401
|
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
|
402
|
Płóciennikowska A, Hromada-Judycka A, Borzęcka K, Kwiatkowska K. Co-operation of TLR4 and raft proteins in LPS-induced pro-inflammatory signaling. Cell Mol Life Sci 2014; 72:557-581. [PMID: 25332099 PMCID: PMC4293489 DOI: 10.1007/s00018-014-1762-5] [Citation(s) in RCA: 565] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/01/2014] [Accepted: 10/13/2014] [Indexed: 11/28/2022]
Abstract
Toll-like receptor 4 (TLR4) is activated by lipopolysaccharide (LPS), a component of Gram-negative bacteria to induce production of pro-inflammatory mediators aiming at eradication of the bacteria. Dysregulation of the host responses to LPS can lead to a systemic inflammatory condition named sepsis. In a typical scenario, activation of TLR4 is preceded by binding of LPS to CD14 protein anchored in cholesterol- and sphingolipid-rich microdomains of the plasma membrane called rafts. CD14 then transfers the LPS to the TLR4/MD-2 complex which dimerizes and triggers MyD88- and TRIF-dependent production of pro-inflammatory cytokines and type I interferons. The TRIF-dependent signaling is linked with endocytosis of the activated TLR4, which is controlled by CD14. In addition to CD14, other raft proteins like Lyn tyrosine kinase of the Src family, acid sphingomyelinase, CD44, Hsp70, and CD36 participate in the TLR4 signaling triggered by LPS and non-microbial endogenous ligands. In this review, we summarize the current state of the knowledge on the involvement of rafts in TLR4 signaling, with an emphasis on how the raft proteins regulate the TLR4 signaling pathways. CD14-bearing rafts, and possibly CD36-rich rafts, are believed to be preferred sites of the assembly of a multimolecular complex which mediates the endocytosis of activated TLR4.
Collapse
Affiliation(s)
- Agnieszka Płóciennikowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093, Warsaw, Poland
| | - Aneta Hromada-Judycka
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093, Warsaw, Poland
| | - Kinga Borzęcka
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093, Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093, Warsaw, Poland.
| |
Collapse
|
403
|
Jovanović I, Ugrenović S, Ljubomirović M, Vasović L, Čukuranović R, Stefanović V. Folliculo-stellate cells – Potential mediators of the inflammaging-induced hyperactivity of the hypothalamic–pituitary–adrenal axis in healthy elderly individuals. Med Hypotheses 2014; 83:501-5. [DOI: 10.1016/j.mehy.2014.08.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/10/2014] [Accepted: 08/15/2014] [Indexed: 10/24/2022]
|
404
|
Palmitoleic acid (n-7) attenuates the immunometabolic disturbances caused by a high-fat diet independently of PPARα. Mediators Inflamm 2014; 2014:582197. [PMID: 25147439 PMCID: PMC4131426 DOI: 10.1155/2014/582197] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/17/2014] [Accepted: 06/17/2014] [Indexed: 12/30/2022] Open
Abstract
Palmitoleic acid (PMA) has anti-inflammatory and antidiabetic activities. Here we tested whether these effects of PMA on glucose homeostasis and liver inflammation, in mice fed with high-fat diet (HFD), are PPAR-α dependent. C57BL6 wild-type (WT) and PPAR-α-knockout (KO) mice fed with a standard diet (SD) or HFD for 12 weeks were treated after the 10th week with oleic acid (OLA, 300 mg/kg of b.w.) or PMA 300 mg/kg of b.w. Steatosis induced by HFD was associated with liver inflammation only in the KO mice, as shown by the increased hepatic levels of IL1-beta, IL-12, and TNF-α; however, the HFD increased the expression of TLR4 and decreased the expression of IL1-Ra in both genotypes. Treatment with palmitoleate markedly attenuated the insulin resistance induced by the HFD, increased glucose uptake and incorporation into muscle in vitro, reduced the serum levels of AST in WT mice, decreased the hepatic levels of IL1-beta and IL-12 in KO mice, reduced the expression of TLR-4 and increased the expression of IL-1Ra in WT mice, and reduced the phosphorylation of NF 𝜅B (p65) in the livers of KO mice. We conclude that palmitoleate attenuates diet-induced insulin resistance, liver inflammation, and damage through mechanisms that do not depend on PPAR-α.
Collapse
|
405
|
Myles IA. Fast food fever: reviewing the impacts of the Western diet on immunity. Nutr J 2014; 13:61. [PMID: 24939238 PMCID: PMC4074336 DOI: 10.1186/1475-2891-13-61] [Citation(s) in RCA: 199] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/23/2014] [Indexed: 02/08/2023] Open
Abstract
While numerous changes in human lifestyle constitute modern life, our diet has been gaining attention as a potential contributor to the increase in immune-mediated diseases. The Western diet is characterized by an over consumption and reduced variety of refined sugars, salt, and saturated fat. Herein our objective is to detail the mechanisms for the Western diet's impact on immune function. The manuscript reviews the impacts and mechanisms of harm for our over-indulgence in sugar, salt, and fat, as well as the data outlining the impacts of artificial sweeteners, gluten, and genetically modified foods; attention is given to revealing where the literature on the immune impacts of macronutrients is limited to either animal or in vitro models versus where human trials exist. Detailed attention is given to the dietary impact on the gut microbiome and the mechanisms by which our poor dietary choices are encoded into our gut, our genes, and are passed to our offspring. While today's modern diet may provide beneficial protection from micro- and macronutrient deficiencies, our over abundance of calories and the macronutrients that compose our diet may all lead to increased inflammation, reduced control of infection, increased rates of cancer, and increased risk for allergic and auto-inflammatory disease.
Collapse
Affiliation(s)
- Ian A Myles
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike Building 33, Room 2W10A, Bethesda, MD, 20892, Maryland.
| |
Collapse
|
406
|
Rutkowsky JM, Knotts TA, Ono-Moore KD, McCoin CS, Huang S, Schneider D, Singh S, Adams SH, Hwang DH. Acylcarnitines activate proinflammatory signaling pathways. Am J Physiol Endocrinol Metab 2014; 306:E1378-87. [PMID: 24760988 PMCID: PMC4059985 DOI: 10.1152/ajpendo.00656.2013] [Citation(s) in RCA: 236] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Incomplete β-oxidation of fatty acids in mitochondria is a feature of insulin resistance and type 2 diabetes mellitus (T2DM). Previous studies revealed that plasma concentrations of medium- and long-chain acylcarnitines (by-products of incomplete β-oxidation) are elevated in T2DM and insulin resistance. In a previous study, we reported that mixed D,L isomers of C12- or C14-carnitine induced an NF-κB-luciferase reporter gene in RAW 264.7 cells, suggesting potential activation of proinflammatory pathways. Here, we determined whether the physiologically relevant L-acylcarnitines activate classical proinflammatory signaling pathways and if these outcomes involve pattern recognition receptor (PRR)-associated pathways. Acylcarnitines induced the expression of cyclooxygenase-2 in a chain length-dependent manner in RAW 264.7 cells. L-C14 carnitine (5-25 μM), used as a representative acylcarnitine, stimulated the expression and secretion of proinflammatory cytokines in a dose-dependent manner. Furthermore, L-C14 carnitine induced phosphorylation of JNK and ERK, common downstream components of many proinflammatory signaling pathways including PRRs. Knockdown of MyD88, a key cofactor in PRR signaling and inflammation, blunted the proinflammatory effects of acylcarnitine. While these results point to potential involvement of PRRs, L-C14 carnitine promoted IL-8 secretion from human epithelial cells (HCT-116) lacking Toll-like receptors (TLR)2 and -4, and did not activate reporter constructs in TLR overexpression cell models. Thus, acylcarnitines have the potential to activate inflammation, but the specific molecular and tissue target(s) involved remain to be identified.
Collapse
Affiliation(s)
- Jennifer M Rutkowsky
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, California
| | - Trina A Knotts
- Obesity and Metabolism Research Unit, US Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California; Department of Nutrition, University of California, Davis, California
| | - Kikumi D Ono-Moore
- Department of Nutrition, University of California, Davis, California; Graduate Group in Nutritional Biology, University of California, Davis, California
| | - Colin S McCoin
- Graduate Group in Molecular, Cellular and Integrative Physiology, University of California, Davis, California; and
| | - Shurong Huang
- Immunity and Disease Prevention Research Unit, US Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Dina Schneider
- Immunity and Disease Prevention Research Unit, US Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Shamsher Singh
- Immunity and Disease Prevention Research Unit, US Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Sean H Adams
- Obesity and Metabolism Research Unit, US Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California; Department of Nutrition, University of California, Davis, California; Graduate Group in Nutritional Biology, University of California, Davis, California; Graduate Group in Molecular, Cellular and Integrative Physiology, University of California, Davis, California; and
| | - Daniel H Hwang
- Department of Nutrition, University of California, Davis, California; Graduate Group in Nutritional Biology, University of California, Davis, California; Immunity and Disease Prevention Research Unit, US Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| |
Collapse
|
407
|
Song R, Ao L, Zhao KS, Zheng D, Venardos N, Fullerton DA, Meng X. Soluble biglycan induces the production of ICAM-1 and MCP-1 in human aortic valve interstitial cells through TLR2/4 and the ERK1/2 pathway. Inflamm Res 2014; 63:703-10. [PMID: 24875140 DOI: 10.1007/s00011-014-0743-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/05/2014] [Accepted: 05/12/2014] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE Mononuclear cell infiltration in valvular tissue is one of the characteristics in calcific aortic valve disease. The inflammatory responses of aortic valve interstitial cells (AVICs) play an important role in valvular inflammation. However, it remains unclear what may evoke AVIC inflammatory responses. Accumulation of biglycan has been found in diseased aortic valve leaflets. Soluble biglycan can function as a danger-associated molecular pattern to induce the production of proinflammatory mediators in cultured macrophages. We tested the hypothesis that soluble biglycan induces AVIC production of proinflammatory mediators involved in mononuclear cell infiltration through Toll-like receptor (TLR)-dependent signaling pathways. METHODS Human AVICs isolated from normal aortic valve leaflets were treated with specific siRNA and neutralizing antibody against TLR2 or TLR4 before biglycan stimulation. The production of ICAM-1 and MCP-1 was assessed. To determine the signaling pathway involved, phosphorylation of ERK1/2 and p38 MAPK was analyzed, and specific inhibitors of ERK1/2 and p38 MAPK were applied. RESULTS Soluble biglycan induced ICAM-1 expression and MCP-1 release in human AVICs, but had no effect on IL-6 release. TLR4 blockade and knockdown reduced ICAM-1 and MCP-1 production induced by biglycan, while knockdown and neutralization of TLR2 resulted in greater suppression of the inflammatory responses. Biglycan induced the phosphorylation of ERK1/2 and p38 MAPK, but ICAM-1 and MCP-1 production was reduced only by inhibition of the ERK1/2 pathway. Further, inhibition of ERK1/2 attenuated NF-κB activation following biglycan treatment. CONCLUSIONS Soluble biglycan induces the expression of ICAM-1 and MCP-1 in human AVICs through TLR2 and TLR4 and requires activation of the ERK1/2 pathway. AVIC inflammatory responses induced by soluble biglycan may contribute to the mechanism of chronic inflammation associated with calcific aortic valve disease.
