1
|
Zhou Y, Zhou Z. Unraveling the causal link: fatty acids and inflammatory bowel disease. Front Immunol 2024; 15:1405790. [PMID: 39119343 PMCID: PMC11306040 DOI: 10.3389/fimmu.2024.1405790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Background Previous observational studies have revealed the strong relationship between fatty acids (FA) and inflammatory bowel disease (IBD). Nonetheless, due to the inherent limitations of retrospective research, the causality between the two has not been clearly established. Methods Genetic variants associated with the 17 FA indicators were derived from genome-wide association studies. Summary statistics for the discovery cohort and testing cohort for IBD, including ulcerative colitis (UC) and Crohn's disease (CD), were available from IIBDGC and FinnGen, respectively. Bidirectional MR analysis and sensitivity analysis with multiple measures were applied to comprehensively investigate the causal link between FA and IBD. Results Combining the results of various MR methods, the validation of testing cohort, and the merging of meta-analysis, we demonstrated that genetically predicted Omega-3 FA levels, Ratio of Omega-3 FA to total FA, Docosahexaenoic acid (DHA) levels, and Ratio of DHA to total FA reduced the risk of IBD, UC, and CD. Meanwhile, multivariate MR suggested that the risk effects of Omega-3 FA and DHA for UC and CD were mainly affected by Saturated FA and Monounsaturated fatty acid (MUFA). Furthermore, although there was the causal association between Ratio of MUFA to total FA as well as Ratio of Polyunsaturated fatty acid (PUFA) to MUFA and CD, sensitivity analysis prompted that the findings were not robust. None of the above results had a reverse causal effect. Conclusion This MR investigation provided evidence of causality between diverse FA and IBD. These findings offered new insights into the treatment and prevention of IBD.
Collapse
Affiliation(s)
| | - Zhenhua Zhou
- Department of General Surgery, Medical Center of Digestive Disease, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| |
Collapse
|
2
|
Hata M, Andriessen EMMA, Hata M, Diaz-Marin R, Fournier F, Crespo-Garcia S, Blot G, Juneau R, Pilon F, Dejda A, Guber V, Heckel E, Daneault C, Calderon V, Des Rosiers C, Melichar HJ, Langmann T, Joyal JS, Wilson AM, Sapieha P. Past history of obesity triggers persistent epigenetic changes in innate immunity and exacerbates neuroinflammation. Science 2023; 379:45-62. [PMID: 36603072 DOI: 10.1126/science.abj8894] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Age-related macular degeneration is a prevalent neuroinflammatory condition and a major cause of blindness driven by genetic and environmental factors such as obesity. In diseases of aging, modifiable factors can be compounded over the life span. We report that diet-induced obesity earlier in life triggers persistent reprogramming of the innate immune system, lasting long after normalization of metabolic abnormalities. Stearic acid, acting through Toll-like receptor 4 (TLR4), is sufficient to remodel chromatin landscapes and selectively enhance accessibility at binding sites for activator protein-1 (AP-1). Myeloid cells show less oxidative phosphorylation and shift to glycolysis, ultimately leading to proinflammatory cytokine transcription, aggravation of pathological retinal angiogenesis, and neuronal degeneration associated with loss of visual function. Thus, a past history of obesity reprograms mononuclear phagocytes and predisposes to neuroinflammation.
Collapse
Affiliation(s)
- Masayuki Hata
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada.,Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Elisabeth M M A Andriessen
- Department of Biomedical Sciences, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Maki Hata
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Roberto Diaz-Marin
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Frédérik Fournier
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Sergio Crespo-Garcia
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada.,Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Guillaume Blot
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada.,Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Rachel Juneau
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Frédérique Pilon
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Agnieszka Dejda
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Vera Guber
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Emilie Heckel
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Centre Hospitalier Universitaire Ste-Justine Research Center, Montreal, Quebec H3T 1C5, Canada
| | - Caroline Daneault
- Department of Nutrition, University of Montreal, Montreal, Quebec, Plateforme métabolomique de l'Institut de Cardiologie de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Virginie Calderon
- Bioinformatics & Molecular Biology Core Facility, Institut de Recherches Cliniques de Montréal, Montreal, Quebec H2W 1R7, Canada
| | - Christine Des Rosiers
- Department of Nutrition, University of Montreal, Montreal, Quebec, Plateforme métabolomique de l'Institut de Cardiologie de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Heather J Melichar
- Department of Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Jean-Sebastien Joyal
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Centre Hospitalier Universitaire Ste-Justine Research Center, Montreal, Quebec H3T 1C5, Canada
| | - Ariel M Wilson
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada.,Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| |
Collapse
|
3
|
Huang H, Wang X, Yang L, He W, Meng T, Zheng K, Xia X, Zhou Y, He J, Liu C, Zou S, Xiao D. The Effects of Fenugreek Extract on Growth Performance, Serum Biochemical Indexes, Immunity and NF-κB Signaling Pathway in Broiler. Front Vet Sci 2022; 9:882754. [PMID: 35812848 PMCID: PMC9260050 DOI: 10.3389/fvets.2022.882754] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
In order to study the regulation of Fenugreek seed extract (FSE) on the immunity of broilers, and explore the appropriate amount of FSE in broilers' production, 1-day-old yellow feather broilers with a total of 420 birds were randomly allocated into seven treatments. Each treatment had six replicates, with 10 birds per replicate. The two control groups were the basic fodder group fed with basal diet and the bacitracin zinc group added 30 mg/kg bacitracin zinc to the basal diet. Experimental groups included five levels of FSE (50, 100, 200, 400, and 800 mg/kg FSE to the basal diet, respectively). The pre-test period was 7 days and the formal test lasted for 56 days. The results showed that the average daily gain (ADG) of 50 and 800 mg/kg FSE groups was significantly increased (P < 0.01), and the feed to gain ratio (F/G) of FSE groups was significantly decreased (P < 0.01) compared with the basic fodder and the bacitracin zinc groups. Compared with the basic fodder group, the serum total cholesterol (TC) content in the FSE groups was significantly decreased (P < 0.05), the serum low density lipoprotein cholesterol (LDL-C) content of 50, 100, and 800 mg/kg FSE groups was significantly lower than that of the basic fodder group (P < 0.05). Compared with the basic fodder and bacitracin zinc groups, the serum immunoglobulins (IgG, IgM, IgA) content of 100 and 200 mg/kg FSE groups were significantly increased (P < 0.05). Compared with the bacitracin zinc group, the serum interleukins (IL-1, IL-10) content of 400 mg/kg FSE group were significantly increased (P ≤ 0.05), and the serum interferon-γ (IFN-γ) content of 100 and 200 mg/kg FSE groups was significantly increased (P < 0.05). Compared with the basic fodder group, the lower doses (0–400 mg/kg) of FSE had no significant effect on the mRNA expression of toll-like receptors 4/ myeloid differentiation factor 88/ nuclear factor-κB (TLR4/MyD88/NF-κB) signaling pathways (P > 0.05). The 800 mg/kg FSE treatment group significantly increased the expression levels of nuclear factor-κB (NF-κB) mRNA in the spleen of broilers (P < 0.05). The zinc bacitracin group significantly increased the expression levels of myeloid differentiation factor 88 (MyD88) and nuclear factor-κB (NF-κB) mRNA (P ≤ 0.05). The results showed that FSE could promote the secretion of immunoglobulins, regulate the body's cytokines, and have a positive effect on immunity in broilers. Furthermore, the recommended supplement of FSE is 100 mg/kg in the broiler diet.
