondence to: Shanthi Srinivasan, MD, Division of Digestive Diseases, Whitehead Biomedical Research Building, 615 Michael Street, Suite 201A, Atlanta, Georgia 30322. fax: (404) 727-5767.Division of Digestive DiseasesWhitehead Biomedical Research Building615 Michael StreetSuite 201AAtlantaGeorgia 30322
| |
Collapse
|
357
|
Han LP, Li CJ, Sun B, Xie Y, Guan Y, Ma ZJ, Chen LM. Protective Effects of Celastrol on Diabetic Liver Injury via TLR4/MyD88/NF-κB Signaling Pathway in Type 2 Diabetic Rats. J Diabetes Res 2016; 2016:2641248. [PMID: 27057550 PMCID: PMC4745324 DOI: 10.1155/2016/2641248] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 12/05/2015] [Accepted: 12/30/2015] [Indexed: 02/06/2023] Open
Abstract
Immune and inflammatory pathways play a central role in the pathogenesis of diabetic liver injury. Celastrol is a potent immunosuppressive and anti-inflammatory agent. So far, there is no evidence regarding the mechanism of innate immune alterations of celastrol on diabetic liver injury in type 2 diabetic animal models. The present study was aimed at investigating protective effects of celastrol on the liver injury in diabetic rats and at elucidating the possible involved mechanisms. We analyzed the liver histopathological and biochemical changes and the expressions of TLR4 mediated signaling pathway. Compared to the normal control group, diabetic rats were found to have obvious steatohepatitis and proinflammatory cytokine activities were significantly upregulated. Celastrol-treated diabetic rats show reduced hepatic inflammation and macrophages infiltration. The expressions of TLR4, MyD88, NF-κB, and downstream inflammatory factors IL-1β and TNFα in the hepatic tissue of treated rats were downregulated in a dose-dependent manner. We firstly found that celastrol treatment could delay the progression of diabetic liver disease in type 2 diabetic rats via inhibition of TLR4/MyD88/NF-κB signaling cascade pathways and its downstream inflammatory effectors.
Collapse
Affiliation(s)
- Li-ping Han
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Chun-jun Li
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Bei Sun
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Yun Xie
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Yue Guan
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Ze-jun Ma
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Li-ming Chen
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
- *Li-ming Chen:
| |
Collapse
|
358
|
Montserrat-de la Paz S, Naranjo MC, Bermudez B, Lopez S, Moreda W, Abia R, Muriana FJG. Postprandial dietary fatty acids exert divergent inflammatory responses in retinal-pigmented epithelium cells. Food Funct 2016; 7:1345-53. [DOI: 10.1039/c6fo00136j] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Postprandial triglyceride-rich lipoproteins (TRLs) lead to a complex series of events that are potentially oxidative and inflammatory.
Collapse
Affiliation(s)
| | - M. Carmen Naranjo
- Laboratory of Cellular and Molecular Nutrition
- Instituto de la Grasa
- CSIC
- 41013 Seville
- Spain
| | - Beatriz Bermudez
- Department of Pharmacology
- School of Pharmacy
- University of Seville
- 41012 Seville
- Spain
| | - Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition
- Instituto de la Grasa
- CSIC
- 41013 Seville
- Spain
| | - Wenceslao Moreda
- Laboratory of Olive Oil Quality
- Purity and Technology
- Instituto de la Grasa
- CSIC
- 41013 Seville
| | - Rocio Abia
- Laboratory of Cellular and Molecular Nutrition
- Instituto de la Grasa
- CSIC
- 41013 Seville
- Spain
| | | |
Collapse
|
359
|
Sutter AG, Palanisamy AP, Lench JH, Esckilsen S, Geng T, Lewin DNB, Cowart LA, Chavin KD. Dietary Saturated Fat Promotes Development of Hepatic Inflammation Through Toll-Like Receptor 4 in Mice. J Cell Biochem 2015; 117:1613-21. [PMID: 26600310 DOI: 10.1002/jcb.25453] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 11/23/2015] [Indexed: 12/15/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) is currently the third most common cause of end stage liver disease necessitating transplantation. The question remains how inflammation and NASH develop in the setting of nonalcoholic fatty liver disease (NAFLD) and steatosis. Understand the roles of toll-like receptor 4 (TLR4) and dietary fats in the development of hepatic inflammation. Wild-type and TLR4 KO mice were fed a standard high fat diet (LD), a high saturated fat diet (MD), or an isocaloric control diet (CD). Sera and tissue were analyzed for development of hepatic steatosis, inflammation, and injury. MD induced features of hepatic steatosis and inflammation in wild-type, but not in TLR4 KO, mice. TLR4 KO prevented MD induced increases in NAFLD activity scores, serum alanine aminotransferase levels, and inflammatory cytokine expression. Inflammatory cell infiltration and cytokine expression were also lower in the TLR4 KO mice livers than wild-type mice fed MD. Hepatic expression of Collagen I transcripts and collagen deposition were also decreased in the TLR4 KO MD animals. Results show that TLR4 plays a critical role in the effects of dietary fat composition on the development of hepatic steatosis, inflammation, and injury consistent with nonalcoholic steatohepatitis. J. Cell. Biochem. 117: 1613-1621, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alton G Sutter
- Division of Transplant Surgery, Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Arun P Palanisamy
- Division of Transplant Surgery, Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Julie H Lench
- Division of Transplant Surgery, Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Scott Esckilsen
- Division of Transplant Surgery, Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Tuoyu Geng
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - David N B Lewin
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Lauren A Cowart
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Kenneth D Chavin
- Division of Transplant Surgery, Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
360
|
Schuett KA, Lehrke M, Marx N, Burgmaier M. High-Risk Cardiovascular Patients: Clinical Features, Comorbidities, and Interconnecting Mechanisms. Front Immunol 2015; 6:591. [PMID: 26635805 PMCID: PMC4655316 DOI: 10.3389/fimmu.2015.00591] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/03/2015] [Indexed: 01/04/2023] Open
Abstract
Cardiovascular disease is the leading cause of death in the Western world with an increase over the last few decades. Atherosclerosis with its different manifestations in the coronary artery tree, the cerebral, as well as peripheral arteries is the basis for cardiovascular events, such as myocardial infarction, stroke, and cardiovascular death. The pathophysiological understanding of the mechanisms that promote the development of vascular disease has changed over the last few decades, leading to the recognition that inflammation and inflammatory processes in the vessel wall are major contributors in atherogenesis. In addition, a subclinical inflammatory status, e.g., in patients with diabetes or the presence of a chronic inflammatory disease, such as rheumatoid arthritis, have been recognized as strong risk factors for cardiovascular disease. The present review will summarize the different inflammatory processes in the vessel wall leading to atherosclerosis and highlight the role of inflammation in diabetes and chronic inflammatory diseases for cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital RWTH Aachen , Aachen , Germany
| | - Nikolaus Marx
- Department of Internal Medicine I, University Hospital RWTH Aachen , Aachen , Germany
| | - Mathias Burgmaier
- Department of Internal Medicine I, University Hospital RWTH Aachen , Aachen , Germany
| |
Collapse
|
361
|
Ghosh SS, Righi S, Krieg R, Kang L, Carl D, Wang J, Massey HD, Sica DA, Gehr TWB, Ghosh S. High Fat High Cholesterol Diet (Western Diet) Aggravates Atherosclerosis, Hyperglycemia and Renal Failure in Nephrectomized LDL Receptor Knockout Mice: Role of Intestine Derived Lipopolysaccharide. PLoS One 2015; 10:e0141109. [PMID: 26580567 PMCID: PMC4651339 DOI: 10.1371/journal.pone.0141109] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/04/2015] [Indexed: 01/06/2023] Open
Abstract
A high fat meal, frequently known as western diet (WD), exacerbates atherosclerosis and diabetes. Both these diseases are frequently associated with renal failure. Recent studies have shown that lipopolysaccharide (LPS) leaks into the circulation from the intestine in the setting of renal failure and after WD. However, it is not clear how renal function and associated disorders are affected by LPS. This study demonstrates that circulatory LPS exacerbates renal insufficiency, atherosclerosis and glucose intolerance. Renal insufficiency was induced by 2/3 nephrectomy in LDL receptor knockout mice. Nx animals were given normal diet (Nx) or WD (Nx+WD). The controls were sham operated animals on normal diet (control) and WD (WD). To verify if LPS plays a role in exaggerating renal insufficiency, polymyxin (PM), a known LPS antagonist, and curcumin (CU), a compound known to ameliorate chronic kidney disease (CKD), was given to Nx animals on western diet (Nx+WD+PM and Nx+WD+CU, respectively). Compared to control, all other groups displayed increased circulatory LPS. The Nx+WD cohort had the highest levels of LPS. Nx group had significant renal insufficiency and glucose intolerance but not atherosclerosis. WD had intense atherosclerosis and glucose intolerance but it did not show signs of renal insufficiency. Compared to other groups, Nx+WD had significantly higher cytokine expression, macrophage infiltration in the kidney, renal insufficiency, glucose intolerance and atherosclerosis. PM treatment blunted the expression of cytokines, deterioration of renal function and associated disorders, albeit not to the levels of Nx, and was significantly inferior to CU. PM is a non-absorbable antibiotic with LPS binding properties, hence its beneficial effect can only be due to its effect within the GI tract. We conclude that LPS may not cause renal insufficiency but can exaggerate kidney failure and associated disorders following renal insufficiency.
Collapse
Affiliation(s)
- Siddhartha S. Ghosh
- Divisions of Nephrology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Samuel Righi
- Divisions of Nephrology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Richard Krieg
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Le Kang
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Daniel Carl
- Divisions of Nephrology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Jing Wang
- Division of Pulmonary Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - H. Davis Massey
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Domenic A. Sica
- Divisions of Nephrology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Todd W. B. Gehr
- Divisions of Nephrology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Shobha Ghosh
- Division of Pulmonary Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
362
|
Snodgrass RG, Boß M, Zezina E, Weigert A, Dehne N, Fleming I, Brüne B, Namgaladze D. Hypoxia Potentiates Palmitate-induced Pro-inflammatory Activation of Primary Human Macrophages. J Biol Chem 2015; 291:413-24. [PMID: 26578520 DOI: 10.1074/jbc.m115.686709] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Indexed: 12/17/2022] Open
Abstract
Pro-inflammatory cytokines secreted by adipose tissue macrophages (ATMs) contribute to chronic low-grade inflammation and obesity-induced insulin resistance. Recent studies have shown that adipose tissue hypoxia promotes an inflammatory phenotype in ATMs. However, our understanding of how hypoxia modulates the response of ATMs to free fatty acids within obese adipose tissue is limited. We examined the effects of hypoxia (1% O2) on the pro-inflammatory responses of human monocyte-derived macrophages to the saturated fatty acid palmitate. Compared with normoxia, hypoxia significantly increased palmitate-induced mRNA expression and protein secretion of IL-6 and IL-1β. Although palmitate-induced endoplasmic reticulum stress and nuclear factor κB pathway activation were not enhanced by hypoxia, hypoxia increased the activation of JNK and p38 mitogen-activated protein kinase signaling in palmitate-treated cells. Inhibition of JNK blocked the hypoxic induction of pro-inflammatory cytokine expression, whereas knockdown of hypoxia-induced transcription factors HIF-1α and HIF-2α alone or in combination failed to reduce IL-6 and only modestly reduced IL-1β gene expression in palmitate-treated hypoxic macrophages. Enhanced pro-inflammatory cytokine production and JNK activity under hypoxia were prevented by inhibiting reactive oxygen species generation. In addition, silencing of dual-specificity phosphatase 16 increased normoxic levels of IL-6 and IL-1β and reduced the hypoxic potentiation in palmitate-treated macrophages. The secretome of hypoxic palmitate-treated macrophages promoted IL-6 and macrophage chemoattractant protein 1 expression in primary human adipocytes, which was sensitive to macrophage JNK inhibition. Our results reveal that the coexistence of hypoxia along with free fatty acids exacerbates macrophage-mediated inflammation.