Collapse
Affiliation(s)
- Rui Song
- Department of Surgery, University of Colorado Denver, 12700 E 19th Avenue, Box C-320, Aurora, CO, 80045, USA
| | | | | | | | | | | | | |
Collapse
|
408
|
Bublin M, Eiwegger T, Breiteneder H. Do lipids influence the allergic sensitization process? J Allergy Clin Immunol 2014; 134:521-9. [PMID: 24880633 PMCID: PMC4151997 DOI: 10.1016/j.jaci.2014.04.015] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/09/2014] [Accepted: 04/17/2014] [Indexed: 11/24/2022]
Abstract
Allergic sensitization is a multifactorial process that is not only influenced by the allergen and its biological function per se but also by other small molecular compounds, such as lipids, that are directly bound as ligands by the allergen or are present in the allergen source. Several members of major allergen families bind lipid ligands through hydrophobic cavities or electrostatic or hydrophobic interactions. These allergens include certain seed storage proteins, Bet v 1–like and nonspecific lipid transfer proteins from pollens and fruits, certain inhalant allergens from house dust mites and cockroaches, and lipocalins. Lipids from the pollen coat and furry animals and the so-called pollen-associated lipid mediators are codelivered with the allergens and can modulate the immune responses of predisposed subjects by interacting with the innate immune system and invariant natural killer T cells. In addition, lipids originating from bacterial members of the pollen microbiome contribute to the outcome of the sensitization process. Dietary lipids act as adjuvants and might skew the immune response toward a TH2-dominated phenotype. In addition, the association with lipids protects food allergens from gastrointestinal degradation and facilitates their uptake by intestinal cells. These findings will have a major influence on how allergic sensitization will be viewed and studied in the future.
Collapse
Affiliation(s)
- Merima Bublin
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Thomas Eiwegger
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Heimo Breiteneder
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
409
|
Zhou YJ, Tang YS, Song YL, Li A, Zhou H, Li Y. Saturated fatty acid induces insulin resistance partially through nucleotide-binding oligomerization domain 1 signaling pathway in adipocytes. ACTA ACUST UNITED AC 2014; 28:211-7. [PMID: 24382222 DOI: 10.1016/s1001-9294(14)60004-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To investigate the potential role of nucleotide-binding oligomerization domain 1 (NOD1), a component of the innate immune system, in mediating lipid-induced insulin resistance in adipocytes. METHODS Adipocytes from Toll-like receptor 4 deficiency mice were used for stimulation experiments. The effect of oleate/palmitate mixture on nuclear factor-κB (NF-κB) activation was analyzed by reporter plasmid assay. The release of proinflammatory chemokine/cytokines production was determined by using real-time PCR. Insulin-stimulated glucose uptake was measured by 2-deoxy-D-[3H] glucose uptake assay. Chemokine/cytokine expression and glucose uptake in adipocytes transfected with small interfering RNA (siRNA) targeting NOD1 upon fatty acids treatment were analyzed. RESULTS Oleate/palmitate mixture activated the NF-κB pathway and induced interleukin-6, tumor necrosis factor-α, and monocyte chemoattractant protein-1 mRNA expressions in adipocytes from mice deficient in Toll-like receptor 4, and these effects were blocked by siRNA targeting NOD1. Furthermore, saturated fatty acids decreased the ability of insulin-stimulated glucose uptake. Importantly, siRNA targeting NOD1 partially reversed saturated fatty acid-induced suppression of insulin-induced glucose uptake. CONCLUSION NOD1 might play an important role in saturated fatty acid-induced insulin resistance in adipocytes, suggesting a mechanism by which reduced NOD1 activity confers beneficial effects on insulin action.
Collapse
Affiliation(s)
| | - Yin-si Tang
- Department of Endocrinology and Metabolism, Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yu-ling Song
- Department of Endocrinology and Metabolism, Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Ai Li
- Department of Endocrinology and Metabolism, Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Hui Zhou
- Department of Endocrinology and Metabolism, Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yan Li
- Department of Endocrinology and Metabolism, Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| |
Collapse
|
410
|
Critical role of TLR2 and MyD88 for functional response of macrophages to a group IIA-secreted phospholipase A2 from snake venom. PLoS One 2014; 9:e93741. [PMID: 24718259 PMCID: PMC3981733 DOI: 10.1371/journal.pone.0093741] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 03/06/2014] [Indexed: 01/10/2023] Open
Abstract
The snake venom MT-III is a group IIA secreted phospholipase A2 (sPLA2) enzyme with functional and structural similarities with mammalian pro-inflammatory sPLA2s of the same group. Previously, we demonstrated that MT-III directly activates the innate inflammatory response of macrophages, including release of inflammatory mediators and formation of lipid droplets (LDs). However, the mechanisms coordinating these processes remain unclear. In the present study, by using TLR2−/− or MyD88−/− or C57BL/6 (WT) male mice, we report that TLR2 and MyD88 signaling have a critical role in MT-III-induced inflammatory response in macrophages. MT-III caused a marked release of PGE2, PGD2, PGJ2, IL-1β and IL-10 and increased the number of LDs in WT macrophages. In MT-III-stimulated TLR2−/− macrophages, formation of LDs and release of eicosanoids and cytokines were abrogated. In MyD88−/− macrophages, MT-III-induced release of PGE2, IL-1β and IL-10 was abrogated, but release of PGD2 and PGJ2 was maintained. In addition, COX-2 protein expression seen in MT-III-stimulated WT macrophages was abolished in both TLR2−/− and MyD88−/− cells, while perilipin 2 expression was abolished only in MyD88−/− cells. We further demonstrated a reduction of saturated, monounsaturated and polyunsaturated fatty acids and a release of the TLR2 agonists palmitic and oleic acid from MT-III-stimulated WT macrophages compared with WT control cells, thus suggesting these fatty acids as major messengers for MT-III-induced engagement of TLR2/MyD88 signaling. Collectively, our findings identify for the first time a TLR2 and MyD88-dependent mechanism that underlies group IIA sPLA2-induced inflammatory response in macrophages.
Collapse
|
411
|
Eguchi K, Manabe I. Toll-like receptor, lipotoxicity and chronic inflammation: the pathological link between obesity and cardiometabolic disease. J Atheroscler Thromb 2014; 21:629-39. [PMID: 24695021 DOI: 10.5551/jat.22533] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The epidemic growth in the prevalence of obesity has made the impact of metabolic syndrome on cardiovascular events increasingly significant. Elevated visceral adiposity, the indispensable component of metabolic syndrome, is thought to play a primary role in the increasing incidence of cardiometabolic disorders. Importantly, obesity is not merely the simple expansion of adipose tissue mass; it also involves the activation of inflammatory processes within visceral adipose tissue. Adipose tissue inflammation on the one hand enhances the production of proinflammatory adipokines and on the other hand increases the release of free fatty acids via the activation of lipolysis. The adipokines and free fatty acids secreted from visceral fat then contribute to a cardiometabolic pathology. We herein summarize recent advances in our understanding of the mechanisms by which visceral obesity leads to the activation of inflammation in cardiovascular and metabolic tissues and promotes cardiometabolic disease. Our focus is on Toll-like receptor 4 signaling and free fatty acids as mediators of chronic inflammation in patients with metabolic syndrome and atherosclerosis.
Collapse
Affiliation(s)
- Kosei Eguchi
- Department of Cardiovascular Medicine, The University of Tokyo
| | | |
Collapse
|
412
|
Cullberg KB, Larsen JØ, Pedersen SB, Richelsen B. Effects of LPS and dietary free fatty acids on MCP-1 in 3T3-L1 adipocytes and macrophages in vitro. Nutr Diabetes 2014; 4:e113. [PMID: 24662749 PMCID: PMC3974034 DOI: 10.1038/nutd.2014.10] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 01/23/2014] [Accepted: 02/07/2014] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND High levels of free fatty acids (FFA) have been suggested to be one of the underlying mechanisms for adipose tissue (AT) inflammation and dysfunction in obesity. Human AT produces several adipokines including monocyte chemoattractant protein-1 (MCP-1), which are involved in the pathogenesis of obesity-mediated inflammation. OBJECTIVE In this study, we investigated the effects of lipopolysaccharide (LPS) and a panel of dietary FFA on MCP-1 gene and protein expression in adipocytes and macrophages. Furthermore, we investigated whether the effect of LPS and FFA were mediated through the toll-like receptor 4 (TLR4). METHODS 3T3-L1 adipocytes and THP-1 macrophages were incubated for 24 h with the following FFA: monounsaturated fatty acid (oleic acid), saturated fatty acid (palmitic acid) and trans fatty acid (elaidic acid; 500 μM) with and without LPS (2 ng ml(-1)), and MCP-1 and TLR4 mRNA expression and MCP-1 protein secretion was determined. RESULTS The results showed that LPS significantly increased MCP-1 and TLR4 expression and MCP-1 secretion in 3T3-L1 adipocytes, and that the MCP-1 expression was blocked by a TLR4 inhibitor (CLI095). The effects of the various FFA on MCP-1 mRNA expression and protein secretion in the adipocytes showed no significant changes either alone or in combination with LPS. In macrophages, palmitic acid increased MCP-1 mRNA expression by 1.8-fold (P<0.05), but oleic acid and elaidic acid had no effects. CONCLUSIONS In conclusion, in 3T3-L1 adipocyte, the TLR4-agonist, LPS, stimulates the proinflammatory chemokine MCP-1. The different classes of FFA did not induce MCP-1 mRNA expression or protein secretion in the adipocytes, but the saturated FFA, palmitic acid, induced MCP-1 mRNA expression in macrophages, possibly because of the higher expression level of TLR4 in the macrophages than the adipocytes. Our results indicate that FFA may induce AT inflammation through proinflammatory stimulation of macrophages.