Collapse
Affiliation(s)
- Hui Huang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xia Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Yiyang Vocational and Technical College, Yiyang, China
| | - Ling Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Wenxiang He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Tiantian Meng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Ke Zheng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xin Xia
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yingjun Zhou
- Geneham Pharmaceutical Co., Ltd., Changsha, China
- College of Xiangya Pharmaceutical Sciences, Central South University, Changsha, China
| | - Jianhua He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Chunming Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Shengwen Zou
- Hunan Wenfeng Agricultural and Animal Husbandry Technology Co., Loudi, China
| | - Dingfu Xiao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- *Correspondence: Dingfu Xiao
| |
Collapse
|
4
|
Rudge JD. A New Hypothesis for Alzheimer's Disease: The Lipid Invasion Model. J Alzheimers Dis Rep 2022; 6:129-161. [PMID: 35530118 PMCID: PMC9028744 DOI: 10.3233/adr-210299] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/05/2022] [Indexed: 02/07/2023] Open
Abstract
This paper proposes a new hypothesis for Alzheimer's disease (AD)-the lipid invasion model. It argues that AD results from external influx of free fatty acids (FFAs) and lipid-rich lipoproteins into the brain, following disruption of the blood-brain barrier (BBB). The lipid invasion model explains how the influx of albumin-bound FFAs via a disrupted BBB induces bioenergetic changes and oxidative stress, stimulates microglia-driven neuroinflammation, and causes anterograde amnesia. It also explains how the influx of external lipoproteins, which are much larger and more lipid-rich, especially more cholesterol-rich, than those normally present in the brain, causes endosomal-lysosomal abnormalities and overproduction of the peptide amyloid-β (Aβ). This leads to the formation of amyloid plaques and neurofibrillary tangles, the most well-known hallmarks of AD. The lipid invasion model argues that a key role of the BBB is protecting the brain from external lipid access. It shows how the BBB can be damaged by excess Aβ, as well as by most other known risk factors for AD, including aging, apolipoprotein E4 (APOE4), and lifestyle factors such as hypertension, smoking, obesity, diabetes, chronic sleep deprivation, stress, and head injury. The lipid invasion model gives a new rationale for what we already know about AD, explaining its many associated risk factors and neuropathologies, including some that are less well-accounted for in other explanations of AD. It offers new insights and suggests new ways to prevent, detect, and treat this destructive disease and potentially other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jonathan D’Arcy Rudge
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| |
Collapse
|
5
|
Huang L, Gong L, Huo X, Lei L, Zhang Q, Hu Y, Kuang Q, Gui Y, Dai Y, Gu Y, Deng Y, Wang D, Guo D. N-acetyldopamine dimer inhibits neuroinflammation through the TLR4/NF-κB and NLRP3/Caspase-1 pathways. Acta Biochim Biophys Sin (Shanghai) 2022; 55:23-33. [PMID: 36017888 PMCID: PMC10157536 DOI: 10.3724/abbs.2022116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Neuroinflammation mediated by microglia is an important pathophysiological mechanism in neurodegenerative diseases. However, there is a lack of effective drugs to treat neuroinflammation. N-acetyldopamine dimer (NADD) is a natural compound from the traditional Chinese medicine Isaria cicada. In our previous study, we found that NADD can attenuate DSS-induced ulcerative colitis by suppressing the NF-κB and MAPK pathways. Does NADD inhibit neuroinflammation, and what is the target of NADD? To answer this question, lipopolysaccharide (LPS)-stimulated BV-2 microglia was used as a cell model to investigate the effect of NADD on neuroinflammation. Nitric oxide (NO) detection, reactive oxygen species (ROS) detection and enzyme-linked immunosorbent assay (ELISA) results show that NADD attenuates inflammatory signals and proinflammatory cytokines in LPS-stimulated BV-2 microglia, including NO, ROS, tumor necrosis factor (TNF)-α, interleukin (IL)-1β and interleukin-6 (IL-6). Western blot analysis show that NADD inhibits the protein levels of Toll-like receptor 4 (TLR4), nuclear factor kappa-B (NF-κB), NOD-like receptor thermal protein domain associated protein 3 (NLRP3), ASC and cysteinyl aspartate specific proteinase (Caspase)-1, indicating that NADD may inhibit neuroinflammation through the TLR4/NF-κB and NLRP3/Caspase-1 signaling pathways. In addition, surface plasmon resonance assays and molecular docking demonstrate that NADD binds with TLR4 directly. Our study reveals a new role of NADD in inhibiting the TLR4/NF-κB and NLRP3/Caspase-1 pathways, and shows that TLR4-MD2 is the direct target of NADD, which may provide a potential therapeutic candidate for the treatment of neuroinflammation.
Collapse
Affiliation(s)
- Lijun Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Leiqiang Gong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xueyan Huo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lirong Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yunjie Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qixuan Kuang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu Gui
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yifei Dai
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yucheng Gu
- Syngenta Jealott's Hill International Research Centre, Berkshire RG426EY, UK
| | - Yun Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dong Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dale Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
6
|
Tsukayama I, Mega T, Hojo N, Toda K, Kawakami Y, Takahashi Y, Suzuki-Yamamoto T. Diosgenin suppresses COX-2 and mPGES-1 via GR and improves LPS-induced liver injury in mouse. Prostaglandins Other Lipid Mediat 2021; 156:106580. [PMID: 34252545 DOI: 10.1016/j.prostaglandins.2021.106580] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/28/2022]
Abstract
Using a wild yam (Dioscorea japonica), we previously found novel anti-inflammatory and anti-carcinogenic effects via the downregulation of cyclooxygenase (COX)-2 and microsomal prostaglandin E synthase (mPGES)-1. One of the substances in wild yam is a steroidal saponin, diosgenin. We demonstrated that diosgenin suppressed COX-2 in human non-small-cell lung carcinoma A549 cells via nuclear factor-kappa B (NF-κB) translocation and the effects were reversed by a glucocorticoid receptor antagonist, RU486. In lipopolysaccharide (LPS)-induced mouse liver injury, COX-2 and mPGES-1 were induced and localized in sinusoidal macrophages and endothelial cells; however, diosgenin administration significantly suppressed Ptgs2 and Ptges expression and decreased COX-2 and mPGES-1 immunopositive cells in the sinusoids. Multiple immunohistochemical analyses showed that diosgenin had an effect on COX-2 and mPGES-1, particularly in the macrophages. Thus, we showed that diosgenin downregulated COX-2 and mPGES-1 via the glucocorticoid receptor and suppressed COX-2 and mPGES-1 in the macrophages of LPS-induced acute mouse liver injury.