Collapse
Affiliation(s)
| | - Marcel Boß
- From the Institute of Biochemistry I and
| | | | | | | | - Ingrid Fleming
- Institute for Vascular Signaling, Center for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | | | | |
Collapse
|
363
|
|
364
|
Feldman N, Rotter-Maskowitz A, Okun E. DAMPs as mediators of sterile inflammation in aging-related pathologies. Ageing Res Rev 2015; 24:29-39. [PMID: 25641058 DOI: 10.1016/j.arr.2015.01.003] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 12/29/2014] [Accepted: 01/19/2015] [Indexed: 12/20/2022]
Abstract
Accumulating evidence indicates that aging is associated with a chronic low-level inflammation, termed sterile-inflammation. Sterile-inflammation is a form of pathogen-free inflammation caused by mechanical trauma, ischemia, stress or environmental conditions such as ultra-violet radiation. These damage-related stimuli induce the secretion of molecular agents collectively termed danger-associated molecular patterns (DAMPs). DAMPs are recognized by virtue of specialized innate immune receptors, such as toll-like receptors (TLRs) and NOD-like receptor family, pyrin domain containing 3 (NLRP3). These receptors initiate signal transduction pathways, which typically drive inflammation in response to microbe-associated molecular patterns (MAMPs) and/or DAMPs. This review summarizes the current knowledge on DAMPs-mediated sterile-inflammation, its associated downstream signaling, and discusses the possibility that DAMPs activating TLRs or NLRP3 complex mediate sterile inflammation during aging and in aging-related pathologies.
Collapse
Affiliation(s)
- Noa Feldman
- The Mina and Everard Goodman Faculty of Life Sciences, The Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan 5290002, Israel
| | - Aviva Rotter-Maskowitz
- The Mina and Everard Goodman Faculty of Life Sciences, The Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan 5290002, Israel
| | - Eitan Okun
- The Mina and Everard Goodman Faculty of Life Sciences, The Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan 5290002, Israel.
| |
Collapse
|
365
|
Gillet C, Spruyt D, Rigutto S, Dalla Valle A, Berlier J, Louis C, Debier C, Gaspard N, Malaisse WJ, Gangji V, Rasschaert J. Oleate Abrogates Palmitate-Induced Lipotoxicity and Proinflammatory Response in Human Bone Marrow-Derived Mesenchymal Stem Cells and Osteoblastic Cells. Endocrinology 2015; 156:4081-93. [PMID: 26327577 DOI: 10.1210/en.2015-1303] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Osteoporosis is a metabolic bone disease associated with unequilibrated bone remodeling resulting from decreased bone formation and/or increased bone resorption, leading to progressive bone loss. In osteoporotic patients, low bone mass is associated with an increase of bone marrow fat resulting from accumulation of adipocytes within the bone marrow. Marrow adipocytes are active secretory cells, releasing cytokines, adipokines and free fatty acids (FA) that influence the bone marrow microenvironment and alter the biology of neighboring cells. Therefore, we examined the effect of palmitate (Palm) and oleate (Ole), 2 highly prevalent FA in human organism and diet, on the function and survival of human mesenchymal stem cells (MSC) and MSC-derived osteoblastic cells. The saturated FA Palm exerted a cytotoxic action via initiation of endoplasmic reticulum stress and activation of the nuclear factor κB (NF-κB) and ERK pathways. In addition, Palm induced a proinflammatory response, as determined by the up-regulation of Toll-like receptor 4 expression as well as the increase of IL-6 and IL-8 expression and secretion. Moreover, we showed that MSC-derived osteoblastic cells were more sensitive to lipotoxicity than undifferentiated MSC. The monounsaturated FA Ole fully neutralized Palm-induced lipotoxicity by impairing activation of the pathways triggered by the saturated FA. Moreover, Ole promoted Palm detoxification by fostering its esterification into triglycerides and storage in lipid droplets. Altogether, our data showed that physiological concentrations of Palm and Ole differently modulated cell death and function in bone cells. We therefore propose that FA could influence skeletal health.
Collapse
Affiliation(s)
- C Gillet
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - D Spruyt
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - S Rigutto
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - A Dalla Valle
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - J Berlier
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - C Louis
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - C Debier
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - N Gaspard
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - W J Malaisse
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - V Gangji
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - J Rasschaert
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| |
Collapse
|
366
|
Kim JA, Jang HJ, Hwang DH. Toll-like receptor 4-induced endoplasmic reticulum stress contributes to impairment of vasodilator action of insulin. Am J Physiol Endocrinol Metab 2015; 309:E767-76. [PMID: 26522062 PMCID: PMC4628943 DOI: 10.1152/ajpendo.00369.2015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/02/2015] [Indexed: 12/28/2022]
Abstract
Impairment of vasodilator action of insulin is associated with endothelial dysfunction and insulin resistance. Activation of Toll-like receptor 4 (TLR4) induces proinflammatory response and endoplasmic reticulum (ER) stress. Saturated fatty acids (SFA) activate TLR4, which induces ER stress and endothelial dysfunction. Therefore, we determined whether TLR4-mediated ER stress is an obligatory step mediating SFA-induced endothelial dysfunction. Palmitate stimulated proinflammatory responses and ER stress, and this was suppressed by knockdown of TLR4 in primary human aortic endothelial cells (HAEC). Next, we examined the role of TLR4 in vasodilatory responses in intact vessels isolated from wild-type (WT, C57BL/6) and TLR4-KO mice after feeding high-fat (HFD) or normal chow diet (NCD) for 12 wk. Arterioles isolated from HFD WT mice exhibited impaired insulin-stimulated vasodilation compared with arterioles isolated from NCD WT mice. Deficiency of TLR4 was protective from HFD-induced impairment of insulin-stimulated vasodilation. There were no differences in acetylcholine (Ach)- or sodium nitroprusside (SNP)-stimulated vasodilation between the two groups. Furthermore, we examined whether ER stress is involved in SFA-induced impairment of vasodilator actions of insulin. Infusion of palmitate showed the impairment of vasodilatory response to insulin, which was ameliorated by coinfusion with tauroursodeoxycholic acid (TUDCA), an ER stress suppressor. Taken together, the results suggest that TLR4-induced ER stress may be an obligatory step mediating the SFA-mediated endothelial dysfunction.
Collapse
Affiliation(s)
- Jeong-A Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama; University of Alabama at Birmingham Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Hyun-Ju Jang
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel H Hwang
- Western Human Nutrition Research Center, United States Department of Agriculture, and Department of Nutrition, University of California, Davis, Davis, California
| |
Collapse
|
367
|
Caspar-Bauguil S, Kolditz CI, Lefort C, Vila I, Mouisel E, Beuzelin D, Tavernier G, Marques MA, Zakaroff-Girard A, Pecher C, Houssier M, Mir L, Nicolas S, Moro C, Langin D. Fatty acids from fat cell lipolysis do not activate an inflammatory response but are stored as triacylglycerols in adipose tissue macrophages. Diabetologia 2015; 58:2627-36. [PMID: 26245186 DOI: 10.1007/s00125-015-3719-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/09/2015] [Indexed: 01/28/2023]
Abstract
AIMS/HYPOTHESIS Activation of macrophages by fatty acids (FAs) is a potential mechanism linking obesity to adipose tissue (AT) inflammation and insulin resistance. Here, we investigated the effects of FAs released during adipocyte lipolysis on AT macrophages (ATMs). METHODS Human THP-1 macrophages were treated with media from human multipotent adipose-derived stem (hMADS) adipocytes stimulated with lipolytic drugs. Macrophages were also treated with mixtures of FAs and an inhibitor of Toll-like receptor 4, since this receptor is activated by saturated FAs. Levels of mRNA and the secretion of inflammation-related molecules were measured in macrophages. FA composition was determined in adipocytes, conditioned media and macrophages. The effect of chronic inhibition or acute activation of fat cell lipolysis on ATM response was investigated in vivo in mice. RESULTS Whereas palmitic acid alone activates THP-1, conditioned media from hMADS adipocyte lipolysis had no effect on IL, chemokine and cytokine gene expression, and secretion by macrophages. Mixtures of FAs representing de novo lipogenesis or habitual dietary conditions also had no effect. FAs derived from adipocyte lipolysis were taken up by macrophages and stored as triacylglycerol droplets. In vivo, chronic treatment with an antilipolytic drug did not modify gene expression and number of ATMs in mice with intact or defective Tlr4. Stimulation of adipocyte lipolysis increased storage of neutral lipids by macrophages without change in number and phenotype. CONCLUSIONS/INTERPRETATION Our data suggest that adipocyte lipolysis does not activate inflammatory pathways in ATMs, which instead may act as scavengers of FAs.
Collapse
Affiliation(s)
- Sylvie Caspar-Bauguil
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
- Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, France
| | - Catherine-Ines Kolditz
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Corinne Lefort
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Isabelle Vila
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Etienne Mouisel
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Diane Beuzelin
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Geneviève Tavernier
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Marie-Adeline Marques
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Alexia Zakaroff-Girard
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
- Inserm, UMR1048, Cytometry Facility, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Inserm, UMR1048, Team 1, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Christiane Pecher
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
- Inserm, UMR1048, Cytometry Facility, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Marianne Houssier
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Lucile Mir
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Sarah Nicolas
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Cédric Moro
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Dominique Langin
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France.
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France.
- Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, France.
| |
Collapse
|
368
|
Intereukin-10 and Kupffer cells protect steatotic mice livers from ischemia-reperfusion injury. Eur Cytokine Netw 2015; 25:69-76. [PMID: 25679269 DOI: 10.1684/ecn.2015.0359] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Steatotic livers are more sensitive to ischemia/reperfusion (I/R) and are thus routinely rejected for transplantation because of their increased rate of primary nonfunction (PNF). Lean livers have less I/R-induced damage and inflammation due to Kupffer cells (KC), which are protective after total, warm, hepatic I/R with associated bowel congestion. This protection has been linked to KC-dependent expression of the potent anti-inflammatory cytokine interleukin-10 (IL-10). We hypothesized that pretreatment with exogenous IL-10 would protect the steatotic livers of genetically obese (ob/ob) mice from inflammation and injury induced by I/R. Lean and ob/ob mice were pretreated with either IL-10 or liposomally-encapsulated bisphosphonate clodronate (shown to deplete KC) prior to total, warm, hepatic I/R. IL-10 pretreatment increased survival of ob/ob animals at 24 hrs post-I/R from 30% to 100%, and significantly decreased serum ALT levels. At six hrs post-I/R, IL-10 pretreatment increased IL-10 mRNA expression, but suppressed up-regulation of the pro-inflammatory cytokine IL-1β mRNA. However, ALT levels were elevated at six hrs post-I/R in KC-depleted animals. These data reveal that pretreatment with IL-10 protects steatotic livers undergoing I/R, and that phagocytically active KC retain a hepatoprotective role in the steatotic environment.
Collapse
|
369
|
Caesar R, Tremaroli V, Kovatcheva-Datchary P, Cani PD, Bäckhed F. Crosstalk between Gut Microbiota and Dietary Lipids Aggravates WAT Inflammation through TLR Signaling. Cell Metab 2015; 22:658-68. [PMID: 26321659 PMCID: PMC4598654 DOI: 10.1016/j.cmet.2015.07.026] [Citation(s) in RCA: 724] [Impact Index Per Article: 72.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 05/25/2015] [Accepted: 07/27/2015] [Indexed: 12/13/2022]
Abstract
Dietary lipids may influence the abundance of circulating inflammatory microbial factors. Hence, inflammation in white adipose tissue (WAT) induced by dietary lipids may be partly dependent on their interaction with the gut microbiota. Here, we show that mice fed lard for 11 weeks have increased Toll-like receptor (TLR) activation and WAT inflammation and reduced insulin sensitivity compared with mice fed fish oil and that phenotypic differences between the dietary groups can be partly attributed to differences in microbiota composition. Trif(-/-) and Myd88(-/-) mice are protected against lard-induced WAT inflammation and impaired insulin sensitivity. Experiments in germ-free mice show that an interaction between gut microbiota and saturated lipids promotes WAT inflammation independent of adiposity. Finally, we demonstrate that the chemokine CCL2 contributes to microbiota-induced WAT inflammation in lard-fed mice. These results indicate that gut microbiota exacerbates metabolic inflammation through TLR signaling upon challenge with a diet rich in saturated lipids.
Collapse
Affiliation(s)
- Robert Caesar
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden.
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Petia Kovatcheva-Datchary
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Patrice D Cani
- Université Catholique de Louvain, Louvain Drug Research Institute, Metabolism and Nutrition Research Group, WELBIO (Walloon Excellence in Life Sciences and BIOtechnology), 1200 Brussels, Belgium
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
370
|
Wang H, Zhang Q, Chai Y, Liu Y, Li F, Wang B, Zhu C, Cui J, Qu H, Zhu M. 1,25(OH)2D3 downregulates the Toll-like receptor 4-mediated inflammatory pathway and ameliorates liver injury in diabetic rats. J Endocrinol Invest 2015; 38:1083-91. [PMID: 25906757 PMCID: PMC4768236 DOI: 10.1007/s40618-015-0287-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/03/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND Fatty acid deposition in the liver can activate a number of pro-inflammatory signaling pathways such as the Toll-like receptor 4 (TLR4) pathway, which may be important in the pathogenesis of nonalcoholic steatohepatitis. 1,25(OH)2D3 downregulates the expression of TLR4 and may represent a novel treatment strategy for reducing hepatocyte injury. Therefore, in this study, we investigated the protective effects of 1,25(OH)2D3 on diabetic liver injury in vivo. METHODS Streptozotocin (STZ)-induced diabetic rats were randomly divided into five groups and treated with low-dose 1,25(OH)2D3 (0.025 μg/kg/day), medium-dose 1,25(OH)2D3 (0.15 μg/kg/day), high-dose 1,25(OH)2D3 (0.3 μg/kg/day), insulin (protamine zinc insulin 16 U/kg/day, subcutaneous injection), or no intervention (the control group). Sixteen weeks later, the rats were killed, and blood samples were obtained to test lipid profiles and hepatic function. The infiltration of inflammatory cells, the level of fibrosis, and the expression levels of TLR4, nuclear factor-kappa B (NF-κB), and tumor necrosis factor-α (TNF-α) in the liver were analyzed. The hepatocytes were treated with vehicle control, LPS (100 ng), high fat [DMEM + FFA (0.1 mM: palmitic acid, oleic acid, 1:2)], LPS + high fat, vehicle + 1,25(OH)2D3 (10(-7) M), LPS + 1,25(OH)2D3, high fat + 1,25(OH)2D3, or LPS + high fat + 1,25(OH)2D3. RNA and protein were extracted to detect the expression of TLR4 and downstream inflammatory factors such as NF-ΚB, TNF-α, and IL-6. Groups of data were compared by single factor variance analysis. RESULTS High-dose 1,25(OH)2D3 administration for 16 weeks downregulated the expression of TLR4, NF-κB, and TNF-α in the liver tissue of diabetic rats and attenuated hepatic inflammation and fibrosis, as shown by immunohistochemical staining, hematoxylin and eosin staining, Masson's trichrome staining, reverse transcription polymerase chain reaction (RT-PCR), and western blotting. In vitro, hepatocytes treated with high fat or LPS exhibited significantly increased expression of TLR4, NF-κB, and downstream inflammatory factors (P < 0.05). Intervention with 1,25(OH)2D3 decreased the expression of TLR4, NF-κB, and inflammatory factors (P < 0.05). CONCLUSIONS 1,25(OH)2D3 exhibited protective effects against diabetes-related liver injury, possibly through downregulation of components of the TLR4 signaling pathway.
Collapse
Affiliation(s)
- H Wang
- Department of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Q Zhang
- Department of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Y Chai
- Department of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Y Liu
- Department of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - F Li
- Department of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - B Wang
- Department of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - C Zhu
- Department of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - J Cui
- Department of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - H Qu
- Division of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, TX, 77030, USA
| | - M Zhu
- Department of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
371
|
Moreno-Navarrete JM, Escoté X, Ortega F, Camps M, Ricart W, Zorzano A, Vendrell J, Vidal-Puig A, Fernández-Real JM. Lipopolysaccharide binding protein is an adipokine involved in the resilience of the mouse adipocyte to inflammation. Diabetologia 2015. [PMID: 26201685 DOI: 10.1007/s00125-015-3692-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
AIMS/HYPOTHESIS Lipopolysaccharide (LPS) binding protein (LBP) is a novel 65 kDa adipokine, linked to adipose tissue (AT) inflammation, obesity and insulin resistance, that inhibits adipocyte differentiation. Here, we investigated the molecular mechanisms behind these detrimental effects on adipogenesis through whole-genome transcriptomics and in vitro experiments. METHODS Permanent and transient knockdown (KD) and co-culture experiments were performed in 3T3-L1 and 3T3-F442A cell lines during adipocyte differentiation. Microarray gene expression was performed using Genechip Affymetrix technology and validated by real-time PCR. RESULTS LBP KD of 3T3-L1 cells led to a potentiated adipocyte differentiation with a dose-response relationship; genes involved in mitochondrial biogenesis, fatty acid metabolism and peroxisome proliferator-activated receptor γ (PPAR-γ) action were dramatically upregulated in parallel to increased insulin signalling. Cells with LBP KD became refractory to proinflammatory cytokines and other inflammatory stimuli (LPS and palmitate). This phenotype, mediated through disrupted nuclear factor κB (NFκB) signalling, was reversed by a soluble factor present in a co-culture with native cells and by exogenous LBP. Double-silencing of LBP and toll-like receptor 4 (TLR4) again rendered these cells insensitive to co-culture, LBP and inflammatory factors. CONCLUSIONS/INTERPRETATION In summary, LBP is a proinflammatory soluble adipokine that acts as a brake for adipogenesis, strengthening the negative effects of palmitate and LPS on adipocyte differentiation.
Collapse
Affiliation(s)
- José María Moreno-Navarrete
- Section of Diabetes, Endocrinology and Nutrition, Hospital of Girona 'Dr Josep Trueta', Carretera de França s/n, 17007, Girona, Spain
- Institut d'Investigació Biomèdica de Girona (IdIBGi), Girona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain
| | - Xavier Escoté
- Department of Endocrinology, Hospital Joan XXIII, Rovira i Virgili University, Tarragona, Spain
- Institut d'Investigació Sanitaria Pere Virgili (IISPV), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Francisco Ortega
- Section of Diabetes, Endocrinology and Nutrition, Hospital of Girona 'Dr Josep Trueta', Carretera de França s/n, 17007, Girona, Spain
- Institut d'Investigació Biomèdica de Girona (IdIBGi), Girona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain
| | - Marta Camps
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Wifredo Ricart
- Section of Diabetes, Endocrinology and Nutrition, Hospital of Girona 'Dr Josep Trueta', Carretera de França s/n, 17007, Girona, Spain
- Institut d'Investigació Biomèdica de Girona (IdIBGi), Girona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain
| | - Antonio Zorzano
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institute for Research in Biomedicine (IRB), Barcelona, Spain
| | - Joan Vendrell
- Department of Endocrinology, Hospital Joan XXIII, Rovira i Virgili University, Tarragona, Spain
- Institut d'Investigació Sanitaria Pere Virgili (IISPV), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Antonio Vidal-Puig
- Department of Clinical Biochemistry, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - José Manuel Fernández-Real
- Section of Diabetes, Endocrinology and Nutrition, Hospital of Girona 'Dr Josep Trueta', Carretera de França s/n, 17007, Girona, Spain.
- Institut d'Investigació Biomèdica de Girona (IdIBGi), Girona, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain, .
| |
Collapse
|
372
|
Abstract
Perturbations in metabolic pathways can cause substantial increases in plasma and tissue concentrations of long-chain acylcarnitines (LCACs). For example, the levels of LCACs and other acylcarnitines rise in the blood and muscle during exercise, as changes in tissue pools of acyl-coenzyme A reflect accelerated fuel flux that is incompletely coupled to mitochondrial energy demand and capacity of the tricarboxylic acid cycle. This natural ebb and flow of acylcarnitine generation and accumulation contrasts with that of inherited fatty acid oxidation disorders (FAODs), cardiac ischaemia or type 2 diabetes mellitus. These conditions are characterized by very high (FAODs, ischaemia) or modestly increased (type 2 diabetes mellitus) tissue and blood levels of LCACs. Although specific plasma concentrations of LCACs and chain-lengths are widely used as diagnostic markers of FAODs, research into the potential effects of excessive LCAC accumulation or the roles of acylcarnitines as physiological modulators of cell metabolism is lacking. Nevertheless, a growing body of evidence has highlighted possible effects of LCACs on disparate aspects of pathophysiology, such as cardiac ischaemia outcomes, insulin sensitivity and inflammation. This Review, therefore, aims to provide a theoretical framework for the potential consequences of tissue build-up of LCACs among individuals with metabolic disorders.