Collapse
Affiliation(s)
- K B Cullberg
- Department of Endocrinology and Internal Medicine, MEA, THG, Aarhus University Hospital, Aarhus C, Denmark
| | - J Ø Larsen
- Department of Endocrinology and Internal Medicine, MEA, THG, Aarhus University Hospital, Aarhus C, Denmark
| | - S B Pedersen
- Department of Endocrinology and Internal Medicine, MEA, THG, Aarhus University Hospital, Aarhus C, Denmark
| | - B Richelsen
- Department of Endocrinology and Internal Medicine, MEA, THG, Aarhus University Hospital, Aarhus C, Denmark
| |
Collapse
|
413
|
Xu G, Zhang Z, Xing Y, Wei J, Ge Z, Liu X, Zhang Y, Huang X. MicroRNA-149 Negatively Regulates TLR-Triggered Inflammatory Response in Macrophages by Targeting MyD88. J Cell Biochem 2014; 115:919-27. [PMID: 24375488 DOI: 10.1002/jcb.24734] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 12/04/2013] [Indexed: 01/07/2023]
Affiliation(s)
- Guangxian Xu
- General Hospital of Ningxia Medical University; Yinchuan 750004 China
- School of Laboratory Medicine; Ningxia Medical University; Yinchuan 750004 China
- Huashan Hospital, Fudan University; Shanghai 200400 China
| | - Zhaobo Zhang
- Huashan Hospital, Fudan University; Shanghai 200400 China
- Department of Laboratory Medicine; Cangzhou Hospital; Cangzhou 061000 Hebei China
| | - Yiwen Xing
- School of Laboratory Medicine; Ningxia Medical University; Yinchuan 750004 China
| | - Jun Wei
- General Hospital of Ningxia Medical University; Yinchuan 750004 China
- School of Laboratory Medicine; Ningxia Medical University; Yinchuan 750004 China
| | - Zhaohui Ge
- General Hospital of Ningxia Medical University; Yinchuan 750004 China
| | - Xiaoming Liu
- College of Life Science; Ningxia University; Yinchuan 750021 China
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology; Bloomberg School of Public Health; Johns Hopkins University; Baltimore Maryland 21205
| | - Xuelan Huang
- General Hospital of Ningxia Medical University; Yinchuan 750004 China
| |
Collapse
|
414
|
LPS response pattern of inflammatory adipokines in an in vitro 3T3-L1 murine adipocyte model. Inflamm Res 2014; 63:495-507. [DOI: 10.1007/s00011-014-0721-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 01/30/2014] [Accepted: 02/05/2014] [Indexed: 12/21/2022] Open
|
415
|
Toll-like receptors and NLRP3 as central regulators of pancreatic islet inflammation in type 2 diabetes. Immunol Cell Biol 2014; 92:314-23. [PMID: 24492799 DOI: 10.1038/icb.2014.4] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/03/2014] [Accepted: 01/06/2014] [Indexed: 12/17/2022]
Abstract
The global health and economic burden of type 2 diabetes (T2D) has reached staggering proportions. Current projections estimate that 592 million people will have diabetes by 2035. T2D-which comprises 90% of cases-is a complex disease, in most cases resulting from a combination of predisposing genes and an unhealthy environment. Clinical onset of the disease occurs when pancreatic β cells fail in the face of insulin resistance. It has long been appreciated that chronic activation of the innate immune system is associated with T2D, and many organs critical to the regulation of glucose homeostasis show signs of a chronic inflammatory process, including the pancreatic islets of Langerhans. Recent clinical trials using IL-1-targeting agents have confirmed that inflammation contributes to β-cell failure in humans with T2D. However, little is known about the nature of the pro-inflammatory response within the islet, and there is considerable debate about the triggers for islet inflammation, which may be systemically derived and/or tissue-specific. In this review, we present evidence that Toll-like receptors 2 and 4 and the NLRP3 (Nucleotide-binding oligomerization domain, Leucine-rich Repeat and Pyrin domain containing 3) inflammasome are triggers for islet inflammation in T2D and propose that the activation of macrophages by these triggers mediates islet endocrine cell dysfunction. Therapeutically targeting these receptors may improve hyperglycemia and protect the β cell in T2D.
Collapse
|
416
|
Myles IA, Pincus NB, Fontecilla NM, Datta SK. Effects of parental omega-3 fatty acid intake on offspring microbiome and immunity. PLoS One 2014; 9:e87181. [PMID: 24489864 PMCID: PMC3906117 DOI: 10.1371/journal.pone.0087181] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 12/20/2013] [Indexed: 02/07/2023] Open
Abstract
The "Western diet" is characterized by increased intake of saturated and omega-6 (n-6) fatty acids with a relative reduction in omega-3 (n-3) consumption. These fatty acids can directly and indirectly modulate the gut microbiome, resulting in altered host immunity. Omega-3 fatty acids can also directly modulate immunity through alterations in the phospholipid membranes of immune cells, inhibition of n-6 induced inflammation, down-regulation of inflammatory transcription factors, and by serving as pre-cursors to anti-inflammatory lipid mediators such as resolvins and protectins. We have previously shown that consumption by breeder mice of diets high in saturated and n-6 fatty acids have inflammatory and immune-modulating effects on offspring that are at least partially driven by vertical transmission of altered gut microbiota. To determine if parental diets high in n-3 fatty acids could also affect offspring microbiome and immunity, we fed breeding mice an n-3-rich diet with 40% calories from fat and measured immune outcomes in their offspring. We found offspring from mice fed diets high in n-3 had altered gut microbiomes and modestly enhanced anti-inflammatory IL-10 from both colonic and splenic tissue. Omega-3 pups were protected during peanut oral allergy challenge with small but measurable alterations in peanut-related serologies. However, n-3 pups displayed a tendency toward worsened responses during E. coli sepsis and had significantly worse outcomes during Staphylococcus aureus skin infection. Our results indicate excess parental n-3 fatty acid intake alters microbiome and immune response in offspring.
Collapse
Affiliation(s)
- Ian A. Myles
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Nathan B. Pincus
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Natalia M. Fontecilla
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sandip K. Datta
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
417
|
Matsuo S, Yang WL, Aziz M, Kameoka S, Wang P. Fatty acid synthase inhibitor C75 ameliorates experimental colitis. Mol Med 2014; 20:1-9. [PMID: 24306512 DOI: 10.2119/molmed.2013.00113] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/25/2013] [Indexed: 12/22/2022] Open
Abstract
Abnormalities of lipid metabolism through overexpression of fatty acid synthase (FASN), which catalyzes the formation of long-chain fatty acids, are associated with the development of inflammatory bowel disease (IBD). C75 is a synthetic α-methylene-γ-butyrolactone compound that inhibits FASN activity. We hypothesized that C75 treatment could effectively reduce the severity of experimental colitis. Male C57BL/6 mice were fed 4% dextran sodium sulfate (DSS) for 7 d. C75 (5 mg/kg body weight) or dimethyl sulfoxide (DMSO) (vehicle) was administered intraperitoneally from d 2 to 6. Clinical parameters were monitored daily. Mice were euthanized on d 8 for histological evaluation and measurements of colon length, chemokine, cytokine and inflammatory mediator expression. C75 significantly reduced body weight loss from 23% to 15% on d 8, compared with the vehicle group. The fecal bleeding, diarrhea and colon histological damage scores in the C75-treated group were significantly lower than scores in the vehicle animals. Colon shortening was significantly improved after C75 treatment. C75 protected colon tissues from DSS-induced apoptosis by inhibiting caspase-3 activity. Macrophage inflammatory protein 2, keratinocyte-derived chemokine, myeloperoxidase activity and proinflammatory cytokines (tumor necrosis factor-α, interleukin [IL]-1β and IL-6) in the colon were significantly downregulated in the C75-treated group, compared with the vehicle group. Treatment with C75 in colitis mice inhibited the elevation of FASN, cyclooxygenase-2 and inducible nitric oxide synthase expression as well as IκB degradation in colon tissues. C75 administration alleviates the severity of colon damage and inhibits the activation of inflammatory pathways in DSS-induced colitis. Thus, inhibition of FASN may represent an attractive therapeutic potential for treating IBD.
Collapse
Affiliation(s)
- Shingo Matsuo
- Department of Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, and The Feinstein Institute for Medical Research, Manhasset, New York, United States of America Department of Surgery II, Tokyo Women's Medical University, Tokyo, Japan
| | - Weng-Lang Yang
- Department of Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, and The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Monowar Aziz
- Department of Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, and The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Shingo Kameoka
- Department of Surgery II, Tokyo Women's Medical University, Tokyo, Japan
| | - Ping Wang
- Department of Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, and The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| |
Collapse
|
418
|
Drosatos-Tampakaki Z, Drosatos K, Siegelin Y, Gong S, Khan S, Van Dyke T, Goldberg IJ, Schulze PC, Schulze-Späte U. Palmitic acid and DGAT1 deficiency enhance osteoclastogenesis, while oleic acid-induced triglyceride formation prevents it. J Bone Miner Res 2014; 29:1183-95. [PMID: 24272998 PMCID: PMC4945760 DOI: 10.1002/jbmr.2150] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 11/01/2013] [Accepted: 11/16/2013] [Indexed: 01/03/2023]
Abstract
Both obesity and diabetes mellitus are associated with alterations in lipid metabolism as well as a change in bone homeostasis and osteoclastogenesis. We hypothesized that increased fatty acid levels affect bone health by altering precursor cell differentiation and osteoclast activation. Here we show that palmitic acid (PA, 16:0) enhances receptor activator of NF-κB ligand (RANKL)-stimulated osteoclastogenesis and is sufficient to induce osteoclast differentiation even in the absence of RANKL. TNFα expression is crucial for PA-induced osteoclastogenesis, as shown by increased TNFα mRNA levels in PA-treated cells and abrogation of PA-stimulated osteoclastogenesis by TNFα neutralizing antibodies. In contrast, oleic acid (OA, 18:1) does not enhance osteoclast differentiation, leads to increased intracellular triglyceride accumulation, and inhibits PA-induced osteoclastogenesis. Adenovirus-mediated expression of diacylglycerol acyl transferase 1 (DGAT1), a gene involved in triglyceride synthesis, also inhibits PA-induced osteoclastogenesis, suggesting a protective role of DGAT1 for bone health. Accordingly, Dgat1 knockout mice have larger bone marrow-derived osteoclasts and decreased bone mass indices. In line with these findings, mice on a high-fat PA-enriched diet have a greater reduction in bone mass and structure than mice on a high-fat OA-enriched diet. Thus, we propose that TNFα mediates saturated fatty acid-induced osteoclastogenesis that can be prevented by DGAT activation or supplementation with OA.