Collapse
Affiliation(s)
- Izumi Tsukayama
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Takuto Mega
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Nana Hojo
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Keisuke Toda
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Yuki Kawakami
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Yoshitaka Takahashi
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Toshiko Suzuki-Yamamoto
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan.
| |
Collapse
|
7
|
Gorabi AM, Kiaie N, Khosrojerdi A, Jamialahmadi T, Al-Rasadi K, Johnston TP, Sahebkar A. Implications for the role of lipopolysaccharide in the development of atherosclerosis. Trends Cardiovasc Med 2021; 32:525-533. [PMID: 34492295 DOI: 10.1016/j.tcm.2021.08.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 08/16/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
Mounting scientific evidence over decades has established that atherosclerosis is a chronic inflammatory disorder. Among the potentially critical sources of vascular inflammation during atherosclerosis are the components of pathogenic bacteria, especially lipopolysaccharide (LPS). Toll-like receptor (TLR)-4, expressed on different inflammatory cells involved with the recognition of bacterial LPS, has been recognized to have mutations that are prevalent in a number of ethnic groups. Such mutations have been associated with a decreased risk of atherosclerosis. In addition, epidemiological investigations have proposed that LPS confers a risk factor for the development of atherosclerosis. Gram-negative bacteria are the major source of LPS in an individual's serum, which may be generated during subclinical infections. The major cell receptors on inflammatory cells involved in the pathogenesis of atherosclerosis, like macrophages, monocytes, and dendritic cells (DCs), are CD14, MD-2, and LPS binding protein (LBP). These receptors have been blamed for the development of atherosclerosis through dysregulated activation following LPS recognition. Lipoproteins may also play a role in modulating the LPS-induced inflammatory events during atherosclerosis development. In this review article, we attempt to clarify the role of LPS in the initiation and progression of atherosclerotic lesion development.
Collapse
Affiliation(s)
- Armita Mahdavi Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasim Kiaie
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Arezou Khosrojerdi
- Department of Medical Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri, 64108, USA.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Wang B, Dong J, Xu J, Qiu Z, Yao F. Ginsenoside CK inhibits obese insulin resistance by activating PPARγ to interfere with macrophage activation. Microb Pathog 2021; 157:105002. [PMID: 34051328 DOI: 10.1016/j.micpath.2021.105002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/08/2021] [Accepted: 05/19/2021] [Indexed: 12/18/2022]
Abstract
Obesity is often accompanied by chronic low-grade inflammation, which aggravates the disorder of lipid metabolism and leads to insulin resistance (IR). Macrophage activation plays an important role in inflammation. Ginsenoside Compound K (CK) is an active metabolite of ginsenoside Rb1, which is adopting to an anti-inflammatory effective substance. In order to clarify the mechanism of ginsenoside CK on the regulation of macrophage activation in adipose tissue, the macrophage model was incubated with the supernatant of hypertrophic adipocytes, and the co-culture models of Raw264.7 and 3T3-L1 were established. The levels of related cytokines, macrophage polarization and protein expression in inflammatory signaling pathway were measured. The results showed that ginsenoside CK significantly inhibited the increase of MCP-1 and TNF-α induced by the supernatant of hypertrophic adipocytes, promoted the expression of IL-10, inhibited the activation of inflammatory macrophages and increased the expression of anti-inflammatory macrophages. Similarly, ginsenoside CK inhibited the migration of Raw264.7, blocked the activation of NF-κB, and up-regulated the expression of PPARγ. In addition, ginsenoside CK also promotes the expression of IRS-1 in insulin signal pathway. The experimental results proved that ginsenoside CK plays a crucial role in alleviating inflammation and insulin resistance in obesity, and inhibits macrophage activation through the key protein PPARγ.
Collapse
Affiliation(s)
- Bei Wang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchum, 130117. China
| | - Jinxiang Dong
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchum, 130117. China
| | - Jie Xu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchum, 130117. China
| | - Zhidong Qiu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchum, 130117. China.
| | - Fan Yao
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchum, 130117. China.
| |
Collapse
|
9
|
Chen G, Liu Y, Xu Y, Zhang M, Guo S, Zhang G. Isoimperatorin exerts anti-inflammatory activity by targeting the LPS-TLR4/MD-2-NF-κB pathway. EUR J INFLAMM 2021. [DOI: 10.1177/20587392211000573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Isoimperatorin (QHS) is a phytoconstituent found in the methanolic extracts obtained from the roots of Angelica dahurica, which contains anti-inflammatory, anti-bacterial, analgesic, anti-tumor, and vasodilatory activities. QHS possesses potent antagonistic activity against lipopolysaccharide (LPS)-induced inflammation; however, the mechanism of action remains unclear. In this study, we investigated the anti-inflammatory effect of QHS and explored the underlying mechanisms. The QHS was purchased from Jiangsu Yongjian Pharmaceutical Co., Ltd. (Jiangsu, China). We performed MTT assay, real-time PCR, ELISA, and western blotting experiments to assess the anti-inflammatory activity and the possible mechanism of QHS in vitro. Molecular docking was performed to study the binding of QHS and myeloid differentiation protein-2 (MD-2) and elucidate the possible anti-inflammatory mechanism. QHS had no significant effect on cell viability. Moreover, pre-treatment with QHS significantly decreased the release of inflammatory cytokines and mediators including NO, TNF-α, IL-6, and IL-1β. In addition, real-time PCR showed that QHS decreased the mRNA expressions of iNOS, COX-2 TNF-α, IL-6, and IL-1β. Western blotting indicated that QHS could inhibit the expression of the proteins associated with the LPS-TLR4/MD-2-NF-κB signaling pathway. Lastly, molecular docking revealed a possible binding mechanism between QHS and MD-2. QHS exhibited anti-inflammatory activity when combined with MD-2, regulating the LPS-TLR4/MD-2-NF-κB signaling pathway, and inhibiting the release and expression of inflammatory cytokines and mediators. Furthermore, QHS can be used as a potential TLR4 antagonist, which blocks MD-2 binding, for treating inflammatory responses induced by LPS.
Collapse
Affiliation(s)
- Guirong Chen
- 967th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Dalian, Liaoning, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yunong Liu
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yubin Xu
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Mingbo Zhang
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Song Guo
- Department of Computer Application, Shenyang Sport University, Shenyang, Liaoning, China
| | - Gang Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Polymyxin B prevents the development of adjuvant arthritis via modulation of TLR/Cox-2 signaling pathway. Life Sci 2020; 259:118250. [PMID: 32791152 DOI: 10.1016/j.lfs.2020.118250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 11/20/2022]
Abstract
AIMS Several microbial toll-like receptor (TLR) ligands, bacterial DNA and bacterial cell wall fragments have been identified in the synovium of rheumatoid arthritis (RA) patients, proving bacterial involvement in the pathogenesis of RA. The current study aimed to verify that low dose polymyxin B could prevent the development of chronic inflammatory arthritis. METHODS Twelve days post adjuvant injection, Sprague-Dawley rats were treated twice weekly with methotrexate (0.5 mg/kg) or daily with polymyxin B (1 mg/kg) or with combination of both for 1 or 2 weeks. Arthritis progression was assessed by hind paw swelling, serum levels of tumor growth factor-1β (TGF-1β), tumor necrosis factor-alpha (TNF-α), high sensitivity C-reactive protein (HS-CRP) and nuclear factor kappa B (NF-κB) were measured using ELISA. Cyclooxygenase-1 (Cox-1) and Cox-2 activities, as well as mRNA expression of TLR-2 and TLR-4 were determined. Histopathological examination of the ankle joint was performed as well as immunohistochemistry for anti-TLR-4. Histopathological assessment of toxic effects on the kidney was performed. KEY FINDINGS Adjuvant arthritis led to a significant swelling of the hind paw and alteration in all serum parameters, TLR-2 and TLR-4 expression, as well as Cox-2 activity. These alterations were associated with histopathological changes of the joints. Polymyxin B reduced significantly all biomarkers of inflammation, showing better effect of the combination in most of the studied parameters, with minimal signs of nephrotoxicity. SIGNIFICANCE In conclusion, results showed that polymyxin B possesses significant anti-arthritic activity which may be attributed to inhibition of the TLR-4, NF-κB and Cox-2 signaling pathway.