Collapse
Affiliation(s)
- Colin S McCoin
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Trina A Knotts
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA
| | - Sean H Adams
- Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, 15 Children's Way, Little Rock, AR 72202, USA
| |
Collapse
|
373
|
Navarro E, Funtikova AN, Fíto M, Schröder H. Can metabolically healthy obesity be explained by diet, genetics, and inflammation? Mol Nutr Food Res 2015; 59:75-93. [PMID: 25418549 DOI: 10.1002/mnfr.201400521] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/18/2014] [Accepted: 11/18/2014] [Indexed: 12/20/2022]
Abstract
A substantial proportion of obese individuals do not present cardiometabolic complications such as diabetes, hypertension, or dyslipidemia. Some, but not all, prospective studies observe similar risk of cardiovascular events and all-cause mortality among individuals with this so-called "metabolically healthy obese" (MHO) phenotype, compared to the metabolically healthy normal weight or metabolically healthy non-obese phenotypes. Compared to the metabolically unhealthy obese (MUO) phenotype, MHO is often characterized by a more favorable inflammatory profile, less visceral fat, less infiltration of macrophages into adipose tissue, and smaller adipocyte cell size. Tipping the inflammation balance in adipose tissue might be particularly important for metabolic health in the obese. While the potential role of genetic predisposition or lifestyle factors such as diet in the MHO phenotype is yet to be clarified, it is well known that diet affects inflammation profile and contributes to the functionality of adipose tissue. This review will discuss genetic predisposition and the molecular mechanisms underlying the potential effect of food on the development of the metabolic phenotype characteristic of obesity.
Collapse
|
374
|
The impact of early life gut colonization on metabolic and obesogenic outcomes: what have animal models shown us? J Dev Orig Health Dis 2015; 7:15-24. [DOI: 10.1017/s2040174415001518] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The rise in the occurrence of obesity to epidemic proportions has made it a global concern. Great difficulty has been experienced in efforts to control this growing problem with lifestyle interventions. Thus, attention has been directed to understanding the events of one of the most critical periods of development, perinatal life. Early life adversity driven by maternal obesity has been associated with an increased risk of metabolic disease and obesity in the offspring later in life. Although a mechanistic link explaining the relationship between maternal and offspring obesity is still under investigation, the gut microbiota has come forth as a new factor that may play a role modulating metabolic function of both the mother and the offspring. Emerging evidence suggests that the gut microbiota plays a much larger role in mediating the risk of developing non-communicable disease, including obesity and metabolic dysfunction in adulthood. With the observation that the early life colonization of the neonatal and postnatal gut is mediated by the perinatal environment, the number of studies investigating early life gut microbial establishment continues to grow. This paper will review early life gut colonization in experimental animal models, concentrating on the role of the early life environment in offspring gut colonization and the ability of the gut microbiota to dictate risk of disease later in life.
Collapse
|
375
|
Bleau C, Karelis AD, St-Pierre DH, Lamontagne L. Crosstalk between intestinal microbiota, adipose tissue and skeletal muscle as an early event in systemic low-grade inflammation and the development of obesity and diabetes. Diabetes Metab Res Rev 2015; 31:545-61. [PMID: 25352002 DOI: 10.1002/dmrr.2617] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 10/01/2014] [Accepted: 10/13/2014] [Indexed: 02/06/2023]
Abstract
Obesity is associated with a systemic chronic low-grade inflammation that contributes to the development of metabolic disorders such as cardiovascular diseases and type 2 diabetes. However, the etiology of this obesity-related pro-inflammatory process remains unclear. Most studies have focused on adipose tissue dysfunctions and/or insulin resistance in skeletal muscle cells as well as changes in adipokine profile and macrophage recruitment as potential sources of inflammation. However, low-grade systemic inflammation probably involves a complex network of signals interconnecting several organs. Recent evidences have suggested that disturbances in the composition of the gut microbial flora and alterations in levels of gut peptides following the ingestion of a high-fat diet may be a cause of low-grade systemic inflammation that may even precede and predispose to obesity, metabolic disorders or type 2 diabetes. This hypothesis is appealing because the gastrointestinal system is first exposed to nutrients and may thereby represent the first link in the chain of events leading to the development of obesity-associated systemic inflammation. Therefore, the present review will summarize the latest advances interconnecting intestinal mucosal bacteria-mediated inflammation, adipose tissue and skeletal muscle in a coordinated circuitry favouring the onset of a high-fat diet-related systemic low-grade inflammation preceding obesity and predisposing to metabolic disorders and/or type 2 diabetes. A particular emphasis will be given to high-fat diet-induced alterations of gut homeostasis as an early initiator event of mucosal inflammation and adverse consequences contributing to the promotion of extended systemic inflammation, especially in adipose and muscular tissues.
Collapse
MESH Headings
- Adipose Tissue, White/immunology
- Adipose Tissue, White/metabolism
- Animals
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/microbiology
- Diet, High-Fat/adverse effects
- Enteritis/etiology
- Enteritis/immunology
- Enteritis/microbiology
- Enteritis/physiopathology
- Gastrointestinal Hormones/metabolism
- Gastrointestinal Microbiome
- Humans
- Immunity, Mucosal
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/microbiology
- Models, Biological
- Muscle, Skeletal/immunology
- Muscle, Skeletal/metabolism
- Myositis/etiology
- Myositis/immunology
- Myositis/microbiology
- Myositis/physiopathology
- Obesity/etiology
- Obesity/immunology
- Obesity/metabolism
- Obesity/microbiology
- Panniculitis/etiology
- Panniculitis/immunology
- Panniculitis/microbiology
- Panniculitis/physiopathology
- Systemic Vasculitis/etiology
- Systemic Vasculitis/immunology
- Systemic Vasculitis/microbiology
- Systemic Vasculitis/physiopathology
Collapse
Affiliation(s)
- Christian Bleau
- Department of Biological Sciences, Université du Québec à Montréal, Montreal, Canada, H3C 3P8
| | - Antony D Karelis
- Department of Kinanthropology, Université du Québec à Montréal, Montreal, Canada, H3C 3P8
| | - David H St-Pierre
- Department of Kinanthropology, Université du Québec à Montréal, Montreal, Canada, H3C 3P8
| | - Lucie Lamontagne
- Department of Biological Sciences, Université du Québec à Montréal, Montreal, Canada, H3C 3P8
| |
Collapse
|
376
|
Oki E, Norde MM, Carioca AAF, Ikeda RE, Souza JMP, Castro IA, Marchioni DML, Fisberg RM, Rogero MM. Interaction of SNP in the CRP gene and plasma fatty acid profile in inflammatory pattern: A cross-sectional population-based study. Nutrition 2015; 32:88-94. [PMID: 26456189 DOI: 10.1016/j.nut.2015.07.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/18/2015] [Accepted: 07/29/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To assess the interaction of three single nucleotide polymorphisms in the C-reactive protein (CRP) gene and plasma fatty acid (FA) levels in modulating inflammatory profile. METHODS A total of 262 subjects, aged >19 y and <60 y, participated in a cross-sectional, population-based study performed in Brazil. Three single nucleotide polymorphisms (rs1205, rs1417938, and rs2808630) spanning the CRP gene were genotyped. Eleven plasma inflammatory biomarkers and plasma FA profile were determined. Cluster analysis was performed to stratify individuals based on eleven inflammatory biomarkers into two groups: an inflammatory (INF) and a noninflammatory group. RESULTS The INF cluster had higher age, waist circumference, systolic blood pressure, and diastolic blood pressure; higher levels of triacylglycerol, high-sensitivity CRP, tumor necrosis factor-α, interleukin (IL)-8, IL-6, IL-1β, IL-12, IL-10, soluble monocyte chemoattractant protein-1, soluble intercellular adhesion molecule-1, C16:0, polyunsaturated fatty acid, and omega (n)-6 polyunsaturated fatty acid; and greater C20:4n-6, C18:1/18:0, and C20:4/20:3 ratios than the noninflammatory group. Statistically significant gene-plasma C16:1n-7 interaction was detected for rs1417938 (P = 0.047). Those with a dominant homozygous rs2808630 had a lower risk of belonging to the INF group with the upper 50th percentile of C20:4n-6, n-3 highly unsaturated FA, and C20:4/20:3 ratio. Regarding rs1205, A allele carriers had lower risk of being in the INF group when C20:5n-3 and n-3 highly unsaturated FA levels were greater than the median. CONCLUSIONS The INF group exhibited changes in metabolic parameters that predispose this group to chronic disease, where polymorphisms in the CRP gene modulated the risk of being in the INF group depending on individual plasma fatty acid and lipid profile.
Collapse
Affiliation(s)
- Erica Oki
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Marina M Norde
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Antônio A F Carioca
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Renata E Ikeda
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - José M P Souza
- Department of Epidemiology, School of Public Health, University of São Paulo, Brazil
| | - Inar A Castro
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Brazil
| | - Dirce M L Marchioni
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Regina M Fisberg
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Marcelo M Rogero
- Nutrition Department, School of Public Health, University of São Paulo, Brazil.
| |
Collapse
|
377
|
de Courten B, Moreno-Navarrete JM, Lyons J, Soldatos G, de Courten M, Dougherty S, Forbes J, Fernández-Real JM. Contrasting association of circulating sCD14 with insulin sensitivity in non-obese and morbidly obese subjects. Mol Nutr Food Res 2015; 60:103-9. [PMID: 26114238 DOI: 10.1002/mnfr.201500102] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 05/21/2015] [Accepted: 05/26/2015] [Indexed: 01/22/2023]
Abstract
SCOPE In experimental studies, moderate to high concentrations of sCD14 (serum cluster of differentiation 14 protein) prevent lipopolysaccharide (LPS)-induced systemic inflammation, while low concentrations may promote inflammation. Given that metabolic endotoxemia is thought to initiate high-fat diet-induced insulin resistance, we explored the association between sCD14 concentrations and insulin sensitivity in humans. METHODS AND RESULTS Healthy non-obese (n = 12, BMI 26 ± 5y), obese (n = 11, BMI 33.45 ± 3.2) and morbidly obese participants (n = 38, BMI 45 ± 7) underwent measurement of body composition (dual energy X-ray absorptiometry) and a hyperinsulinemic-euglycemic clamp to measure insulin sensitivity (M value). Circulating sCD14 concentrations were measured by ELISA. Non-obese participants had lower circulating sCD14 concentrations compared to obese (p = 0.03). Circulating sCD14 concentrations were positively associated with percent body fat, waist circumference and white blood cell count and negatively associated with insulin sensitivity. In contrast, circulating sCD14 were positively associated with insulin sensitivity in morbidly obese participants. In regression analysis, insulin sensitivity (r = 0.52, p = 0.004) and fasting triglycerides (r = 0.49, p = 0.005) contributed independently to circulating sCD14 variance after controlling for age, sex and BMI in these morbidly obese subjects. CONCLUSION These findings suggest that circulating sCD14 concentrations, through its compensatory (in non-obese subjects) or buffering role (in morbidly obese subjects), could exert an important role in modulating insulin sensitivity.