Collapse
Affiliation(s)
- Zoi Drosatos-Tampakaki
- Division of Periodontics, Columbia University College of Dental Medicine, New York, NY, USA
| | - Konstantinos Drosatos
- Division of Preventive Medicine and Nutrition, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Yasemin Siegelin
- Division of Periodontics, Columbia University College of Dental Medicine, New York, NY, USA
| | - Shan Gong
- Division of Periodontics, Columbia University College of Dental Medicine, New York, NY, USA
| | | | | | - Ira J Goldberg
- Division of Preventive Medicine and Nutrition, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - P Christian Schulze
- Division of Cardiology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Ulrike Schulze-Späte
- Division of Periodontics, Columbia University College of Dental Medicine, New York, NY, USA
| |
Collapse
|
419
|
Shi X, Wei X, Koo I, Schmidt RH, Yin X, Kim SH, Vaughn A, McClain CJ, Arteel GE, Zhang X, Watson WH. Metabolomic analysis of the effects of chronic arsenic exposure in a mouse model of diet-induced Fatty liver disease. J Proteome Res 2013; 13:547-554. [PMID: 24328084 DOI: 10.1021/pr400719u] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Arsenic is a widely distributed environmental component that is associated with a variety of cancer and non-cancer adverse health effects. Additional lifestyle factors, such as diet, contribute to the manifestation of disease. Recently, arsenic was found to increase inflammation and liver injury in a dietary model of fatty liver disease. The purpose of the present study was to investigate potential mechanisms of this diet-environment interaction via a high-throughput metabolomics approach. GC×GC-TOF MS was used to identify metabolites that were significantly increased or decreased in the livers of mice fed a Western diet (a diet high in fat and cholesterol) and co-exposed to arsenic-contaminated drinking water. The results showed that there are distinct hepatic metabolomic profiles associated with eating a high fat diet, drinking arsenic-contaminated water, and the combination of the two. Among the metabolites that were decreased when arsenic exposure was combined with a high fat diet were short-chain and medium-chain fatty acid metabolites and the anti-inflammatory amino acid, glycine. These results are consistent with the observed increase in inflammation and cell death in the livers of these mice and point to potentially novel mechanisms by which these metabolic pathways could be altered by arsenic in the context of diet-induced fatty liver disease.
Collapse
Affiliation(s)
- Xue Shi
- Department of Chemistry, University of Louisville, Louisville, KY 40292
| | - Xiaoli Wei
- Department of Chemistry, University of Louisville, Louisville, KY 40292
| | - Imhoi Koo
- Department of Chemistry, University of Louisville, Louisville, KY 40292
| | - Robin H Schmidt
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292.,Department of Alcohol Research Center, University of Louisville, Louisville, KY 40292
| | - Xinmin Yin
- Department of Chemistry, University of Louisville, Louisville, KY 40292
| | - Seong Ho Kim
- Biostatistics Core, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Andrew Vaughn
- Department of Medicine, University of Louisville, Louisville, KY 40292
| | - Craig J McClain
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292.,Department of Alcohol Research Center, University of Louisville, Louisville, KY 40292.,Department of Medicine, University of Louisville, Louisville, KY 40292.,Department of Robley Rex VAMC, Louisville, KY 40292
| | - Gavin E Arteel
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292.,Department of Alcohol Research Center, University of Louisville, Louisville, KY 40292
| | - Xiang Zhang
- Department of Chemistry, University of Louisville, Louisville, KY 40292
| | - Walter H Watson
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292.,Department of Alcohol Research Center, University of Louisville, Louisville, KY 40292.,Department of Medicine, University of Louisville, Louisville, KY 40292
| |
Collapse
|
420
|
Moreno-Navarrete JM, Escoté X, Ortega F, Serino M, Campbell M, Michalski MC, Laville M, Xifra G, Luche E, Domingo P, Sabater M, Pardo G, Waget A, Salvador J, Giralt M, Rodriguez-Hermosa JI, Camps M, Kolditz CI, Viguerie N, Galitzky J, Decaunes P, Ricart W, Frühbeck G, Villarroya F, Mingrone G, Langin D, Zorzano A, Vidal H, Vendrell J, Burcelin R, Vidal-Puig A, Fernández-Real JM. A role for adipocyte-derived lipopolysaccharide-binding protein in inflammation- and obesity-associated adipose tissue dysfunction. Diabetologia 2013; 56:2524-37. [PMID: 23963324 DOI: 10.1007/s00125-013-3015-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 07/10/2013] [Indexed: 01/12/2023]
Abstract
AIMS/HYPOTHESIS Circulating lipopolysaccharide-binding protein (LBP) is an acute-phase reactant known to be increased in obesity. We hypothesised that LBP is produced by adipose tissue (AT) in association with obesity. METHODS LBP mRNA and LBP protein levels were analysed in AT from three cross-sectional (n = 210, n = 144 and n = 28) and three longitudinal (n = 8, n = 25, n = 20) human cohorts; in AT from genetically manipulated mice; in isolated adipocytes; and in human and murine cell lines. The effects of a high-fat diet and exposure to lipopolysaccharide (LPS) and peroxisome proliferator-activated receptor (PPAR)γ agonist were explored. Functional in vitro and ex vivo experiments were also performed. RESULTS LBP synthesis and release was demonstrated to increase with adipocyte differentiation in human and mouse AT, isolated adipocytes and human and mouse cell lines (Simpson-Golabi-Behmel syndrome [SGBS], human multipotent adipose-derived stem [hMAD] and 3T3-L1 cells). AT LBP expression was robustly associated with inflammatory markers and increased with metabolic deterioration and insulin resistance in two independent cross-sectional human cohorts. AT LBP also increased longitudinally with weight gain and excessive fat accretion in both humans and mice, and decreased with weight loss (in two other independent cohorts), in humans with acquired lipodystrophy, and after ex vivo exposure to PPARγ agonist. Inflammatory agents such as LPS and TNF-α led to increased AT LBP expression in vivo in mice and in vitro, while this effect was prevented in Cd14-knockout mice. Functionally, LBP knockdown using short hairpin (sh)RNA or anti-LBP antibody led to increases in markers of adipogenesis and decreased adipocyte inflammation in human adipocytes. CONCLUSIONS/INTERPRETATION Collectively, these findings suggest that LBP might have an essential role in inflammation- and obesity-associated AT dysfunction.
Collapse
Affiliation(s)
- José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Hospital Universitari 'Dr Josep Trueta', Carretera de França s/n, 17007, Girona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
421
|
Zeng H, Chi H. The interplay between regulatory T cells and metabolism in immune regulation. Oncoimmunology 2013; 2:e26586. [PMID: 24404429 PMCID: PMC3881602 DOI: 10.4161/onci.26586] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/21/2013] [Accepted: 09/23/2013] [Indexed: 01/04/2023] Open
Abstract
Regulatory T cells (Tregs) are crucial for peripheral tolerance and are intimately involved in immunological diseases and cancer. Recent studies have highlighted a key role for Tregs in metabolic disorders, for instance as they accumulate in the adipose tissue to protect against obesity-related inflammation and insulin resistance. Conversely, the generation and immunosuppressive functions of Tregs are influenced by both systemic and cellular metabolism. The nutritional status as well as metabolic cues such as those provided by leptin impinge upon the proliferation of Tregs. In addition, the mTOR-dependent lipid metabolism has a crucial role in programming the activity of Tregs under steady-state conditions as well as upon activation. This review discusses the intricate interaction between Tregs and metabolism, focusing on the roles of Tregs in systemic and local metabolic circuitries as well as on the regulation of Treg abundance and function by metabolic signals.
Collapse
Affiliation(s)
- Hu Zeng
- Department of Immunology; St. Jude Children's Research Hospital; Memphis, TN USA
| | - Hongbo Chi
- Department of Immunology; St. Jude Children's Research Hospital; Memphis, TN USA
| |
Collapse
|
422
|
Yin XL, Lu WN, Feng LY. Role of LPS/TLR4 signaling pathway in nonalcoholic fatty liver disease. Shijie Huaren Xiaohua Zazhi 2013; 21:2957-2962. [DOI: 10.11569/wcjd.v21.i28.2957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The LPS/TLR4 signaling pathway is involved in the pathogenesis of nonalcoholic fatty liver disease. The up-regulation of expression of LPS receptor TLR4 induces inflammatory response and promotes liver cell damage in nonalcoholic fatty liver disease. When nonalcoholic fatty liver disease occurs, disordered intestinal bacterial growth leads to increased production of LPS and increased permeability of the intestinal wall and therefore induces intestinal endotoxemia. Via MyD88-dependent and -independent pathways, LPS up-regulates the expression of TLR4 in Kupffer cells, activates NF-kB, induces strong inflammatory responses and mediates liver injury. This review aims to elucidate the role of the LPS/TLR4 signaling pathway in nonalcoholic fatty liver disease.