Collapse
|
11
|
Zhang C, Jia Y, Liu B, Wang G, Zhang Y. TLR4 knockout upregulates the expression of Mfn2 and PGC-1α in a high-fat diet and ischemia-reperfusion mice model of liver injury. Life Sci 2020; 254:117762. [PMID: 32437795 DOI: 10.1016/j.lfs.2020.117762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/01/2020] [Accepted: 05/05/2020] [Indexed: 12/30/2022]
Abstract
AIMS Patients with nonalcoholic fatty liver disease (NAFLD) have less tolerance to ischemia-reperfusion injury (IRI) of the liver than those with the healthy liver; hence have a higher incidence of severe complications after surgery. This study aimed to investigate the dynamics of the liver and mitochondrial damage and the impact of TLR4 knockout (TLR4KO) on Mfn2 expression in the composite model of NAFLD and IRI. MAIN METHODS We performed high-fat diet (HFD) feeding and ischemia reperfusion (IR) on wild type (WT) and TLR4 knockout TLR4KO mice. KEY FINDINGS The degree of structural and functional injuries to the liver and mitochondria (NAFLD and IRI) is greater than that caused by a single factor (NAFLD or IRI) or a simple superposition of both. The IL-6 and TNF-α expressions were significantly suppressed (P < .05), while PGC-1α and Mfn2 expressions were up-regulated considerably (P < .05) after TLR4KO. Furthermore, mitochondrial fusion increased, while ATP consumption and ROS production decreased significantly after TLR4KO (P < .05). The degree of reduction of compound injury by TLR4KO is more significant than the reduction degree of single factor injury. Also, TNF-α and IL-6 levels can be used predictive markers for mitochondrial damage and liver tolerance to NAFLD and IRI. SIGNIFICANCE TLR4KO upregulates the expression of Mfn2 and PGC-1α in the composite model of NAFLD and IRI. This pathway may be related to IL-6 and TNF-α. This evidence provides theoretical and experimental basis for the subsequent Toll-like receptor 4 (TLR4) receptor targeted therapy.
Collapse
Affiliation(s)
- Chaoyang Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yinzhao Jia
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Guoliang Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Bao M, Zhang K, Wei Y, Hua W, Gao Y, Li X, Ye L. Therapeutic potentials and modulatory mechanisms of fatty acids in bone. Cell Prolif 2020; 53:e12735. [PMID: 31797479 PMCID: PMC7046483 DOI: 10.1111/cpr.12735] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 02/05/2023] Open
Abstract
Bone metabolism is a lifelong process that includes bone formation and resorption. Osteoblasts and osteoclasts are the predominant cell types associated with bone metabolism, which is facilitated by other cells such as bone marrow mesenchymal stem cells (BMMSCs), osteocytes and chondrocytes. As an important component in our daily diet, fatty acids are mainly categorized as long-chain fatty acids including polyunsaturated fatty acids (LCPUFAs), monounsaturated fatty acids (LCMUFAs), saturated fatty acids (LCSFAs), medium-/short-chain fatty acids (MCFAs/SCFAs) as well as their metabolites. Fatty acids are closely associated with bone metabolism and associated bone disorders. In this review, we summarized the important roles and potential therapeutic implications of fatty acids in multiple bone disorders, reviewed the diverse range of critical effects displayed by fatty acids on bone metabolism, and elucidated their modulatory roles and mechanisms on specific bone cell types. The evidence supporting close implications of fatty acids in bone metabolism and disorders suggests fatty acids as potential therapeutic and nutritional agents for the treatment and prevention of metabolic bone diseases.
Collapse
Affiliation(s)
- Minyue Bao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Kaiwen Zhang
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yangyini Wei
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Weihan Hua
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yanzi Gao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xin Li
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ling Ye
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesDepartment of Cariology and EndodonticsWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
13
|
Ceotto Freitas-Lima L, Merlo E, Campos Zicker M, Navia-Pelaez JM, de Oliveira M, Dos Santos Aggum Capettini L, Nogueira CR, Versiani Matos Ferreira A, Sousa Santos SH, Bernardes Graceli J. Tributyltin impacts in metabolic syndrome development through disruption of angiotensin II receptor signaling pathways in white adipose tissue from adult female rats. Toxicol Lett 2018; 299:21-31. [PMID: 30172001 DOI: 10.1016/j.toxlet.2018.08.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 08/22/2018] [Accepted: 08/28/2018] [Indexed: 01/09/2023]
Abstract
White adipose tissue (WAT) dysfunction and obesity are a consequence of a low-grade inflammation state. These WAT irregularities could result from abnormal metabolic renin-angiotensin system (RAS) control. Recently, tributyltin (TBT) has been found to play a critical role in these metabolic irregularities. However, TBT actions on the WAT-RAS functions are not currently well understood. In this study, we assessed whether TBT exposure resulted in metabolic syndrome (MetS) development and other metabolic complications as a result of abnormal modulation of WAT-RAS pathways. TBT (100 ng/kg/day) was administered to adult female Wistar rats, and their WAT morphophysiology and adipokine profiles were assessed. We further assessed the expression of Angiotensin-II receptor proteins (AT1R and AT2R) and proteins involved in downstream pathways mediating inflammation and adipogenesis modulation. TBT-exposed rats exhibited increases in body weight and adiposity. TBT rats present dyslipidemia and insulin resistance, suggesting MetS development. TBT promoted WAT inflammatory infiltration, AT1R protein overexpression and reduced Angiotensin-(1-7) expression. These TBT WAT abnormalities are reflected by NFκB activation, with higher adipokine levels (leptin, TNF-α and IL-6) and overexpression of AKT, ERK, P38, FAS and PPARγ protein. In vitro, TBT exposure stimulates lipid accumulation, reduces AT2R protein expression, and increases leptin, AKT and ERK protein expression in 3T3L1 cells. These findings suggest that TBT exposure participates in MetS development via the improper function of WAT-RAS metabolic control.