Collapse
Affiliation(s)
- Barbora de Courten
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Monash Centre for Health, Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - José Maria Moreno-Navarrete
- University Hospital of Girona, Institut d'Investigació Biomèdica de Girona (IDIBGi) and CIBERobn Pathophysiology of Obesity and Nutrition, Spain
| | - Jasmine Lyons
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Georgia Soldatos
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Monash Centre for Health, Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Maximilian de Courten
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Sonia Dougherty
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Josephine Forbes
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia.,Mater Research at TRI, Woolloongabba, Australia.,Mater Clinical School, University of Queensland, St Lucia, Australia
| | - José Manuel Fernández-Real
- University Hospital of Girona, Institut d'Investigació Biomèdica de Girona (IDIBGi) and CIBERobn Pathophysiology of Obesity and Nutrition, Spain
| |
Collapse
|
378
|
Abstract
Acne vulgaris, an epidemic inflammatory skin disease of adolescence, is closely related to Western diet. Three major food classes that promote acne are: 1) hyperglycemic carbohydrates, 2) milk and dairy products, 3) saturated fats including trans-fats and deficient ω-3 polyunsaturated fatty acids (PUFAs). Diet-induced insulin/insulin-like growth factor (IGF-1)-signaling is superimposed on elevated IGF-1 levels during puberty, thereby unmasking the impact of aberrant nutrigenomics on sebaceous gland homeostasis. Western diet provides abundant branched-chain amino acids (BCAAs), glutamine, and palmitic acid. Insulin and IGF-1 suppress the activity of the metabolic transcription factor forkhead box O1 (FoxO1). Insulin, IGF-1, BCAAs, glutamine, and palmitate activate the nutrient-sensitive kinase mechanistic target of rapamycin complex 1 (mTORC1), the key regulator of anabolism and lipogenesis. FoxO1 is a negative coregulator of androgen receptor, peroxisome proliferator-activated receptor-γ (PPARγ), liver X receptor-α, and sterol response element binding protein-1c (SREBP-1c), crucial transcription factors of sebaceous lipogenesis. mTORC1 stimulates the expression of PPARγ and SREBP-1c, promoting sebum production. SREBP-1c upregulates stearoyl-CoA- and Δ6-desaturase, enhancing the proportion of monounsaturated fatty acids in sebum triglycerides. Diet-mediated aberrations in sebum quantity (hyperseborrhea) and composition (dysseborrhea) promote Propionibacterium acnes overgrowth and biofilm formation with overexpression of the virulence factor triglyceride lipase increasing follicular levels of free palmitate and oleate. Free palmitate functions as a "danger signal," stimulating toll-like receptor-2-mediated inflammasome activation with interleukin-1β release, Th17 differentiation, and interleukin-17-mediated keratinocyte proliferation. Oleate stimulates P. acnes adhesion, keratinocyte proliferation, and comedogenesis via interleukin-1α release. Thus, diet-induced metabolomic alterations promote the visible sebofollicular inflammasomopathy acne vulgaris. Nutrition therapy of acne has to increase FoxO1 and to attenuate mTORC1/SREBP-1c signaling. Patients should balance total calorie uptake and restrict refined carbohydrates, milk, dairy protein supplements, saturated fats, and trans-fats. A paleolithic-like diet enriched in vegetables and fish is recommended. Plant-derived mTORC1 inhibitors and ω-3-PUFAs are promising dietary supplements supporting nutrition therapy of acne vulgaris.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Germany
| |
Collapse
|
379
|
Olthof ED, Gülich AF, Renne MF, Landman S, Joosten LAB, Roelofs HMJ, Wanten GJA. Immune activation by medium-chain triglyceride-containing lipid emulsions is not modulated by n-3 lipids or toll-like receptor 4. Toxicol In Vitro 2015; 29:1851-8. [PMID: 26162596 DOI: 10.1016/j.tiv.2015.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 07/02/2015] [Accepted: 07/06/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Saturated medium-chain triglycerides (MCT) as part of the parenteral lipid regimen (50% MCT and 50% long chain triglycerides (LCT)) activate the immune system in vitro. Fish oil (FO)-derived n-3 fatty acids (FA) inhibit saturated FA-induced immune activation via a toll-like receptor (TLR)-4 mediated mechanism. We hypothesized that effects of parenteral MCTs on immune cells involve TLR-4 signaling and that these effects are modulated by n-3 FA that are present in FO. MATERIALS AND METHODS To test this hypothesis we assessed effects of addition of various commercially available mixed parenteral lipid emulsions, n-3 FA and of TLR-4 inhibition on MCT-induced human immune cell activation by evaluation of the expression of leukocyte membrane activation markers and reactive oxygen species (ROS) production. RESULTS All MCT-containing lipid emulsions activated leukocytes by inducing changes in expression of membrane markers and stimulus induced ROS production, whereas MCT-free lipid emulsions lacked this effect. Moreover, addition of n-3 FA to LCT/MCT did not prevent MCT-induced immune activation. TLR-4 inhibitors did not distinctly modulate MCT-induced changes in immune function. CONCLUSION Taken together, these findings suggest that leukocyte activation by parenteral MCTs does not involve TLR-4 signaling and is not modulated by n-3 FA in FO-, but is exerted via different signaling pathways.
Collapse
Affiliation(s)
- Evelyn D Olthof
- Intestinal Failure Unit, Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Alexandra F Gülich
- Intestinal Failure Unit, Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Mike F Renne
- Intestinal Failure Unit, Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands; Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research and Institute of Biomembranes, Utrecht University, Utrecht, The Netherlands.
| | - Sija Landman
- Intestinal Failure Unit, Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Laboratory Medicine - Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Hennie M J Roelofs
- Intestinal Failure Unit, Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Geert J A Wanten
- Intestinal Failure Unit, Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
380
|
Ciesielska A, Kwiatkowska K. Modification of pro-inflammatory signaling by dietary components: The plasma membrane as a target. Bioessays 2015; 37:789-801. [PMID: 25966354 DOI: 10.1002/bies.201500017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
You are what you eat - this well-known phrase properly describes the phenomenon of the effects of diet on acute and chronic inflammation. Several lipids and lipophilic compounds that are delivered with food or are produced in situ in pathological conditions exert immunomodulatory activity due to their interactions with the plasma membrane. This group of compounds includes cholesterol and its oxidized derivatives, fatty acids, α-tocopherol, and polyphenols. Despite their structural heterogeneity, all these compounds ultimately induce changes in plasma membrane architecture and fluidity. By doing this, they modulate the dynamics of plasma membrane receptors, such as TLR4. This receptor is activated by lipopolysaccharide, triggering acute inflammation during bacterial infection, which often leads to sepsis and is linked with diverse chronic inflammatory diseases. In this review, we discuss how the impact on plasma membrane properties contributes to the immunomodulatory activity of dietary compounds, pointing to the therapeutic potential of some of them. Also watch the Video Abstract.
Collapse
Affiliation(s)
- Anna Ciesielska
- Nencki Institute of Experimental Biology, Laboratory of Molecular Membrane Biology, Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Nencki Institute of Experimental Biology, Laboratory of Molecular Membrane Biology, Warsaw, Poland
| |
Collapse
|
381
|
Pomés A, Chruszcz M, Gustchina A, Minor W, Mueller GA, Pedersen LC, Wlodawer A, Chapman MD. 100 Years later: Celebrating the contributions of x-ray crystallography to allergy and clinical immunology. J Allergy Clin Immunol 2015; 136:29-37.e10. [PMID: 26145985 PMCID: PMC4502579 DOI: 10.1016/j.jaci.2015.05.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/21/2015] [Accepted: 05/14/2015] [Indexed: 01/07/2023]
Abstract
Current knowledge of molecules involved in immunology and allergic disease results from the significant contributions of x-ray crystallography, a discipline that just celebrated its 100th anniversary. The histories of allergens and x-ray crystallography are intimately intertwined. The first enzyme structure to be determined was lysozyme, also known as the chicken food allergen Gal d 4. Crystallography determines the exact 3-dimensional positions of atoms in molecules. Structures of molecular complexes in the disciplines of immunology and allergy have revealed the atoms involved in molecular interactions and mechanisms of disease. These complexes include peptides presented by MHC class II molecules, cytokines bound to their receptors, allergen-antibody complexes, and innate immune receptors with their ligands. The information derived from crystallographic studies provides insights into the function of molecules. Allergen function is one of the determinants of environmental exposure, which is essential for IgE sensitization. Proteolytic activity of allergens or their capacity to bind LPSs can also contribute to allergenicity. The atomic positions define the molecular surface that is accessible to antibodies. In turn, this surface determines antibody specificity and cross-reactivity, which are important factors for the selection of allergen panels used for molecular diagnosis and the interpretation of clinical symptoms. This review celebrates the contributions of x-ray crystallography to clinical immunology and allergy, focusing on new molecular perspectives that influence the diagnosis and treatment of allergic diseases.