Collapse
|
423
|
Camell C, Smith CW. Dietary oleic acid increases m2 macrophages in the mesenteric adipose tissue. PLoS One 2013; 8:e75147. [PMID: 24098682 PMCID: PMC3787090 DOI: 10.1371/journal.pone.0075147] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 08/09/2013] [Indexed: 02/07/2023] Open
Abstract
Several studies have implicated fatty-acids as inflammatory regulators, suggesting that there may be a direct role for common dietary fatty-acids in regulating innate immune cells. In humans, a single high-fat meal increases systemic cytokines and leukocytes. In mice, short term high-fat feeding increases adipose tissue (AT) leukocytes and alters the inflammatory profile of AT macrophages. We have seen that short term high fat feeding to C57BL/6J male mice increases palmitic and oleic acid within AT depots, but oleic acid increase is highest in the mesenteric AT (MAT). In vitro, oleic acid increases M2 macrophage markers (CD206, MGL1, and ARG1) in a murine macrophage cell line, while addition of palmitic acid is able to inhibit that increase. Three day supplementation of a chow diet, with oleic acid, induced an increase in M2 macrophage markers in the MAT, but not in the epididymal AT. We tested whether increases in M2 macrophages occur during short term ad lib feeding of a high fat diet, containing oleic acid. Experiments revealed two distinct populations of macrophages were altered by a three day high milk-fat diet. One population, phenotypically intermediate for F4/80, showed diet-induced increases in CD206, an anti-inflammatory marker characteristic of M2 macrophages intrinsic to the AT. Evidence for a second population, phenotypically F4/80(HI)CD11b(HI) macrophages, showed increased association with the MAT following short term feeding that is dependent on the adhesion molecule, ICAM-1. Collectively, we have shown that short term feeding of a high-fat diet changes two population of macrophages, and that dietary oleic acid is responsible for increases in M2 macrophage polarization.
Collapse
Affiliation(s)
- Christina Camell
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - C. Wayne Smith
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Children’s Nutrition Research Center, Houston, Texas, United States of America
| |
Collapse
|
424
|
Melnik BC, Plewig G. Impaired Notch-MKP-1 signalling in hidradenitis suppurativa: an approach to pathogenesis by evidence from translational biology. Exp Dermatol 2013; 22:172-7. [PMID: 23489419 DOI: 10.1111/exd.12098] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2013] [Indexed: 12/13/2022]
Abstract
Recent findings in familial hidradenitis suppurativa (HS) demonstrated loss-of-function mutations of components of the γ-secretase (GS) complex leading to decreased protease cleaving activity, which may compromise canonical Notch signalling. Appropriate Notch signalling is of pivotal importance for maintaining the inner and outer root sheath of the hair follicle and skin appendages. This viewpoint on the pathogenesis of HS is primarily supported by circumstantial evidence derived from translational biology. Impaired Notch signalling is proposed to be the major pathogenic mechanism of HS. Deficient Notch signalling switches the fate of outer root sheath cells, resulting in conversion of hair follicles to keratin-enriched epidermal cysts. Impaired Notch signalling may compromise apocrine gland homoeostasis as well. Damage-associated molecular pattern molecules released by either ruptured epidermal cysts exposing keratin fibres or altered structural components of less maintained apocrine glands may both stimulate TLR-mediated innate immunity. All aggravating factors of HS, that is, smoking, obesity, skin occlusion, androgens and progesterone, may further promote inflammation by release of proinflammatory cytokines derived from activated monocyte/macrophages. Inappropriate Notch signalling may not only initiate inflammation in HS but may lead to insufficient feedback inhibition of overstimulated innate immunity. Regular Notch signalling via induction of MAPK phosphatase-1 (MKP-1) terminates TLR-MAPK-signalling in macrophages and IL-23 secreting DCs, the key players for Th17 cell polarization. Thus, impaired Notch signalling links HS to other Th17-driven comorbidities. All major therapeutic interventions in HS appear to attenuate increased MAPK activation of innate immune cells due to impaired Notch-mediated feedback regulation of innate immunity.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Osnabrück, Germany.
| | | |
Collapse
|
425
|
Activation and regulation of the pattern recognition receptors in obesity-induced adipose tissue inflammation and insulin resistance. Nutrients 2013; 5:3757-78. [PMID: 24064574 PMCID: PMC3798933 DOI: 10.3390/nu5093757] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 08/14/2013] [Accepted: 09/11/2013] [Indexed: 12/19/2022] Open
Abstract
Obesity-associated chronic tissue inflammation is a key contributing factor to type 2 diabetes mellitus, and a number of studies have clearly demonstrated that the immune system and metabolism are highly integrated. Recent advances in deciphering the various immune cells and signaling networks that link the immune and metabolic systems have contributed to our understanding of the pathogenesis of obesity-associated inflammation. Other recent studies have suggested that pattern recognition receptors in the innate immune system recognize various kinds of endogenous and exogenous ligands, and have a crucial role in initiating or promoting obesity-associated chronic inflammation. Importantly, these mediators act on insulin target cells or on insulin-producing cells impairing insulin sensitivity and its secretion. Here, we discuss how various pattern recognition receptors in the immune system underlie the etiology of obesity-associated inflammation and insulin resistance, with a particular focus on the TLR (Toll-like receptor) family protein Radioprotective 105 (RP105)/myeloid differentiation protein-1 (MD-1).
Collapse
|
426
|
Snodgrass RG, Huang S, Choi IW, Rutledge JC, Hwang DH. Inflammasome-mediated secretion of IL-1β in human monocytes through TLR2 activation; modulation by dietary fatty acids. THE JOURNAL OF IMMUNOLOGY 2013; 191:4337-47. [PMID: 24043885 DOI: 10.4049/jimmunol.1300298] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Many studies have shown that TLR4- and TLR2-deficient mice are protected from high-fat diet-induced inflammation and insulin resistance, suggesting that saturated fatty acids derived from the high-fat diet activate TLR-mediated proinflammatory signaling pathways and induce insulin resistance. However, evidence that palmitic acid, the major dietary saturated fatty acid, can directly activate TLR has not been demonstrated. In this article, we present multiple lines of evidence showing that palmitic acid directly activates TLR2, a major TLR expressed on human monocytes, by inducing heterodimerization with TLR1 in an NADPH oxidase-dependent manner. Dimerization of TLR2 with TLR1 was inhibited by the n-3 fatty acid docosahexaenoic acid. Activation of TLR2 by palmitic acid leads to expression of pro-IL-1β that is cleaved by caspase-1, which is constitutively present in monocytes, to release mature IL-1β. Our results reveal mechanistic insight about how palmitic acid activates TLR2, upregulates NALP3 expression, and induces inflammasome-mediated IL-1β production in human monocytes, which can trigger enhanced inflammation in peripheral tissues, and suggest that these processes are dynamically modulated by the types of dietary fat we consume.
Collapse
Affiliation(s)
- Ryan G Snodgrass
- U.S. Department of Agriculture, Agricultural Research Service Western Human Nutrition Research Center, Davis, CA 95616
| | | | | | | | | |
Collapse
|
427
|
Lachance C, Wojewodka G, Skinner TAA, Guilbault C, De Sanctis JB, Radzioch D. Fenretinide corrects the imbalance between omega-6 to omega-3 polyunsaturated fatty acids and inhibits macrophage inflammatory mediators via the ERK pathway. PLoS One 2013; 8:e74875. [PMID: 24069363 PMCID: PMC3771966 DOI: 10.1371/journal.pone.0074875] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 08/09/2013] [Indexed: 12/24/2022] Open
Abstract
We previously identified Fragile X-related protein 1 (FXR1) as an RNA-binding protein involved in the post-transcriptional control of TNF and other cytokines in macrophages. Macrophages derived from FXR1-KO mice overexpress several inflammatory cytokines including TNF. Recently, we showed that fenretinide (4HPR) is able to inhibit several inflammatory cytokines in the lungs of cystic fibrosis mice, which also have abnormal immune responses. Therefore, we hypothesized that 4HPR might also be able to downregulate excessive inflammation even in macrophages with ablated FXR1. Indeed, our results demonstrate that 4HPR inhibited the excessive production of inflammatory mediators, including TNF, IL-6, CCL2 and CCL-5 in LPS-stimulated FXR1-KO macrophages, by selectively inhibiting phosphorylation of ERK1/2, which is naturally more phosphorylated in FXR1-KO cells. We also found that LPS stimulation of FXR1-KO macrophages led to significantly higher ratio of arachidonic acid/docosahexaenoic acid than observed in FXR1-WT macrophages. Interestingly, treatment with 4HPR was associated with the normalization of arachidonic acid/docosahexaenoic acid ratio in macrophages, which we found to impact phosphorylation of ERK1/2. Overall, this study shows for the first time that 4HPR modulates inflammatory cytokine expression in macrophages by correcting a phospholipid-bound fatty acid imbalance that impacts the phosphorylation of ERK1/2.
Collapse
Affiliation(s)
- Claude Lachance
- McGill University, Department of Medicine and Department of Human Genetics, McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Gabriella Wojewodka
- McGill University, Department of Medicine and Department of Human Genetics, McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Tom A. A. Skinner
- McGill University, Department of Medicine and Department of Human Genetics, McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Claudine Guilbault
- McGill University, Department of Medicine and Department of Human Genetics, McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Juan B. De Sanctis
- Central University of Venezuela, Institute of Immunology, Caracas, Venezuela
| | - Danuta Radzioch
- McGill University, Department of Medicine and Department of Human Genetics, McGill University Health Center Research Institute, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
428
|
L'homme L, Esser N, Riva L, Scheen A, Paquot N, Piette J, Legrand-Poels S. Unsaturated fatty acids prevent activation of NLRP3 inflammasome in human monocytes/macrophages. J Lipid Res 2013; 54:2998-3008. [PMID: 24006511 DOI: 10.1194/jlr.m037861] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The NLRP3 inflammasome is involved in many obesity-associated diseases, such as type 2 diabetes, atherosclerosis, and gouty arthritis, through its ability to induce interleukin (IL)-1β release. The molecular link between obesity and inflammasome activation is still unclear, but free fatty acids have been proposed as one triggering event. Here we reported opposite effects of saturated fatty acids (SFAs) compared with unsaturated fatty acids (UFAs) on NLRP3 inflammasome in human monocytes/macrophages. Palmitate and stearate, both SFAs, triggered IL-1β secretion in a caspase-1/ASC/NLRP3-dependent pathway. Unlike SFAs, the UFAs oleate and linoleate did not lead to IL-1β secretion. In addition, they totally prevented the IL-1β release induced by SFAs and, with less efficiency, by a broad range of NLRP3 inducers, including nigericin, alum, and monosodium urate. UFAs did not affect the transcriptional effect of SFAs, suggesting a specific effect on the NLRP3 activation. These results provide a new anti-inflammatory mechanism of UFAs by preventing the activation of the NLRP3 inflammasome and, therefore, IL-1β processing. By this way, UFAs might play a protective role in NLRP3-associated diseases.