Collapse
Affiliation(s)
| | - Eduardo Merlo
- Department of Morphology, Healthy Sciences Center, Federal University of Espírito Santo, Brazil
| | - Marina Campos Zicker
- Department. of Food Science, Faculty of Pharmacy, Federal University of Minas Gerais, Brazil
| | | | - Miriane de Oliveira
- Department of Internal Medicine, Botucatu School of Medicine, University of São Paulo State, Botucatu, SP, Brazil
| | | | - Célia Regina Nogueira
- Department of Internal Medicine, Botucatu School of Medicine, University of São Paulo State, Botucatu, SP, Brazil
| | | | - Sérgio Henrique Sousa Santos
- Health Science Graduate Program, UNIMONTES, Montes Claros, MG, Brazil; Institute of Agricultural Sciences, Food Engineering College, Federal University of Minas Gerais, Montes Claros, MG, Brazil
| | - Jones Bernardes Graceli
- Department of Morphology, Healthy Sciences Center, Federal University of Espírito Santo, Brazil.
| |
Collapse
|
14
|
Wang J, Jin Y, Li J. Protective role of fentanyl in lipopolysaccharide-induced neuroinflammation in BV-2 cells. Exp Ther Med 2018; 16:3740-3744. [PMID: 30233733 DOI: 10.3892/etm.2018.6590] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/30/2018] [Indexed: 12/31/2022] Open
Abstract
Neurosurgery always results in neuroinflammation, which may activate microglial cells. Previous studies have demonstrated that fentanyl could be used for the induction or maintenance of anesthesia prior to surgery. However, it is unknown if fentanyl attenuates neuroinflammation prophylactically. Cell viability in groups that were treated with different concentrations of fentanyl (0.01, 0.1, 1 or 5 µmol/l) was analyzed by an MTT assay. BV-2 microglial cells were treated with lipopolysaccharide (LPS) at a concentration of 1 µg/ml to mimic neuroinflammation in vitro. BV-2 cells were pretreated with 5 µmol/l fentanyl prior to stimulation by LPS. The protein levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-10 in the culture medium were assessed by ELISA. The mRNA level of toll-like receptor (TLR)4 was evaluated by reverse transcription-quantitative polymerase chain reaction analysis. The protein levels of TLR4, glycogen synthase kinase (GSK)-3β and phosphorylated (p)-GSK-3β in BV-2 cells were assessed by western blot analysis. The MTT assay demonstrated that low concentrations of fentanyl (0.01, 0.1 or 1 µmol/l) did not affect the cell viability of BV-2 cells, while 5 µmol/l fentanyl significantly reduced BV-2 cell viability. The results of ELISA revealed that LPS significantly upregulated the release of TNF-α, IL-1β and IL-10, which were repressed by fentanyl pretreatment. Fentanyl pretreatment significantly reduced the LPS-induced elevation of TLR4 at mRNA and protein levels as well as p-GSK-3β protein levels in BV-2 cells. In conclusion, fentanyl pretreatment protects BV-2 cells from LPS-induced neuroinflammation by inhibiting TLR4 expression and GSK-3β activation. Neuroinflammation induced by surgery serves an important role in the development of postoperative cognitive dysfunction (POCD) and targeting the TLR4 and GSK-3β signaling pathway may provide a novel therapeutic approach for the treatment of POCD.
Collapse
Affiliation(s)
- Jian Wang
- Department of Anesthesiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yingjie Jin
- Department of Anesthesiology, Shengze Hospital of Jiangsu, Suzhou, Jiangsu 215228, P.R. China
| | - Jianchun Li
- Department of Anesthesiology, Shengze Hospital of Jiangsu, Suzhou, Jiangsu 215228, P.R. China
| |
Collapse
|
15
|
Abstract
The Western world has witnessed a tremendous increase in the occurrence of allergy and autoimmunity in the second half of the 20th century. Extensive efforts have been made to explain this phenomenon and various hypotheses have been formulated. Among them, two concepts have attracted the most attention: the "hygiene hypothesis," identifying the reduced exposure to environmental microorganisms as a driving force behind the observed epidemiological trends; and the "diet hypotheses," pointing to the importance of changes in our dietary habits. In this review, we discuss the interplay between the Western diet, microbiota, and inflammatory conditions, with particular emphasis on respiratory diseases. This is followed by an in-depth overview of the immunomodulatory potential of different dietary fatty acids. We conclude by identifying the outstanding questions, which, if answered, could shed further light on the impact of dietary habits on immunity and interconnect it with postulates proposed by the hygiene hypothesis. Linking these two concepts will be an important step towards understanding how Western lifestyle shapes disease susceptibility.
Collapse
|
16
|
Ann SJ, Kim KK, Cheon EJ, Noh HM, Hwang I, Yu JW, Park S, Kang SM, Manabe I, Miller YI, Kim S, Lee SH. Palmitate and minimally-modified low-density lipoprotein cooperatively promote inflammatory responses in macrophages. PLoS One 2018. [PMID: 29518116 PMCID: PMC5843266 DOI: 10.1371/journal.pone.0193649] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Increased consumption of Western-type diets and environmental insults lead to wide-spread increases in the plasma levels of saturated fatty acids and lipoprotein oxidation. The aim of this study is to examine whether palmitate and minimally modified low-density lipoprotein (mmLDL) exert an additive effect on macrophage activation. We found that CXCL2 and TNF-α secretion as well as ERK and p38 phosphorylation were additively increased by co-treatment of J774 macrophages with palmitate and mmLDL in the presence of lipopolysaccharide (LPS). Furthermore, the analysis of differentially expressed genes using the KEGG database revealed that several pathways, including cytokine-cytokine receptor interaction, and genes were significantly altered. These results were validated with real-time PCR, showing upregulation of Il-6, Csf3, Il-1β, and Clec4d. The present study demonstrated that palmitate and mmLDL additively potentiate the LPS-induced activation of macrophages. These results suggest the existence of synergistic mechanisms by which saturated fatty acids and oxidized lipoproteins activate immune cells.
Collapse
Affiliation(s)
- Soo-jin Ann
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ka-Kyung Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Jeong Cheon
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hye-Min Noh
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Inhwa Hwang
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea
| | - Je-Wook Yu
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea
| | - Sungha Park
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Seok-Min Kang
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ichiro Manabe
- Department of Disease Biology and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yury I. Miller
- Department of Medicine, University of California, San Diego, La Jolla, United States of America
| | - Sangwoo Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (SH Lee); (S Kim)
| | - Sang-Hak Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (SH Lee); (S Kim)
| |
Collapse
|
17
|
He W, Yuan T, Choezom D, Hunkler H, Annamalai K, Lupse B, Maedler K. Ageing potentiates diet-induced glucose intolerance, β-cell failure and tissue inflammation through TLR4. Sci Rep 2018; 8:2767. [PMID: 29426925 PMCID: PMC5807311 DOI: 10.1038/s41598-018-20909-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/25/2018] [Indexed: 12/25/2022] Open
Abstract
Ageing and obesity are two major risk factors for the development of type 2 diabetes (T2D). A chronic, low-grade, sterile inflammation contributes to insulin resistance and β-cell failure. Toll-like receptor-4 (TLR4) is a major pro-inflammatory pathway; its ligands as well as downstream signals are increased systemically in patients with T2D and at-risk individuals. In the present study we investigated the combined effects of high fat/high sucrose diet (HFD) feeding, ageing and TLR4-deficiency on tissue inflammation, insulin resistance and β-cell failure. In young mice, a short-term HFD resulted in a mildly impaired glucose tolerance and reduced insulin secretion, together with a β-cell mass compensation. In older mice, HFD further deteriorated insulin secretion and induced a significantly impaired glucose tolerance and augmented tissue inflammation in adipose, liver and pancreatic islets, all of which was attenuated by TLR4 deficiency. Our results show that ageing exacerbates HFD-induced impairment of glucose homeostasis and pancreatic β-cell function and survival, and deteriorates HFD-induced induction of mRNA expression of inflammatory cytokines and pro-inflammatory macrophage markers. TLR4-deficiency protects against these combined deleterious effects of a high fat diet and ageing through a reduced expression of inflammatory products in both insulin sensitive tissues and pancreatic islets.