Collapse
Affiliation(s)
- Anna Pomés
- Basic Research, INDOOR Biotechnologies, Charlottesville, Va.
| | - Maksymilian Chruszcz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC
| | - Alla Gustchina
- Macromolecular Crystallography Laboratory, National Cancer Institute, Frederick, Md
| | - Wladek Minor
- Department of Molecular Physiology and Biological Physic, University of Virginia, Charlottesville, Va
| | - Geoffrey A Mueller
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Lars C Pedersen
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Alexander Wlodawer
- Macromolecular Crystallography Laboratory, National Cancer Institute, Frederick, Md
| | | |
Collapse
|
382
|
McMillan RP, Wu Y, Voelker K, Fundaro G, Kavanaugh J, Stevens JR, Shabrokh E, Ali M, Harvey M, Anderson AS, Boutagy NE, Mynatt RL, Frisard MI, Hulver MW. Selective overexpression of Toll-like receptor-4 in skeletal muscle impairs metabolic adaptation to high-fat feeding. Am J Physiol Regul Integr Comp Physiol 2015; 309:R304-13. [PMID: 26084695 DOI: 10.1152/ajpregu.00139.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/11/2015] [Indexed: 12/22/2022]
Abstract
Toll-like receptor-4 (TLR-4) is elevated in skeletal muscle of obese humans, and data from our laboratory have shown that activation of TLR-4 in skeletal muscle via LPS results in decreased fatty acid oxidation (FAO). The purpose of this study was to determine whether overexpression of TLR-4 in skeletal muscle alters mitochondrial function and whole body metabolism in the context of a chow and high-fat diet. C57BL/6J mice (males, 6-8 mo of age) with skeletal muscle-specific overexpression of the TLR-4 (mTLR-4) gene were created and used for this study. Isolated mitochondria and whole muscle homogenates from rodent skeletal muscle (gastrocnemius and quadriceps) were investigated. TLR-4 overexpression resulted in a significant reduction in FAO in muscle homogenates; however, mitochondrial respiration and reactive oxygen species (ROS) production did not appear to be affected on a standard chow diet. To determine the role of TLR-4 overexpression in skeletal muscle in response to high-fat feeding, mTLR-4 mice and WT control mice were fed low- and high-fat diets for 16 wk. The high-fat diet significantly decreased FAO in mTLR-4 mice, which was observed in concert with elevated body weight and fat, greater glucose intolerance, and increase in production of ROS and cellular oxidative damage compared with WT littermates. These findings suggest that TLR-4 plays an important role in the metabolic response in skeletal muscle to high-fat feeding.
Collapse
Affiliation(s)
- Ryan P McMillan
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia; Metabolic Phenotyping Core, Virginia Tech, Blacksburg, Virginia; Fralin Translational Obesity Research Center, Virginia Tech, Blacksburg, Virginia
| | - Yaru Wu
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia
| | - Kevin Voelker
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia
| | - Gabrielle Fundaro
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia
| | - John Kavanaugh
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia
| | - Joseph R Stevens
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia; Fralin Translational Obesity Research Center, Virginia Tech, Blacksburg, Virginia
| | - Elika Shabrokh
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan; and
| | - Mostafa Ali
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia
| | - Mordecai Harvey
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia
| | - Angela S Anderson
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia
| | - Nabil E Boutagy
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia; Fralin Translational Obesity Research Center, Virginia Tech, Blacksburg, Virginia
| | - Randall L Mynatt
- Gene Nutrient Interactions, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Madlyn I Frisard
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia; Metabolic Phenotyping Core, Virginia Tech, Blacksburg, Virginia; Fralin Translational Obesity Research Center, Virginia Tech, Blacksburg, Virginia
| | - Matthew W Hulver
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia; Metabolic Phenotyping Core, Virginia Tech, Blacksburg, Virginia; Fralin Translational Obesity Research Center, Virginia Tech, Blacksburg, Virginia;
| |
Collapse
|
383
|
Chan KL, Pillon NJ, Sivaloganathan DM, Costford SR, Liu Z, Théret M, Chazaud B, Klip A. Palmitoleate Reverses High Fat-induced Proinflammatory Macrophage Polarization via AMP-activated Protein Kinase (AMPK). J Biol Chem 2015; 290:16979-88. [PMID: 25987561 DOI: 10.1074/jbc.m115.646992] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Indexed: 12/16/2022] Open
Abstract
A rise in tissue-embedded macrophages displaying "M1-like" proinflammatory polarization is a hallmark of metabolic inflammation during a high fat diet or obesity. Here we show that bone marrow-derived macrophages (BMDM) from high fat-fed mice retain a memory of their dietary environment in vivo (displaying the elevated proinflammatory genes Cxcl1, Il6, Tnf, Nos2) despite 7-day differentiation and proliferation ex vivo. Notably, 6-h incubation with palmitoleate (PO) reversed the proinflammatory gene expression and cytokine secretion seen in BMDM from high fat-fed mice. BMDM from low fat-fed mice exposed to palmitate (PA) for 18 h ex vivo also showed elevated expression of proinflammatory genes (Cxcl1, Il6, Tnf, Nos2, and Il12b) associated with M1 polarization. Conversely, PO treatment increased anti-inflammatory genes (Mrc1, Tgfb1, Il10, Mgl2) and oxidative metabolism, characteristic of M2 macrophages. Therefore, saturated and unsaturated fatty acids bring about opposite macrophage polarization states. Coincubation of BMDM with both fatty acids counteracted the PA-induced Nos2 expression in a PO dose-dependent fashion. PO also prevented PA-induced IκBα degradation, RelA nuclear translocation, NO production, and cytokine secretion. Mechanistically, PO exerted its anti-inflammatory function through AMP-activated protein kinase as AMP kinase knockout or inhibition by Compound C offset the PO-dependent prevention of PA-induced inflammation. These results demonstrate a nutritional memory of BMDM ex vivo, highlight the plasticity of BMDM polarization in response to saturated and unsaturated fatty acids, and identify the potential to reverse diet- and saturated fat-induced M1-like polarization by administering palmitoleate. These findings could have applicability to reverse obesity-linked inflammation in metabolically relevant tissues.
Collapse
Affiliation(s)
- Kenny L Chan
- From the Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada, the Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada, and
| | - Nicolas J Pillon
- From the Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | | | - Sheila R Costford
- From the Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Zhi Liu
- From the Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Marine Théret
- the Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, Villeurbanne 69622, France
| | - Benedicte Chazaud
- the Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, Villeurbanne 69622, France
| | - Amira Klip
- From the Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada, the Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada, and
| |
Collapse
|
384
|
Tilg H, Moschen AR. Food, immunity, and the microbiome. Gastroenterology 2015; 148:1107-19. [PMID: 25575570 DOI: 10.1053/j.gastro.2014.12.036] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 12/12/2022]
Abstract
There is increasing evidence that ingested diet-borne components are involved in the pathogenesis of disorders such as inflammatory bowel diseases, atherosclerosis, and type 2 diabetes. Nutrients can have short- and long-term effects in shaping the composition of the microbiota. Western diets (enriched in fat, phosphatidylcholine, and L-carnitine) promote inflammation and atherosclerosis through specific fatty acids and degradation products such as trimethylamine N-oxide. Other dietary factors such as carbazoles or tryptophan-enriched proteins have anti-inflammatory properties-partly via activation of aryl hydrocarbon receptors. The microbiota and its metabolic machinery produce a myriad of metabolites that serve as important messengers between the diet, microbiota, and host. Short-chain fatty acids affect immune responses and epithelial integrity via G-protein-coupled receptors and epigenetic mechanisms. By increasing our understanding of interactions between diet, immunity, and the microbiota, we might develop food-based approaches to prevent or treat many diseases. There now is scientific evidence to support the adage "we are what we eat," and this process begins in early life.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Endocrinology, Gastroenterology and Metabolism, Medical University Innsbruck, Innsbruck, Austria.
| | - Alexander R Moschen
- Department of Internal Medicine I, Endocrinology, Gastroenterology and Metabolism, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
385
|
Malandrino MI, Fucho R, Weber M, Calderon-Dominguez M, Mir JF, Valcarcel L, Escoté X, Gómez-Serrano M, Peral B, Salvadó L, Fernández-Veledo S, Casals N, Vázquez-Carrera M, Villarroya F, Vendrell JJ, Serra D, Herrero L. Enhanced fatty acid oxidation in adipocytes and macrophages reduces lipid-induced triglyceride accumulation and inflammation. Am J Physiol Endocrinol Metab 2015; 308:E756-69. [PMID: 25714670 DOI: 10.1152/ajpendo.00362.2014] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 02/19/2015] [Indexed: 12/31/2022]
Abstract
Lipid overload in obesity and type 2 diabetes is associated with adipocyte dysfunction, inflammation, macrophage infiltration, and decreased fatty acid oxidation (FAO). Here, we report that the expression of carnitine palmitoyltransferase 1A (CPT1A), the rate-limiting enzyme in mitochondrial FAO, is higher in human adipose tissue macrophages than in adipocytes and that it is differentially expressed in visceral vs. subcutaneous adipose tissue in both an obese and a type 2 diabetes cohort. These observations led us to further investigate the potential role of CPT1A in adipocytes and macrophages. We expressed CPT1AM, a permanently active mutant form of CPT1A, in 3T3-L1 CARΔ1 adipocytes and RAW 264.7 macrophages through adenoviral infection. Enhanced FAO in palmitate-incubated adipocytes and macrophages reduced triglyceride content and inflammation, improved insulin sensitivity in adipocytes, and reduced endoplasmic reticulum stress and ROS damage in macrophages. We conclude that increasing FAO in adipocytes and macrophages improves palmitate-induced derangements. This indicates that enhancing FAO in metabolically relevant cells such as adipocytes and macrophages may be a promising strategy for the treatment of chronic inflammatory pathologies such as obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Maria Ida Malandrino
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Raquel Fucho
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Minéia Weber
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - María Calderon-Dominguez
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Francesc Mir
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Lorea Valcarcel
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Xavier Escoté
- Endocrinology and Diabetes Unit, Joan XXIII University Hospital, Institut d'Investigació Sanitària Pere i Virgili, Universitat Rovira i Virgili, Tarragona, Spain; Institut de Biomedicina de la Universitat de Barcelona de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - María Gómez-Serrano
- Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Belén Peral
- Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Laia Salvadó
- Institut de Biomedicina de la Universitat de Barcelona de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain; Pharmacology Unit, Department of Pharmacology and Therapeutic Chemistry and Institut de Biomedicina de la Universitat de Barcelona, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain; and
| | - Sonia Fernández-Veledo
- Endocrinology and Diabetes Unit, Joan XXIII University Hospital, Institut d'Investigació Sanitària Pere i Virgili, Universitat Rovira i Virgili, Tarragona, Spain; Institut de Biomedicina de la Universitat de Barcelona de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Núria Casals
- Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Manuel Vázquez-Carrera
- Institut de Biomedicina de la Universitat de Barcelona de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain; Pharmacology Unit, Department of Pharmacology and Therapeutic Chemistry and Institut de Biomedicina de la Universitat de Barcelona, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain; and
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Joan J Vendrell
- Endocrinology and Diabetes Unit, Joan XXIII University Hospital, Institut d'Investigació Sanitària Pere i Virgili, Universitat Rovira i Virgili, Tarragona, Spain; Institut de Biomedicina de la Universitat de Barcelona de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain;
| |
Collapse
|
386
|
Haneklaus M, O'Neill LAJ. NLRP3 at the interface of metabolism and inflammation. Immunol Rev 2015; 265:53-62. [DOI: 10.1111/imr.12285] [Citation(s) in RCA: 245] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Moritz Haneklaus
- School of Biochemistry & Immunology; Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin 2 Ireland
| | - Luke A. J. O'Neill
- School of Biochemistry & Immunology; Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin 2 Ireland
| |
Collapse
|
387
|
Ballak DB, van Asseldonk EJP, van Diepen JA, Jansen H, Hijmans A, Joosten LAB, Tack CJ, Netea MG, Stienstra R. TLR-3 is present in human adipocytes, but its signalling is not required for obesity-induced inflammation in adipose tissue in vivo. PLoS One 2015; 10:e0123152. [PMID: 25867514 PMCID: PMC4395029 DOI: 10.1371/journal.pone.0123152] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 02/21/2015] [Indexed: 12/16/2022] Open
Abstract
Innate immunity plays a pivotal role in obesity-induced low-grade inflammation originating from adipose tissue. Key receptors of the innate immune system including Toll-like receptors-2 and -4 (TLRs) are triggered by nutrient excess to promote inflammation. The role of other TLRs in this process is largely unknown. In addition to double-stranded viral mRNA, TLR-3 can also recognize mRNA from dying endogenous cells, a process that is frequently observed within obese adipose tissue. Here, we identified profound expression of TLR-3 in adipocytes and investigated its role during diet-induced obesity. Human adipose tissue biopsies (n=80) and an adipocyte cell-line were used to study TLR-3 expression and function. TLR-3-/- and WT animals were exposed to a high-fat diet (HFD) for 16 weeks to induce obesity. Expression of TLR-3 was significantly higher in human adipocytes compared to the non-adipocyte cells part of the adipose tissue. In vitro, TLR-3 expression was induced during differentiation of adipocytes and stimulation of the receptor led to elevated expression of pro-inflammatory cytokines. In vivo, TLR-3 deficiency did not significantly influence HFD-induced obesity, insulin sensitivity or inflammation. In humans, TLR-3 expression in adipose tissue did not correlate with BMI or insulin sensitivity (HOMA-IR). Together, our results demonstrate that TLR-3 is highly expressed in adipocytes and functionally active. However, TLR-3 appears to play a redundant role in obesity-induced inflammation and insulin resistance.