Collapse
Affiliation(s)
- Laurent L'homme
- Laboratory of Virology and Immunology, GIGA-Signal Transduction
| | | | | | | | | | | | | |
Collapse
|
429
|
Xu G, Zhang Z, Wei J, Zhang Y, Zhang Y, Guo L, Liu X. microR-142-3p down-regulates IRAK-1 in response to Mycobacterium bovis BCG infection in macrophages. Tuberculosis (Edinb) 2013; 93:606-11. [PMID: 24053976 DOI: 10.1016/j.tube.2013.08.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 07/31/2013] [Accepted: 08/10/2013] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs) have been demonstrated to play a pivotal role in the regulation of target gene expression at the post-transcriptional level. In order to better understand the role of miRNA in the immunological regulation of macrophages against Mycobacterium bovis BCG infection, we explored the alteration of immune-related miRNA profile in macrophage RAW264.7 cells in response to BCG infection in this study. Our results demonstrated that miR-142-3p was a potential to negatively regulate the production of pro-inflammatory mediators NF-κB (NF-κB1), TNF-α and IL-6 in the macrophages in part through a mechanism of targeting IRAK-1 gene and post-transcriptionally down-regulating IRAK-1 protein expression.
Collapse
Affiliation(s)
- Guangxian Xu
- General Hospital of Ningxia Medical University, Yinchuan 750004, China; School of Laboratory Medicine, Ningxia Medical University, Yinchuan 750004, China.
| | | | | | | | | | | | | |
Collapse
|
430
|
Myles IA, Fontecilla NM, Janelsins BM, Vithayathil PJ, Segre JA, Datta SK. Parental dietary fat intake alters offspring microbiome and immunity. THE JOURNAL OF IMMUNOLOGY 2013; 191:3200-9. [PMID: 23935191 DOI: 10.4049/jimmunol.1301057] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Mechanisms underlying modern increases in prevalence of human inflammatory diseases remain unclear. The hygiene hypothesis postulates that decreased microbial exposure has, in part, driven this immune dysregulation. However, dietary fatty acids also influence immunity, partially through modulation of responses to microbes. Prior reports have described the direct effects of high-fat diets on the gut microbiome and inflammation, and some have additionally shown metabolic consequences for offspring. Our study sought to expand on these previous observations to identify the effects of parental diet on offspring immunity using mouse models to provide insights into challenging aspects of human health. To test the hypothesis that parental dietary fat consumption during gestation and lactation influences offspring immunity, we compared pups of mice fed either a Western diet (WD) fatty acid profile or a standard low-fat diet. All pups were weaned onto the control diet to specifically test the effects of early developmental fat exposure on immune development. Pups from WD breeders were not obese or diabetic, but still had worse outcomes in models of infection, autoimmunity, and allergic sensitization. They had heightened colonic inflammatory responses, with increased circulating bacterial LPS and muted systemic LPS responsiveness. These deleterious impacts of the WD were associated with alterations of the offspring gut microbiome. These results indicate that parental fat consumption can leave a "lard legacy" impacting offspring immunity and suggest inheritable microbiota may contribute to the modern patterns of human health and disease.
Collapse
Affiliation(s)
- Ian A Myles
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Natalia M Fontecilla
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Brian M Janelsins
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Paul J Vithayathil
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Julia A Segre
- Epithelial Biology Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Sandip K Datta
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
431
|
Lipids derived from virulent Francisella tularensis broadly inhibit pulmonary inflammation via toll-like receptor 2 and peroxisome proliferator-activated receptor α. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:1531-40. [PMID: 23925884 DOI: 10.1128/cvi.00319-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Francisella tularensis is a Gram-negative facultative intracellular pathogen that causes an acute lethal respiratory disease in humans. The heightened virulence of the pathogen is linked to its unique ability to inhibit Toll-like receptor (TLR)-mediated inflammatory responses. The bacterial component and mechanism of this inhibition are unknown. Here we show that lipids isolated from virulent but not attenuated strains of F. tularensis are not detected by host cells, inhibit production of proinflammatory cytokines by primary macrophages in response to known TLR ligands, and suppress neutrophil recruitment in vivo. We further show that lipid-mediated inhibition of inflammation is dependent on TLR2, MyD88, and the nuclear hormone and fatty acid receptor peroxisome proliferator-activated receptor α (PPARα). Pathogen lipid-mediated interference with inflammatory responses through the engagement of TLR2 and PPARα represents a novel manipulation of host signaling pathways consistent with the ability of highly virulent F. tularensis to efficiently evade host immune responses.
Collapse
|
432
|
Kavanagh K, Wylie AT, Tucker KL, Hamp TJ, Gharaibeh RZ, Fodor AA, Cullen JMC. Dietary fructose induces endotoxemia and hepatic injury in calorically controlled primates. Am J Clin Nutr 2013; 98:349-57. [PMID: 23783298 PMCID: PMC3712547 DOI: 10.3945/ajcn.112.057331] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Controversy exists regarding the causative role of dietary fructose in obesity and fatty liver diseases. Clinical trials have indicated that negative health consequences may occur only when fructose is consumed within excess calories. Animal studies have suggested that fructose impairs intestinal integrity and leads to hepatic steatosis (HS). OBJECTIVES We assessed nonhuman primates after chronic ad libitum and short-term calorically controlled consumption of a high-fructose (HFr), low-fat diet (24% of calories). Microbial translocation (MT), microbiome, and metabolic health indexes were evaluated. DESIGN Seventeen monkeys fed 0.3–7 y of an HFr ad libitum diet were compared with 10 monkeys fed a low-fructose, low-fat diet (control). Ten middle-aged, weight-stable, fructose-naive monkeys were stratified into HFr and control groups fed for 6 wk at caloric amounts required to maintain weight stability. Metabolic endpoints, feces, liver, small and large intestinal biopsies, and portal blood samples were collected. RESULTS Monkeys allowed ad libitum HFr developed HS in contrast to the control diet, and the extent of ectopic fat was related to the duration of feeding. Diabetes incidence also increased. Monkeys that consumed calorically controlled HFr showed significant increases in biomarkers of liver damage, endotoxemia, and MT indexes and a trend for greater hepatitis that was related to MT; however, HS did not develop. CONCLUSIONS Even in the absence of weight gain, fructose rapidly causes liver damage that we suggest is secondary to endotoxemia and MT. HS relates to the duration of fructose consumption and total calories consumed. These data support fructose inducing both MT and ectopic fat deposition in primates.
Collapse
Affiliation(s)
- Kylie Kavanagh
- Department of Pathology Section on Comparative Medicine and Lipid Sciences, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27127, USA.
| | | | | | | | | | | | | |
Collapse
|
433
|
Mueller GA, Pedersen LC, Lih FB, Glesner J, Moon AF, Chapman MD, Tomer KB, London RE, Pomés A. The novel structure of the cockroach allergen Bla g 1 has implications for allergenicity and exposure assessment. J Allergy Clin Immunol 2013; 132:1420-6. [PMID: 23915714 DOI: 10.1016/j.jaci.2013.06.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 05/28/2013] [Accepted: 06/19/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Sensitization to cockroach allergens is a major risk factor for asthma. The cockroach allergen Bla g 1 has multiple repeats of approximately 100 amino acids, but the fold of the protein and its biological function are unknown. OBJECTIVE We sought to determine the structure of Bla g 1, investigate the implications for allergic disease, and standardize cockroach exposure assays. METHODS nBla g 1 and recombinant constructs were compared by using ELISA with specific murine IgG and human IgE. The structure of Bla g 1 was determined by x-ray crystallography. Mass spectrometry and nuclear magnetic resonance spectroscopy were used to examine the ligand-binding properties of the allergen. RESULTS The structure of an rBla g 1 construct with comparable IgE and IgG reactivity to the natural allergen was solved by x-ray crystallography. The Bla g 1 repeat forms a novel fold with 6 helices. Two repeats encapsulate a large and nearly spherical hydrophobic cavity, defining the basic structural unit. Lipids in the cavity varied depending on the allergen origin. Palmitic, oleic, and stearic acids were associated with nBla g 1 from cockroach frass. One unit of Bla g 1 was equivalent to 104 ng of allergen. CONCLUSIONS Bla g 1 has a novel fold with a capacity to bind various lipids, which suggests a digestive function associated with nonspecific transport of lipid molecules in cockroaches. Defining the basic structural unit of Bla g 1 facilitates the standardization of assays in absolute units for the assessment of environmental allergen exposure.
Collapse
Affiliation(s)
- Geoffrey A Mueller
- Laboratory of Structural Biology, National Institute of Environmental Health Sciences, Research Triangle Park, NC.
| | | | | | | | | | | | | | | | | |
Collapse
|
434
|
Borén J, Taskinen MR, Olofsson SO, Levin M. Ectopic lipid storage and insulin resistance: a harmful relationship. J Intern Med 2013; 274:25-40. [PMID: 23551521 DOI: 10.1111/joim.12071] [Citation(s) in RCA: 164] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Obesity increases the risk of metabolic diseases, including insulin resistance and type 2 diabetes, as well as cardiovascular disease. In addition to lipid accumulation in adipose tissue, obesity is associated with increased lipid storage in ectopic tissues, such as skeletal muscle and liver. Furthermore, lipid accumulation in the heart may result in cardiac dysfunction and heart failure. It has recently been demonstrated that intracellular lipid accumulation in ectopic tissues leads to pathological responses and impaired insulin signalling. Here, we will review the current understanding of how lipid storage and lipid droplet physiology affect the risk of developing metabolic diseases.