Collapse
Affiliation(s)
- Wei He
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany.
| | - Ting Yuan
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Dolma Choezom
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Hannah Hunkler
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Karthika Annamalai
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Blaz Lupse
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Kathrin Maedler
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany.
| |
Collapse
|
18
|
Echeverría S, Leiguez E, Guijas C, do Nascimento NG, Acosta O, Teixeira C, Leiva LC, Rodríguez JP. Evaluation of pro-inflammatory events induced by Bothrops alternatus snake venom. Chem Biol Interact 2017; 281:24-31. [PMID: 29248447 DOI: 10.1016/j.cbi.2017.12.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/05/2017] [Accepted: 12/13/2017] [Indexed: 12/19/2022]
Abstract
Inflammation is a major local feature of envenomation by bothropic snakes being characterized by a prominent local edema, pain, and extensive swelling. There are reports demonstrating that whole Bothrops snake venoms and toxins isolated from them are able to activate macrophages functions, such as phagocytosis, production of reactive oxygen, cytokines and eicosanoids, however, little is known about the effects of Bothrops alternatus (B.a.) venom on macrophages. In this work, we evaluated the proinflammatory effects of B.a. venom with in vivo and in vitro experiments using the Raw 264.7 cell line and mouse peritoneal macrophages. We detected that B.a. venom augments cell permeability (2-fold), and cellular extravasation (mainly neutrophils), increase proinflammatory cytokines IL1 (∼300-fold), IL12 (∼200-fold), and TNFα (∼80-fold) liberation and induce the expression of enzymes related to lipid signaling, such as cPLA2α and COX-2. Additionally, using lipidomic techniques we detected that this venom produces a release of arachidonic acid (∼10 nMol/mg. Protein) and other fatty acids (16:0 and 18:1 n-9c). Although much of these findings were described in inflammatory processes induced by other bothropic venoms, here we demonstrate that B.a. venom also stimulates pro-inflammatory pathways involving lipid mediators of cell signaling. In this sense, lipidomics analysis of macrophages stimulated with B.a. venom evidenced that the main free fatty acids are implicated in the inflammatory response, and also demonstrated that this venom, is able to activate lipid metabolism even with a low content of PLA2.
Collapse
Affiliation(s)
- Silvina Echeverría
- Laboratorio de Investigación en Proteínas, Instituto de Química Básica y Aplicada del Nordeste Argentino (UNNE - CONICET), Argentina
| | - Elbio Leiguez
- Laboratorio de Farmacología, Instituto Butantan, Sao Paulo, Sao Paulo State, Brazil
| | - Carlos Guijas
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, 47003 Valladolid, Spain
| | | | - Ofelia Acosta
- Laboratorio de Farmacología, Facultad de Ciencias Veterinarias, Universidad Nacional del Nordeste (UNNE), Argentina
| | - Catarina Teixeira
- Laboratorio de Farmacología, Instituto Butantan, Sao Paulo, Sao Paulo State, Brazil
| | - Laura C Leiva
- Laboratorio de Investigación en Proteínas, Instituto de Química Básica y Aplicada del Nordeste Argentino (UNNE - CONICET), Argentina
| | - Juan Pablo Rodríguez
- Laboratorio de Investigación en Proteínas, Instituto de Química Básica y Aplicada del Nordeste Argentino (UNNE - CONICET), Argentina.
| |
Collapse
|
19
|
Toll-like receptor-4 signaling mediates inflammation and tissue injury in diabetic nephropathy. J Nephrol 2017; 30:719-727. [PMID: 28933050 DOI: 10.1007/s40620-017-0432-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/14/2017] [Indexed: 12/15/2022]
|
20
|
Francisqueti FV, Nascimento AF, Minatel IO, Dias MC, Luvizotto RDAM, Berchieri-Ronchi C, Ferreira ALA, Corrêa CR. Metabolic syndrome and inflammation in adipose tissue occur at different times in animals submitted to a high-sugar/fat diet. J Nutr Sci 2017; 6:e41. [PMID: 29152245 PMCID: PMC5672321 DOI: 10.1017/jns.2017.42] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 05/23/2017] [Accepted: 06/29/2017] [Indexed: 12/16/2022] Open
Abstract
Obesity is associated with low-grade inflammation, triggered in adipose tissue, which may occur due to an excess of SFA from the diet that can be recognised by Toll-like receptor-4. This condition is involved in the development of components of the metabolic syndrome associated with obesity, especially insulin resistance. The aim of the study was to evaluate the manifestation of the metabolic syndrome and adipose tissue inflammation as a function of the period of time in which rats were submitted to a high-sugar/fat diet (HSF). Male Wistar rats were divided into six groups to receive the control diet (C) or the HSF for 6, 12 or 24 weeks. HSF increased the adiposity index in all HSF groups compared with the C group. HSF was associated with higher plasma TAG, glucose, insulin and leptin levels. Homeostasis model assessment increased in HSF compared with C rats at 24 weeks. Both TNF-α and IL-6 were elevated in the epididymal adipose tissue of HSF rats at 24 weeks compared with HSF at 6 weeks and C at 24 weeks. Only the HSF group at 24 weeks showed increased expression of both Toll-like receptor-4 and NF-κB. More inflammatory cells were found in the HSF group at 24 weeks. We can conclude that the metabolic syndrome occurs independently of the inflammatory response in adipose tissue and that inflammation is associated with hypertrophy of adipocytes, which varies according to duration of exposure to the HSF.
Collapse
Affiliation(s)
| | | | - Igor Otávio Minatel
- São Paulo State University, Institute of Bioscience, Botucatu, São Paulo, Brazil
| | - Marcos Correa Dias
- Institute of Health Sciences, Federal University of Mato Grosso (UFMT), Sinop, Mato Grosso, Brazil
| | | | | | - Ana Lúcia A. Ferreira
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo, Brazil
| | - Camila Renata Corrêa
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo, Brazil
| |
Collapse
|
21
|
Le Roux G, Moche H, Nieto A, Benoit JP, Nesslany F, Lagarce F. Cytotoxicity and genotoxicity of lipid nanocapsules. Toxicol In Vitro 2017; 41:189-199. [PMID: 28323104 DOI: 10.1016/j.tiv.2017.03.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/01/2022]
Abstract
Lipid nanocapsules (LNCs) offer a promising method for the entrapment and nanovectorisation of lipophilic molecules. This new type of nanocarrier, formulated according to a solvent-free process and using only regulatory-approved components, exhibits many prerequisites for being well tolerated. Although toxicological reference values have already been obtained in mice, interaction of LNCs at the cell level needs to be elucidated. LNCs, measuring from 27.0±0.1nm (25nm LNCs) and 112.1±1.8nm (100nm LNCs) and with a zeta potential between -38.7±1.2mV and +9.18±0.4mV, were obtained by a phase inversion process followed by post-insertion of carboxy- or amino-DSPE-PEG. Trypan blue, MTS and neutral red uptake (NRU) assays were performed to evaluate the cytotoxicity of LNCs on mouse macrophage-like cells RAW264.7 after 24h of exposure. The determination of 50% lethal concentration (LC50) showed a size effect of LNCs on toxicity profiles: LC50 ranged from 1.036mg/L (MTS) and 0.477mg/mL (NRU) for 25nm LNCs, to 4.42mg/mL (MTS) and 2.18mg/mL (NRU) for 100nm LNCs. Surfactant Solutol® HS15 has been shown to be the only constituent to exhibit cytotoxicity; its LC50 reached 0.427mg/mL. Moreover, LNCs were not more toxic than their components in simple mixtures. At sublethal concentration, 100nm LNCs only were able to induce a significant production of nitric oxide (NO) by RAW264.7 cells, as assessed by the Griess reaction. Again, surfactant was the only component responsible for an increased NO release (1.8±0.2-fold). Genotoxicity assays revealed no DNA damage on human lymphocytes in both the in vitro Comet and micronucleus assays using 4-hour and 24-hour treatments, respectively.