Collapse
Affiliation(s)
- Dov B. Ballak
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Janna A. van Diepen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Henry Jansen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anneke Hijmans
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A. B. Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cees J. Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rinke Stienstra
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Nutrition, Wageningen University and Research Centre, Wageningen, The Netherlands
| |
Collapse
|
388
|
Wauquier F, Léotoing L, Philippe C, Spilmont M, Coxam V, Wittrant Y. Pros and cons of fatty acids in bone biology. Prog Lipid Res 2015; 58:121-45. [PMID: 25835096 DOI: 10.1016/j.plipres.2015.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/06/2015] [Accepted: 03/23/2015] [Indexed: 12/12/2022]
Abstract
Despite the growing interest in deciphering the causes and consequences of obesity-related disorders, the mechanisms linking fat intake to bone behaviour remain unclear. Since bone fractures are widely associated with increased morbidity and mortality, most notably in elderly and obese people, bone health has become a major social and economic issue. Consistently, public health system guidelines have encouraged low-fat diets in order to reduce associated complications. However, from a bone point of view, mechanisms linking fat intake to bone alteration remain quite controversial. Thus, after more than a decade of dedicated studies, this timely review offers a comprehensive overview of the relationships between bone and fatty acids. Using clinical evidences as a starting-point to more complex molecular elucidation, this work highlights the complexity of the system and reveals that bone alteration that cannot be solved simply by taking ω-3 pills. Fatty acid effects on bone metabolism can be both direct and indirect and require integrated investigations. Furthermore, even at the level of a single cell, one fatty acid is able to trigger several different independent pathways (receptors, metabolites…) which may all have a say in the final cellular metabolic response.
Collapse
Affiliation(s)
- Fabien Wauquier
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Laurent Léotoing
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Claire Philippe
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Mélanie Spilmont
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Véronique Coxam
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Yohann Wittrant
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France.
| |
Collapse
|
389
|
Da Silva MS, Rudkowska I. Dairy nutrients and their effect on inflammatory profile in molecular studies. Mol Nutr Food Res 2015; 59:1249-63. [DOI: 10.1002/mnfr.201400569] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 12/23/2014] [Accepted: 12/29/2014] [Indexed: 12/17/2022]
Affiliation(s)
- Marine S. Da Silva
- Department of Endocrinology and Nephrology; CHU de Québec Research Center; Quebec QC Canada
| | - Iwona Rudkowska
- Department of Endocrinology and Nephrology; CHU de Québec Research Center; Quebec QC Canada
| |
Collapse
|
390
|
Kondo K, Ishida T, Yasuda T, Nakajima H, Mori K, Tanaka N, Mori T, Monguchi T, Shinohara M, Irino Y, Toh R, Rikitake Y, Kiyomizu K, Tomiyama Y, Yamamoto J, Hirata KI. Trans-fatty acid promotes thrombus formation in mice by aggravating antithrombogenic endothelial functions via Toll-like receptors. Mol Nutr Food Res 2015; 59:729-40. [PMID: 25546502 DOI: 10.1002/mnfr.201400537] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 11/14/2014] [Accepted: 12/17/2014] [Indexed: 11/06/2022]
Abstract
SCOPE Since excessive intake of trans-fatty acid (TFA) increases the risk of myocardial infarction, we investigated the effects of TFA on thrombus formation using animal and cell culture experiments. METHODS AND RESULTS C57BL/6 mice were fed a diet containing TFA or cis-fatty acid (5% each of total calories) or a chow diet for 4 weeks, and thrombus formation was induced in the carotid artery by He-Ne laser irradiation. The high-TFA diet significantly promoted thrombus formation in the carotid artery compared to the chow or cis-fatty acid diet. TFA activated the inflammatory signaling pathway in cultured endothelial cells and in mice; aortic gene expression levels of antithrombogenic molecules, including thrombomodulin and tissue factor pathway inhibitor, were decreased, and the expression levels of prothrombogenic molecules were increased in TFA-treated mice. TFA markedly upregulated the prothrombogenic molecules and downregulated the antithrombogenic molecules in endothelial cells. In addition, TFA induced phosphorylation of c-Jun N-terminal kinase, extracellular signal-regulated kinase, and nuclear factor-κB. The TFA-activated signal pathways and prothrombogenic phenotypic changes of endothelial cells were inhibited by genetic or pharmacological inactivation of Toll-like receptors 2 and 4. CONCLUSION TFA aggravates the antithrombogenic phenotypes of vascular endothelial cells via Toll-like receptors and promotes thrombus formation in mice.
Collapse
Affiliation(s)
- Kensuke Kondo
- Division of Cardiovascular Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
391
|
Shaikh SR, Haas KM, Beck MA, Teague H. The effects of diet-induced obesity on B cell function. Clin Exp Immunol 2015; 179:90-9. [PMID: 25169121 DOI: 10.1111/cei.12444] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2014] [Indexed: 12/12/2022] Open
Abstract
B-1 and B-2 B cell subsets carry out a diverse array of functions that range broadly from responding to innate stimuli, antigen presentation, cytokine secretion and antibody production. In this review, we first cover the functional roles of the major murine B cell subsets. We then highlight emerging evidence, primarily in preclinical rodent studies, to show that select B cell subsets are a therapeutic target in obesity and its associated co-morbidities. High fat diets promote accumulation of select murine B cell phenotypes in visceral adipose tissue. As a consequence, B cells exacerbate inflammation and thereby insulin sensitivity through the production of autoantibodies and via cross-talk with select adipose resident macrophages, CD4(+) and CD8(+) T cells. In contrast, interleukin (IL)-10-secreting regulatory B cells counteract the proinflammatory profile and improve glucose sensitivity. We subsequently review data from rodent studies that show pharmacological supplementation of obesogenic diets with long chain n-3 polyunsaturated fatty acids or specialized pro-resolving lipid mediators synthesized from endogenous n-3 polyunsaturated fatty acids boost B cell activation and antibody production. This may have potential benefits for improving inflammation in addition to combating the increased risk of viral infection that is an associated complication of obesity and type II diabetes. Finally, we propose potential underlying mechanisms throughout the review by which B cell activity could be differentially regulated in response to high fat diets.
Collapse
Affiliation(s)
- S R Shaikh
- Department of Biochemistry and Molecular Biology, East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, USA; Department of Microbiology and Immunology, East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | | | | | | |
Collapse
|
392
|
Yoon JH, Kim D, Jang JH, Ghim J, Park S, Song P, Kwon Y, Kim J, Hwang D, Bae YS, Suh PG, Berggren PO, Ryu SH. Proteomic analysis of the palmitate-induced myotube secretome reveals involvement of the annexin A1-formyl peptide receptor 2 (FPR2) pathway in insulin resistance. Mol Cell Proteomics 2015; 14:882-92. [PMID: 25616869 DOI: 10.1074/mcp.m114.039651] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Indexed: 11/06/2022] Open
Abstract
Elevated levels of the free fatty acid palmitate are found in the plasma of obese patients and induce insulin resistance. Skeletal muscle secretes myokines as extracellular signaling mediators in response to pathophysiological conditions. Here, we identified and characterized the skeletal muscle secretome in response to palmitate-induced insulin resistance. Using a quantitative proteomic approach, we identified 36 secretory proteins modulated by palmitate-induced insulin resistance. Bioinformatics analysis revealed that palmitate-induced insulin resistance induced cellular stress and modulated secretory events. We found that the decrease in the level of annexin A1, a secretory protein, depended on palmitate, and that annexin A1 and its receptor, formyl peptide receptor 2 agonist, played a protective role in the palmitate-induced insulin resistance of L6 myotubes through PKC-θ modulation. In mice fed with a high-fat diet, treatment with the formyl peptide receptor 2 agonist improved systemic insulin sensitivity. Thus, we identified myokine candidates modulated by palmitate-induced insulin resistance and found that the annexin A1- formyl peptide receptor 2 pathway mediated the insulin resistance of skeletal muscle, as well as systemic insulin sensitivity.