Collapse
Affiliation(s)
- J Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | | | | | | |
Collapse
|
435
|
Ruiz-Núñez B, Pruimboom L, Dijck-Brouwer DJ, Muskiet FA. Lifestyle and nutritional imbalances associated with Western diseases: causes and consequences of chronic systemic low-grade inflammation in an evolutionary context. J Nutr Biochem 2013; 24:1183-201. [DOI: 10.1016/j.jnutbio.2013.02.009] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 01/03/2013] [Accepted: 02/18/2013] [Indexed: 12/26/2022]
|
436
|
Xie Y, Matsumoto H, Nalbantoglu ILK, Kerr TA, Luo J, Rubin DC, Kennedy S, Davidson NO. Intestine-Specific Mttp Deletion Increases the Severity of Experimental Colitis and Leads to Greater Tumor Burden in a Model of Colitis Associated Cancer. PLoS One 2013; 8:e67819. [PMID: 23805328 PMCID: PMC3689718 DOI: 10.1371/journal.pone.0067819] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 05/22/2013] [Indexed: 12/19/2022] Open
Abstract
Background Gut derived lipid factors have been implicated in systemic injury and inflammation but the precise pathways involved are unknown. In addition, dietary fat intake and obesity are independent risk factors for the development of colorectal cancer. Here we studied the severity of experimental colitis and the development of colitis associated cancer (CAC) in mice with an inducible block in chylomicron secretion and fat malabsorption, following intestine-specific deletion of microsomal triglyceride transfer protein (Mttp-IKO). Methodology/Principal Findings Mttp-IKO mice exhibited more severe injury with ∼90% mortality following dextran sodium sulfate (DSS) induced colitis, compared to <20% in controls. Intestinal permeability was increased in Mttp-IKO mice compared to controls, both at baseline and after DSS administration, in association with increased circulating levels of TNFα. DSS treatment increased colonic mRNA expression of IL-1β and IL-17A as well as inflammasome expression in both genotypes, but the abundance of TNFα was selectively increased in DSS treated Mttp-IKO mice. There was a 2-fold increase in colonic tumor burden in Mttp-IKO mice following azoxymethane/DSS treatment, which was associated with increased colonic inflammation as well as alterations in cytokine expression. To examine the pathways by which alterations in fatty acid abundance might interact with cytokine signaling to regulate colonic epithelial growth, we used primary murine myofibroblasts to demonstrate that palmitate induced expression of amphiregulin and epiregulin and augmented the increase in both of these growth mediators when added to IL-1βor to TNFα. Conclusions These studies demonstrate that Mttp-IKO mice, despite absorbing virtually no dietary fat, exhibit augmented fatty acid dependent signaling that in turn exacerbates colonic injury and increases tumor formation.
Collapse
Affiliation(s)
- Yan Xie
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Hitoshi Matsumoto
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - ILKe Nalbantoglu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Thomas A. Kerr
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jianyang Luo
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Deborah C. Rubin
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Susan Kennedy
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Nicholas O. Davidson
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
437
|
Gao M, Ma Y, Liu D. Rutin suppresses palmitic acids-triggered inflammation in macrophages and blocks high fat diet-induced obesity and fatty liver in mice. Pharm Res 2013; 30:2940-50. [PMID: 23783345 DOI: 10.1007/s11095-013-1125-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 06/12/2013] [Indexed: 12/15/2022]
Abstract
PURPOSE To elucidate the mechanism of rutin in blocking macrophage-mediated inflammation and high fat diet-induced obesity and fatty liver. METHODS Both in vitro and in vivo approaches were taken in evaluating the effects of rutin on palmitic acids-triggered inflammation in cultured macrophages, and on weight gain and development of fatty liver of mice fed a high fat diet. RESULTS Palmitic acids increase mRNA levels of pro-inflammatory cytokines, and elevate the production of TNFα in cultured macrophages. Pre-exposure of rutin to cells greatly suppressed these elevations. The suppressed inflammation by rutin was correlated with a decrease in transcription of genes responsible for ER stress and production of reactive oxygen species. In vivo, rutin protects mice from high fat diet-induced obesity, fatty liver and insulin resistance. The protective effects were associated with lack of hypertrophy and crown-like structures in the white adipose tissue, decreased mRNA levels of marker genes for macrophages including F4/80, Cd11c and Cd68, and repressed transcription of genes involved in chronic inflammation such as Mcp1 and Tnfα in white adipose tissue. In addition, rutin increases the expression of genes responsible for energy expenditure in brown adipose tissue including Pgc1α and Dio2. Furthermore, rutin suppresses transcription of Srebp1c and Cd36 in the liver, leading to a blockade of fatty liver development. CONCLUSION These results suggest that supplementation of rutin is a promising strategy for blocking macrophage-mediated inflammation and inflammation-induced obesity and its associated complications.
Collapse
Affiliation(s)
- Mingming Gao
- Department of Pharmaceutical and Biomedical Sciences College of Pharmacy, University of Georgia, 450 Pharmacy South 250 West Green Street, Athens, Georgia, 30602, USA
| | | | | |
Collapse
|
438
|
Muñoz A, Costa M. Nutritionally mediated oxidative stress and inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:610950. [PMID: 23844276 PMCID: PMC3697417 DOI: 10.1155/2013/610950] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 04/23/2013] [Indexed: 01/04/2023]
Abstract
There are many sources of nutritionally mediated oxidative stress that trigger inflammatory cascades along short and long time frames. These events are primarily mediated via NF κ B. On the short-term scale postprandial inflammation is characterized by an increase in circulating levels of IL-6 and TNF- α and is mirrored on the long-term by proinflammatory gene expression changes in the adipocytes and peripheral blood mononuclear cells (PBMCs) of obese individuals. Specifically the upregulation of CCL2/MCP-1, CCL3/MIP-1 α , CCL4/MIP-1 β , CXCL2/MIP-2 α , and CXCL3/MIP-2 β is noted because these changes have been observed in both adipocytes and PBMC of obese humans. In comparing numerous human intervention studies it is clear that pro-inflammatory and anti-inflammatory consumption choices mediate gene expression in humans adipocytes and peripheral blood mononuclear cells. Arachidonic acid and saturated fatty acids (SFAs) both demonstrate an ability to increase pro-inflammatory IL-8 along with numerous other inflammatory factors including IL-6, TNF α , IL-1 β , and CXCL1 for arachidonic acid and IGB2 and CTSS for SFA. Antioxidant rich foods including olive oil, fruits, and vegetables all demonstrate an ability to lower levels of IL-6 in PBMCs. Thus, dietary choices play a complex role in the mediation of unavoidable oxidative stress and can serve to exacerbate or dampen the level of inflammation.
Collapse
Affiliation(s)
- Alexandra Muñoz
- New York University School of Medicine, Nelson Institute of Environmental Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | - Max Costa
- New York University School of Medicine, Nelson Institute of Environmental Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
- Department of Environmental Medicine, New York University, 57 Old Forge Road, Tuxedo, NY 10987, USA
| |
Collapse
|
439
|
Gupte AA, Lyon CJ, Hsueh WA. Nuclear factor (erythroid-derived 2)-like-2 factor (Nrf2), a key regulator of the antioxidant response to protect against atherosclerosis and nonalcoholic steatohepatitis. Curr Diab Rep 2013; 13:362-71. [PMID: 23475581 DOI: 10.1007/s11892-013-0372-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tissue oxidative stress is a common hallmark of atherosclerosis and non-alcoholic steatohepatitis (NASH), 2 conditions linked epidemiologically and pathophysiologically. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is the master regulator of inducible antioxidant responses, that can attenuate cellular injury from oxidative stress induced by obesity and other redox insults. Nrf2 expression and activation is reduced in mouse and human vessels that harbor accelerated atherosclerosis and in livers with histologic criteria of NASH. Systemic antioxidants have thus been attractive therapeutic targets, but clinical trials have been largely unsuccessful in improving cardiovascular health. Macrophage-selective Nrf2 activation may, however, provide an approach to reduce vascular and hepatocyte injury without the complications of systemic antioxidants, since macrophages play key roles in the development and progression of both atherosclerosis and NASH. In this article, we review the common mechanisms of oxidative stress and inflammation in atherosclerosis and NASH, and discuss the role of Nrf2 in vascular and hepatocyte protection.
Collapse
Affiliation(s)
- Anisha A Gupte
- The Methodist Diabetes and Metabolism Institute, Center for Diabetes Research in The Methodist Hospital Research Institute, Weill Cornell Medical College, R8-111, 6670 Bertner Ave, Houston, 77030 TX, USA
| | | | | |
Collapse
|
440
|
Wei J, Huang X, Zhang Z, Jia W, Zhao Z, Zhang Y, Liu X, Xu G. MyD88 as a target of microRNA-203 in regulation of lipopolysaccharide or Bacille Calmette-Guerin induced inflammatory response of macrophage RAW264.7 cells. Mol Immunol 2013; 55:303-9. [PMID: 23522925 DOI: 10.1016/j.molimm.2013.03.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 02/27/2013] [Accepted: 03/04/2013] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) have been demonstrated to play a pivotal role in the regulation of target gene expression at the post-transcriptional level. In order to better understand the role of microRNA-203 (miR-203) in the immunological regulation, the function of miR-203 was explored in the macrophage RAW264.7 cells against lipopolysaccharide (LPS) or Bacille Calmette-Guerin (BCG) stimulation. The results evidenced that myeloid differentiation factor 88 (MyD88) was a novel target of miR-203, miR-203 was capable of directly targeting the 3' untranslated region (3'UTR) of MyD88 and post-transcriptionally down-regulating the expression of protein. In addition, an overexpression of miR-203 in RAW264.7 cells was correlated with repressions of MyD88, as well as its downstream signaling of NF-κB (NF-κB1), TNF-α and IL-6. These results suggest that miR-203 may be an important regulator in macrophages against LPS or mycobacteria infection, which may through a mechanism of negatively regulating MyD88-dependent Toll-like receptors signaling pathway.
Collapse
Affiliation(s)
- Jun Wei
- General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | | | | | | | | | | | | | | |
Collapse
|
441
|
Estadella D, da Penha Oller do Nascimento CM, Oyama LM, Ribeiro EB, Dâmaso AR, de Piano A. Lipotoxicity: effects of dietary saturated and transfatty acids. Mediators Inflamm 2013; 2013:137579. [PMID: 23509418 PMCID: PMC3572653 DOI: 10.1155/2013/137579] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 12/19/2012] [Accepted: 12/19/2012] [Indexed: 01/18/2023] Open
Abstract
The ingestion of excessive amounts of saturated fatty acids (SFAs) and transfatty acids (TFAs) is considered to be a risk factor for cardiovascular diseases, insulin resistance, dyslipidemia, and obesity. The focus of this paper was to elucidate the influence of dietary SFA and TFA intake on the promotion of lipotoxicity to the liver and cardiovascular, endothelial, and gut microbiota systems, as well as on insulin resistance and endoplasmic reticulum stress. The saturated and transfatty acids favor a proinflammatory state leading to insulin resistance. These fatty acids can be involved in several inflammatory pathways, contributing to disease progression in chronic inflammation, autoimmunity, allergy, cancer, atherosclerosis, hypertension, and heart hypertrophy as well as other metabolic and degenerative diseases. As a consequence, lipotoxicity may occur in several target organs by direct effects, represented by inflammation pathways, and through indirect effects, including an important alteration in the gut microbiota associated with endotoxemia. Interactions between these pathways may perpetuate a feedback process that exacerbates an inflammatory state. The importance of lifestyle modification, including an improved diet, is recommended as a strategy for treatment of these diseases.