Collapse
Affiliation(s)
- Gaël Le Roux
- L'UNAM Université, Inserm U1066 MINT, CHU d'Angers, 49933 Angers Cedex 9, France.
| | - Hélène Moche
- Laboratoire de Toxicologie, Institut Pasteur de Lille, EA 4483, 59019 Lille Cedex, France
| | - Alejandro Nieto
- L'UNAM Université, Inserm U1066 MINT, CHU d'Angers, 49933 Angers Cedex 9, France
| | - Jean-Pierre Benoit
- L'UNAM Université, Inserm U1066 MINT, CHU d'Angers, 49933 Angers Cedex 9, France
| | - Fabrice Nesslany
- Laboratoire de Toxicologie, Institut Pasteur de Lille, EA 4483, 59019 Lille Cedex, France
| | - Frédéric Lagarce
- L'UNAM Université, Inserm U1066 MINT, CHU d'Angers, 49933 Angers Cedex 9, France
| |
Collapse
|
22
|
Philippe C, Wauquier F, Landrier JF, Bonnet L, Miot-Noirault E, Rochefort GY, Sadoine J, Asrih M, Jornayvaz FR, Bernalier A, Coxam V, Wittrant Y. GPR40 mediates potential positive effects of a saturated fatty acid enriched diet on bone. Mol Nutr Food Res 2016; 61. [PMID: 27611773 DOI: 10.1002/mnfr.201600219] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 07/29/2016] [Accepted: 09/02/2016] [Indexed: 01/13/2023]
Abstract
SCOPE The stimulation of the free fatty acid receptor G-protein coupled receptor (GPR) 40 by GW9508 prevents bone loss by inhibiting osteoclast activity, both in vitro and in vivo. Here, we questioned whether the stimulation of the GPR40 receptor by dietary fatty acids may lead to the same beneficial effect on bone. METHODS AND RESULTS We investigated (i) the impact of a fatty acid enriched diet (high-fat diet [HFD]) on bone health in C57/BL6 female mice depending on (ii) the estrogen status (ovariectomy) and (iii) the genotype (GPR40+/+ or GPR40-/- ). Bone mineral density (BMD), body composition, weight, inflammation and bone remodeling parameters were monitored. HFD decreased BMD in HFD-SH-GPR40+/+ mice but OVX failed to further impact BMD in HFD-OVX-GPR40+/+ mice, while additional bone loss was observed in HFD-OVX-GPR40-/- animals. These data suggest that when stimulated by fatty acid enriched diets GPR40 contributes to counteract ovariectomy-induced bone alteration. The sparing effect is supported by the modulation of both the osteoprotegerin/receptor activator of nuclear factor kappa-B ligand (OPG/RANKL) ratio in blood stream and the expression level of inflammatory markers in adipose tissues. Bone preservation by GPR40 stimulation is dependent on the presence of long-chain saturated fatty acids. CONCLUSION GPR40 contributes to counter ovariectomy-induced bone loss in a context of saturated fatty acid enrichment.
Collapse
Affiliation(s)
- Claire Philippe
- INRA, UMR 1019, UNH, CRNH Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, Clermont-Ferrand, France.,Equipe Alimentation, Squelette et Métabolismes, Unité de Nutrition Humaine, Centre de Recherche INRA Auvergne Rhône Alpes, Site de Theix, 63122 Saint Genés Champanelle, France
| | - Fabien Wauquier
- INRA, UMR 1019, UNH, CRNH Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, Clermont-Ferrand, France.,Equipe Alimentation, Squelette et Métabolismes, Unité de Nutrition Humaine, Centre de Recherche INRA Auvergne Rhône Alpes, Site de Theix, 63122 Saint Genés Champanelle, France
| | - Jean-François Landrier
- INRA, UMR1260, Nutriments Lipidiques et Prévention des Maladies Métaboliques, Marseille, France.,Faculté de Médecine, Université de la Méditerranée Aix-Marseille 1 et 2, Marseille, France
| | - Lauriane Bonnet
- INRA, UMR1260, Nutriments Lipidiques et Prévention des Maladies Métaboliques, Marseille, France.,Faculté de Médecine, Université de la Méditerranée Aix-Marseille 1 et 2, Marseille, France
| | - Elisabeth Miot-Noirault
- Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, Clermont-Ferrand, France.,INSERM, UMR990, IMTV, Clermont-Ferrand, France
| | - Gaël Y Rochefort
- EA 2496 Pathologie, Imagerie et Biothérapies Orofaciales, UFR Odontologie, Université Paris Descartes and PIPA, PRES Sorbonne Paris Cité, Montrouge, France
| | - Jérémy Sadoine
- EA 2496 Pathologie, Imagerie et Biothérapies Orofaciales, UFR Odontologie, Université Paris Descartes and PIPA, PRES Sorbonne Paris Cité, Montrouge, France
| | - Mohamed Asrih
- Service d'Endocrinologie, Diabétologie et Métabolisme, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - François R Jornayvaz
- Service d'Endocrinologie, Diabétologie et Métabolisme, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | - Véronique Coxam
- INRA, UMR 1019, UNH, CRNH Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, Clermont-Ferrand, France.,Equipe Alimentation, Squelette et Métabolismes, Unité de Nutrition Humaine, Centre de Recherche INRA Auvergne Rhône Alpes, Site de Theix, 63122 Saint Genés Champanelle, France
| | - Yohann Wittrant
- INRA, UMR 1019, UNH, CRNH Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, Clermont-Ferrand, France.,Equipe Alimentation, Squelette et Métabolismes, Unité de Nutrition Humaine, Centre de Recherche INRA Auvergne Rhône Alpes, Site de Theix, 63122 Saint Genés Champanelle, France
| |
Collapse
|
23
|
Koska J, Ozias MK, Deer J, Kurtz J, Salbe AD, Harman SM, Reaven PD. A human model of dietary saturated fatty acid induced insulin resistance. Metabolism 2016; 65:1621-1628. [PMID: 27733250 DOI: 10.1016/j.metabol.2016.07.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/25/2016] [Accepted: 07/30/2016] [Indexed: 01/22/2023]
Abstract
BACKGROUND Increased consumption of high-fat diets is associated with the development of insulin resistance and type 2 diabetes. Current models to study the mechanisms of high-fat diet-induced IR in humans are limited by their long duration or low efficacy. In the present study we developed and characterized an acute dietary model of saturated fatty acid-enriched diet induced insulin resistance. METHODS High caloric diets enriched with saturated fatty acids (SFA) or carbohydrates (CARB) were evaluated in subjects with normal and impaired glucose tolerance (NGT or IGT). Both diets were compared to a standard eucaloric American Heart Association (AHA) control diet in a series of crossover studies. Whole body insulin resistance was estimated as steady state plasma glucose (SSPG) concentrations during the last 30min of a 3-h insulin suppression test. RESULTS SSPG was increased after a 24-h SFA diet (by 83±74% vs. control, n=38) in the entire cohort, which was comprised of participants with NGT (92±82%, n=22) or IGT (65±55%, n=16) (all p<0.001). SSPG was also increased after a single SFA breakfast (55±32%, p=0.008, n=7). The increase in SSPG was less pronounced after an overnight fast following a daylong SFA diet (24±31%, p=0.04, n=10), and further attenuated 24h after returning to the control diet (19±35%, p=0.09, n=11). SSPG was not increased after a 24-h CARB diet (26±50%, p=0.11, n=12). CONCLUSIONS A short-term SFA-enriched diet induced whole body insulin resistance in both NGT and IGT subjects. Insulin resistance persisted overnight after the last SFA meal and was attenuated by one day of a healthy diet. This model offers opportunities for identifying early mechanisms and potential treatments of dietary saturated fat induced insulin resistance.