Collapse
Affiliation(s)
| | - Dayea Kim
- From the ‡Department of Life Sciences
| | - Jin-Hyeok Jang
- §School of Interdisciplinary Bioscience and Bioengineering
| | | | | | | | | | - Jaeyoon Kim
- ‖The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Daehee Hwang
- §School of Interdisciplinary Bioscience and Bioengineering, ¶Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk 790-784, Republic of Korea, ‖‖Center for Plant Aging Research, Institute for Basic Science and Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 711-873, Republic of Korea
| | - Yoe-Sik Bae
- **Department of Biological Science, Sungkyunkwan University, Suwon 440-746, Republic of Korea, ‡‡Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 135-710, Republic of Korea
| | - Pann-Ghill Suh
- §§School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, 689-798, Republic of Korea
| | - Per-Olof Berggren
- ‖The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm SE-171 77, Sweden, Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 790-784, Republic of Korea
| | | |
Collapse
|
393
|
Santander AM, Lopez-Ocejo O, Casas O, Agostini T, Sanchez L, Lamas-Basulto E, Carrio R, Cleary MP, Gonzalez-Perez RR, Torroella-Kouri M. Paracrine Interactions between Adipocytes and Tumor Cells Recruit and Modify Macrophages to the Mammary Tumor Microenvironment: The Role of Obesity and Inflammation in Breast Adipose Tissue. Cancers (Basel) 2015; 7:143-78. [PMID: 25599228 PMCID: PMC4381255 DOI: 10.3390/cancers7010143] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/05/2015] [Indexed: 01/11/2023] Open
Abstract
The relationship between obesity and breast cancer (BC) has focused on serum factors. However, the mammary gland contains adipose tissue (AT) which may enable the crosstalk between adipocytes and tumor cells contributing to tumor macrophage recruitment. We hypothesize that the breast AT (bAT) is inflamed in obese females and plays a major role in breast cancer development. The effects of this interplay on macrophage chemotaxis were examined in vitro, using co-cultures of mouse macrophages, mammary tumor cells and adipocytes. Macrophages were exposed to the adipocyte and tumor paracrine factors leptin, CCL2 and lauric acid (alone or in combinations). In cell supernatants Luminex identified additional molecules with chemotactic and other pro-tumor functions. Focus on the adipokine leptin, which has been shown to have a central role in breast cancer pathogenesis, indicated it modulates macrophage phenotypes and functions. In vivo experiments demonstrate that mammary tumors from obese mice are larger and that bAT from obese tumor-bearers contains higher numbers of macrophages/CLS and hypertrophic adipocytes than bAT from lean tumor-bearers, thus confirming it is more inflamed. Also, bAT distal from the tumor is more inflamed in obese than in lean mice. Our results reveal that bAT plays a role in breast cancer development in obesity.
Collapse
Affiliation(s)
- Ana M Santander
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA.
| | - Omar Lopez-Ocejo
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA.
| | - Olivia Casas
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA.
| | - Thais Agostini
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA.
| | - Lidia Sanchez
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA.
| | - Eduardo Lamas-Basulto
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA.
| | - Roberto Carrio
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA.
| | - Margot P Cleary
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA.
| | - Ruben R Gonzalez-Perez
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30314, USA.
| | - Marta Torroella-Kouri
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA.
| |
Collapse
|
394
|
Abstract
Lipid metabolism is regulated by multiple signaling pathways, and generates a variety of bioactive lipid molecules. These bioactive lipid molecules known as signaling molecules, such as fatty acid, eicosanoids, diacylglycerol, phosphatidic acid, lysophophatidic acid, ceramide, sphingosine, sphingosine-1-phosphate, phosphatidylinositol-3 phosphate, and cholesterol, are involved in the activation or regulation of different signaling pathways. Lipid metabolism participates in the regulation of many cellular processes such as cell growth, proliferation, differentiation, survival, apoptosis, inflammation, motility, membrane homeostasis, chemotherapy response, and drug resistance. Bioactive lipid molecules promote apoptosis via the intrinsic pathway by modulating mitochondrial membrane permeability and activating different enzymes including caspases. In this review, we discuss recent data in the fields of lipid metabolism, lipid-mediated apoptosis, and cancer therapy. In conclusion, understanding the underlying molecular mechanism of lipid metabolism and the function of different lipid molecules could provide the basis for cancer cell death rationale, discover novel and potential targets, and develop new anticancer drugs for cancer therapy.
Collapse
|
395
|
Quercetin protects against obesity-induced skeletal muscle inflammation and atrophy. Mediators Inflamm 2014; 2014:834294. [PMID: 25614714 PMCID: PMC4295595 DOI: 10.1155/2014/834294] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/22/2014] [Accepted: 11/23/2014] [Indexed: 01/06/2023] Open
Abstract
Skeletal muscle inflammation and atrophy are closely associated with metabolic impairment such as insulin resistance. Quercetin, a natural polyphenol flavonoid, is known to elicit anti-inflammatory and antioxidant activities. In this study, we investigated its effect on obesity-induced skeletal muscle inflammation and atrophy in mice. Male C57BL/6 mice were fed a regular diet, a high-fat diet (HFD), and an HFD supplemented with quercetin for nine weeks. Quercetin reduced levels of inflammatory cytokines and macrophage accumulation in the skeletal muscle of the HFD-fed obese mice. It also reduced transcript and protein levels of the specific atrophic factors, Atrogin-1 and MuRF1, in the skeletal muscle of the HFD-fed obese mice, and protected against the reduction of muscle mass and muscle fiber size. In vitro, quercetin markedly diminished transcript levels of inflammatory receptors and activation of their signaling molecules (ERK, p38 MAPK, and NF-κB) in cocultured myotubes/macrophages, and this was accompanied by reduced expression of the atrophic factors. Together, these findings suggest that quercetin reduces obesity-induced skeletal muscle atrophy by inhibiting inflammatory receptors and their signaling pathway. Quercetin may be useful for preventing obesity-induced muscle inflammation and sarcopenia.
Collapse
|
396
|
Honda KL, Lamon-Fava S, Matthan NR, Wu D, Lichtenstein AH. EPA and DHA exposure alters the inflammatory response but not the surface expression of Toll-like receptor 4 in macrophages. Lipids 2014; 50:121-9. [PMID: 25408476 DOI: 10.1007/s11745-014-3971-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/03/2014] [Indexed: 12/31/2022]
Abstract
Dietary intake of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) and their respective enrichment in cell membranes have been negatively associated with atherosclerotic lesion development. This effect may be mediated, in part, by dampened inflammatory response of macrophages triggered by toll-like receptor 4 (TLR4) activation. This study investigated the influence of membrane fatty acid profile on TLR4-mediated inflammation in RAW 264.7 macrophages. Cells pretreated with myristic acid (MA), EPA, DHA or vehicle control for 24 h were stimulated with ultra-pure LPS, a specific TLR4 agonist, for 6 or 24 h, corresponding to early and late stages of TNFα and IL-6 protein induction. Treatment significantly increased cell membrane MA, EPA, and DHA by 4.5-, 20.6-, and 8.9-fold, respectively. MA significantly increased IL-6 secretion 6 h post-exposure to the fatty acid, but did not change TNFα secretion in response to any other treatment condition. EPA and DHA significantly reduced TNFα secretion by 36 and 41 %, respectively, in cells stimulated for 24 h but not 6 h. In contrast, EPA and DHA significantly reduced IL-6 secretion at both 6 h (67 and 72%, respectively) and 24 h (69 and 72%, respectively). MA or DHA treatment had no significant effect compared to vehicle on factors influencing cellular LPS recognition, including LPS-cell association, and cell surface expression of TLR4, TLR4-MD2 complex, and CD14. These data suggest that membrane fatty acid profiles influence the TLR4-mediated inflammatory response in macrophages, via mechanisms that occur downstream of TLR4 receptor activation.
Collapse
Affiliation(s)
- Kaori L Honda
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA, 02111, USA,
| | | | | | | | | |
Collapse
|
397
|
Ageing, adipose tissue, fatty acids and inflammation. Biogerontology 2014; 16:235-48. [PMID: 25367746 DOI: 10.1007/s10522-014-9536-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 10/20/2014] [Indexed: 12/22/2022]
Abstract
A common feature of ageing is the alteration in tissue distribution and composition, with a shift in fat away from lower body and subcutaneous depots to visceral and ectopic sites. Redistribution of adipose tissue towards an ectopic site can have dramatic effects on metabolic function. In skeletal muscle, increased ectopic adiposity is linked to insulin resistance through lipid mediators such as ceramide or DAG, inhibiting the insulin receptor signalling pathway. Additionally, the risk of developing cardiovascular disease is increased with elevated visceral adipose distribution. In ageing, adipose tissue becomes dysfunctional, with the pathway of differentiation of preadipocytes to mature adipocytes becoming impaired; this results in dysfunctional adipocytes less able to store fat and subsequent fat redistribution to ectopic sites. Low grade systemic inflammation is commonly observed in ageing, and may drive the adipose tissue dysfunction, as proinflammatory cytokines are capable of inhibiting adipocyte differentiation. Beyond increased ectopic adiposity, the effect of impaired adipose tissue function is an elevation in systemic free fatty acids (FFA), a common feature of many metabolic disorders. Saturated fatty acids can be regarded as the most detrimental of FFA, being capable of inducing insulin resistance and inflammation through lipid mediators such as ceramide, which can increase risk of developing atherosclerosis. Elevated FFA, in particular saturated fatty acids, maybe a driving factor for both the increased insulin resistance, cardiovascular disease risk and inflammation in older adults.
Collapse
|
398
|
Saturated fatty acids up-regulate COX-2 expression in prostate epithelial cells via toll-like receptor 4/NF-κB signaling. Inflammation 2014; 37:467-77. [PMID: 24221358 DOI: 10.1007/s10753-013-9760-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cyclooxygenase-2 (COX-2) has been implicated in prostate carcinogenesis, and recently it has been confirmed to be a molecular target of saturated fatty acids (SFAs). In the present study, we investigated the effect of stearic acid (SA) and palmitic acid (PA), two of the most abundant SFAs contained in dietary fat, on COX-2 expression in prostate epithelial cells and the signaling transduction pathway involved. First, we demonstrated that both SA and PA increased the mRNA and protein expression of COX-2, and consistently induced the activation of NF-κB in RWPE-1, BPH-1 and PC-3 prostate epithelial cell lines. The effect of SA and PA on COX-2 over-expression and NF-κB activation was in a dose-dependent manner, and PA was more potent than SA at the same concentration. Then, we demonstrated inhibition of NF-κB using its specific inhibitor strikingly attenuated PA-induced COX-2 expression. Toll-like receptor 4 (TLR4) was revealed to be expressed on RWPE-1, BPH-1 and PC-3 cell lines by PCR and immunofluorescence staining, and blocking its signaling significantly inhibited PA induced COX-2 over-expression and NF-κB activation. Taken together, we demonstrated that SFAs can up-regulate COX-2 expression in prostate epithelial cells, and this effect was mediated mainly through the TLR4/NF-κB signaling pathway.
Collapse
|
399
|
Niu N, Li B, Hu Y, Li X, Li J, Zhang H. Protective effects of scoparone against lipopolysaccharide-induced acute lung injury. Int Immunopharmacol 2014; 23:127-33. [DOI: 10.1016/j.intimp.2014.08.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/28/2014] [Accepted: 08/13/2014] [Indexed: 11/27/2022]
|
400
|
Bergamo P, Luongo D, Miyamoto J, Cocca E, Kishino S, Ogawa J, Tanabe S, Rossi M. Immunomodulatory activity of a gut microbial metabolite of dietary linoleic acid, 10-hydroxy-cis-12-octadecenoic acid, associated with improved antioxidant/detoxifying defences. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.10.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|