Collapse
Affiliation(s)
- Débora Estadella
- Programa de Pós-Graduação em Nutrição, Disciplina de Fisiologia da Nutrição, EPM, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862 Edifício de Ciências Biomédicas, 2 andar, Vila Clementino, 04023-060 São Paulo, SP, Brazil
| | - Claudia M. da Penha Oller do Nascimento
- Programa de Pós-Graduação em Nutrição, Disciplina de Fisiologia da Nutrição, EPM, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862 Edifício de Ciências Biomédicas, 2 andar, Vila Clementino, 04023-060 São Paulo, SP, Brazil
| | - Lila M. Oyama
- Programa de Pós-Graduação em Nutrição, Disciplina de Fisiologia da Nutrição, EPM, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862 Edifício de Ciências Biomédicas, 2 andar, Vila Clementino, 04023-060 São Paulo, SP, Brazil
| | - Eliane B. Ribeiro
- Programa de Pós-Graduação em Nutrição, Disciplina de Fisiologia da Nutrição, EPM, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862 Edifício de Ciências Biomédicas, 2 andar, Vila Clementino, 04023-060 São Paulo, SP, Brazil
| | - Ana R. Dâmaso
- Departamento de Biociências, UNIFESP, Campus Baixada Santista, 11060-001 Santos, SP, Brazil
| | - Aline de Piano
- Programa de Pós-Graduação em Nutrição, Disciplina de Fisiologia da Nutrição, EPM, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862 Edifício de Ciências Biomédicas, 2 andar, Vila Clementino, 04023-060 São Paulo, SP, Brazil
| |
Collapse
|
442
|
Lager S, Gaccioli F, Ramirez VI, Jones HN, Jansson T, Powell TL. Oleic acid stimulates system A amino acid transport in primary human trophoblast cells mediated by toll-like receptor 4. J Lipid Res 2012; 54:725-733. [PMID: 23275648 DOI: 10.1194/jlr.m033050] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Obese women have an increased risk to deliver large babies. However, the mechanisms underlying fetal overgrowth in these pregnancies are not well understood. Obese pregnant women typically have elevated circulating lipid levels. We tested the hypothesis that fatty acids stimulate placental amino acid transport, mediated via toll-like receptor 4 (TLR4) and mammalian target of rapamycin (mTOR) signaling pathways. Circulating NEFA levels and placental TLR4 expression were assessed in women with varying prepregnancy body mass index (BMI). The effects of oleic acid on system A and system L amino acid transport, and on the activation of the mTOR (4EBP1, S6K1, rpS6), TLR4 (IĸB, JNK, p38 MAPK), and STAT3 signaling pathways were determined in cultured primary human trophoblast cells. Maternal circulating NEFAs (n = 33), but not placental TLR4 mRNA expression (n = 16), correlated positively with BMI (P < 0.05). Oleic acid increased trophoblast JNK and STAT3 phosphorylation (P < 0.05), whereas mTOR activity was unaffected. Furthermore, oleic acid doubled trophoblast system A activity (P < 0.05), without affecting system L activity. siRNA-mediated silencing of TLR4 expression prevented the stimulatory effect of oleic acid on system A activity. Our data suggest that maternal fatty acids can increase placental nutrient transport via TLR4, thereby potentially affecting fetal growth.
Collapse
Affiliation(s)
- Susanne Lager
- Center for Pregnancy and Newborn Research, Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX; Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Francesca Gaccioli
- Center for Pregnancy and Newborn Research, Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX
| | - Vanessa I Ramirez
- Center for Pregnancy and Newborn Research, Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX
| | - Helen N Jones
- Center for Molecular Fetal Therapy, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Thomas Jansson
- Center for Pregnancy and Newborn Research, Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX
| | - Theresa L Powell
- Center for Pregnancy and Newborn Research, Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX
| |
Collapse
|
443
|
Li X, Gonzalez O, Shen X, Barnhart S, Kramer F, Kanter JE, Vivekanandan-Giri A, Tsuchiya K, Handa P, Pennathur S, Kim F, Coleman RA, Schaffer JE, Bornfeldt KE. Endothelial acyl-CoA synthetase 1 is not required for inflammatory and apoptotic effects of a saturated fatty acid-rich environment. Arterioscler Thromb Vasc Biol 2012; 33:232-40. [PMID: 23241406 DOI: 10.1161/atvbaha.112.252239] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Saturated fatty acids, such as palmitic and stearic acid, cause detrimental effects in endothelial cells and have been suggested to contribute to macrophage accumulation in adipose tissue and the vascular wall, in states of obesity and insulin resistance. Long-chain fatty acids are believed to require conversion into acyl-CoA derivatives to exert most of their detrimental effects, a reaction catalyzed by acyl-CoA synthetases (ACSLs). The objective of this study was to investigate the role of ACSL1, an ACSL isoform previously shown to mediate inflammatory effects in myeloid cells, in regulating endothelial cell responses to a saturated fatty acid-rich environment in vitro and in vivo. METHODS AND RESULTS Saturated fatty acids caused increased inflammatory activation, endoplasmic reticulum stress, and apoptosis in mouse microvascular endothelial cells. Forced ACSL1 overexpression exacerbated the effects of saturated fatty acids on apoptosis and endoplasmic reticulum stress. However, endothelial ACSL1 deficiency did not protect against the effects of saturated fatty acids in vitro, nor did it protect insulin-resistant mice fed a saturated fatty acid-rich diet from macrophage adipose tissue accumulation or increased aortic adhesion molecule expression. CONCLUSIONS Endothelial ACSL1 is not required for inflammatory and apoptotic effects of a saturated fatty acid-rich environment.
Collapse
Affiliation(s)
- Xin Li
- Departments of Pathology, University of Washington, Seattle WA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
444
|
Bagaitkar J, Zeller I, Renaud DE, Scott DA. Cotinine inhibits the pro-inflammatory response initiated by multiple cell surface Toll-like receptors in monocytic THP cells. Tob Induc Dis 2012. [PMID: 23176969 PMCID: PMC3541225 DOI: 10.1186/1617-9625-10-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background The primary, stable metabolite of nicotine [(S)-3-(1-methyl-2-pyrrolidinyl) pyridine] in humans is cotinine [(S)-1-methyl-5-(3-pyridinyl)-2-pyrrolidinone]. We have previously shown that cotinine exposure induces convergence and amplification of the GSK3β-dependent PI3 kinase and cholinergic anti-inflammatory systems. The consequence is reduced pro-inflammatory cytokine secretion by human monocytes responding to bacteria or LPS, a TLR4 agonist. Findings Here we show that cotinine-induced inflammatory suppression may not be restricted to individual Toll-like receptors (TLRs). Indeed, in monocytic cells, cotinine suppresses the cytokine production that is normally resultant upon agonist-specific engagement of all of the major surface exposed TLRs (TLR 2/1; 2/6; 4 and 5), although the degree of suppression varies by TLR. Conclusions These results provide further mechanistic insight into the increased susceptibility to multiple bacterial infections known to occur in smokers. They also establish THP-1 cells as a potentially suitable model with which to study the influence of tobacco components and metabolites on TLR-initiated inflammatory events.
Collapse
Affiliation(s)
- Juhi Bagaitkar
- Microbiology and Immunology, University of Louisville, Louisville, KY, 40292, USA.
| | | | | | | |
Collapse
|
445
|
Pillon NJ, Arane K, Bilan PJ, Chiu TT, Klip A. Muscle cells challenged with saturated fatty acids mount an autonomous inflammatory response that activates macrophages. Cell Commun Signal 2012; 10:30. [PMID: 23078640 PMCID: PMC3507850 DOI: 10.1186/1478-811x-10-30] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 10/16/2012] [Indexed: 12/11/2022] Open
Abstract
Obesity is associated with chronic low-grade inflammation. Within adipose tissue of mice fed a high fat diet, resident and infiltrating macrophages assume a pro-inflammatory phenotype characterized by the production of cytokines which in turn impact on the surrounding tissue. However, inflammation is not restricted to adipose tissue and high fat-feeding is responsible for a significant increase in pro-inflammatory cytokine expression in muscle. Although skeletal muscle is the major disposer of dietary glucose and a major determinant of glycemia, the origin and consequence of muscle inflammation in the development of insulin resistance are poorly understood. We used a cell culture approach to investigate the vectorial crosstalk between muscle cells and macrophages upon exposure to physiological, low levels of saturated and unsaturated fatty acids. Inflammatory pathway activation and cytokine expression were analyzed in L6 muscle cells expressing myc-tagged GLUT4 (L6GLUT4myc) exposed to 0.2 mM palmitate or palmitoleate. Conditioned media thereof, free of fatty acids, were then tested for their ability to activate RAW264.7 macrophages. Palmitate -but not palmitoleate- induced IL-6, TNFα and CCL2 expression in muscle cells, through activation of the NF-κB pathway. Palmitate (0.2 mM) alone did not induce insulin resistance in muscle cells, yet conditioned media from palmitate-challenged muscle cells selectively activated macrophages towards a pro-inflammatory phenotype. These results demonstrate that low concentrations of palmitate activate autonomous inflammation in muscle cells to release factors that turn macrophages pro-inflammatory. We hypothesize that saturated fat-induced, low-grade muscle cell inflammation may trigger resident skeletal muscle macrophage polarization, possibly contributing to insulin resistance in vivo.
Collapse
Affiliation(s)
- Nicolas J Pillon
- Program in Cell Biology, The Hospital for Sick Children,Toronto, Ontario, M5G 1X8, Canada.
| | | | | | | | | |
Collapse
|