Collapse
Affiliation(s)
| | | | - James Deer
- Phoenix VA Health Care System, Phoenix, AZ
| | | | | | - S Mitchell Harman
- Phoenix VA Health Care System, Phoenix, AZ; Kronos Longevity Research Institute, Phoenix, AZ
| | | |
Collapse
|
24
|
HDL inhibits saturated fatty acid mediated augmentation of innate immune responses in endothelial cells by a novel pathway. Atherosclerosis 2016; 259:83-96. [PMID: 28340361 DOI: 10.1016/j.atherosclerosis.2016.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 09/01/2016] [Accepted: 09/06/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Peripheral insulin resistance is associated with several metabolic abnormalities, including elevated serum fatty acids that contribute to vascular injury and atherogenesis. Our goals were to examine whether saturated fatty acids can modify innate immune responses to subclinical concentrations of lipopolysaccharide (LPS) in endothelial cells, and to explore the underlying pathway and determine whether it is modified by high density lipoprotein (HDL) and other factors commonly altered in insulin resistance. METHODS Physiologic concentrations of palmitic acid were added to human aortic endothelial cells with and without a variety of inhibitors or HDL and measures of cell inflammation and function assessed. RESULTS Palmitic acid significantly amplified human aortic endothelial cell inflammatory responses to LPS. Similar results were obtained from lipolysis products of triglyceride rich lipoproteins. Metabolism of palmitic acid to ceramide and subsequent activation of PKC-ζ, MAPK and ATF3 appeared critical in amplifying LPS induced inflammation. The amplified response to palmitic acid/LPS was decreased by HDL, dose dependently, and this inhibition was dependent on activation of PI3K/AKT and reduction in ATF3. CONCLUSIONS These results indicate that endothelial cell innate immune responses are modified by metabolic abnormalities commonly present in insulin resistance and provide evidence for a novel mechanism by which HDL may reduce vascular inflammation.
Collapse
|
25
|
Basson A, Trotter A, Rodriguez-Palacios A, Cominelli F. Mucosal Interactions between Genetics, Diet, and Microbiome in Inflammatory Bowel Disease. Front Immunol 2016; 7:290. [PMID: 27531998 PMCID: PMC4970383 DOI: 10.3389/fimmu.2016.00290] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/19/2016] [Indexed: 12/12/2022] Open
Abstract
Numerous reviews have discussed gut microbiota composition changes during inflammatory bowel diseases (IBD), particularly Crohn’s disease (CD). However, most studies address the observed effects by focusing on studying the univariate connection between disease and dietary-induced alterations to gut microbiota composition. The possibility that these effects may reflect a number of other interconnected (i.e., pantropic) mechanisms, activated in parallel, particularly concerning various bacterial metabolites, is in the process of being elucidated. Progress seems, however, hampered by various difficult-to-study factors interacting at the mucosal level. Here, we highlight some of such factors that merit consideration, namely: (1) the contribution of host genetics and diet in altering gut microbiome, and in turn, the crosstalk among secondary metabolic pathways; (2) the interdependence between the amount of dietary fat, the fatty acid composition, the effects of timing and route of administration on gut microbiota community, and the impact of microbiota-derived fatty acids; (3) the effect of diet on bile acid composition, and the modulator role of bile acids on the gut microbiota; (4) the impact of endogenous and exogenous intestinal micronutrients and metabolites; and (5) the need to consider food associated toxins and chemicals, which can introduce confounding immune modulating elements (e.g., antioxidant and phytochemicals in oils and proteins). These concepts, which are not mutually exclusive, are herein illustrated paying special emphasis on physiologically inter-related processes.
Collapse
Affiliation(s)
- Abigail Basson
- Digestive Health Research Institute, Case Western Reserve University , Cleveland, OH , USA
| | - Ashley Trotter
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA; University Hospitals Case Medical Center, Cleveland, OH, USA
| | | | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA; University Hospitals Case Medical Center, Cleveland, OH, USA
| |
Collapse
|
26
|
Peng M, Biswas D. Short chain and polyunsaturated fatty acids in host gut health and foodborne bacterial pathogen inhibition. Crit Rev Food Sci Nutr 2016; 57:3987-4002. [PMID: 27438132 DOI: 10.1080/10408398.2016.1203286] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Mengfei Peng
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA
- Biological Sciences Program Molecular and Cellular Biology Concentration, University of Maryland, College Park, Maryland, USA
| | - Debabrata Biswas
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA
- Biological Sciences Program Molecular and Cellular Biology Concentration, University of Maryland, College Park, Maryland, USA
- Center for Food Safety and Security Systems, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
27
|
Ono-Moore KD, Snodgrass RG, Huang S, Singh S, Freytag TL, Burnett DJ, Bonnel EL, Woodhouse LR, Zunino SJ, Peerson JM, Lee JY, Rutledge JC, Hwang DH. Postprandial Inflammatory Responses and Free Fatty Acids in Plasma of Adults Who Consumed a Moderately High-Fat Breakfast with and without Blueberry Powder in a Randomized Placebo-Controlled Trial. J Nutr 2016; 146:1411-9. [PMID: 27306892 PMCID: PMC4926849 DOI: 10.3945/jn.115.223909] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 05/06/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Saturated fatty acids (FAs) released from triglyceride-rich lipoproteins (TGRLs) activate Toll-like receptor 2 (TLR-2) and induce the expression of proinflammatory cytokines in monocytes. Certain plant polyphenols inhibit TLR-mediated signaling pathways. OBJECTIVE We determined whether plasma free FAs (FFAs) after a moderately high-fat (MHF, 40% kcal from fat) breakfast modulate the inflammatory status of postprandial blood, and whether blueberry intake suppresses FFA-induced inflammatory responses in healthy humans. METHODS Twenty-three volunteers with a mean ± SEM age and body mass index (in kg/m(2)) of 30 ± 3 y and 21.9 ± 0.4, respectively, consumed an MHF breakfast with either a placebo powder or 2 or 4 servings of blueberry powder in a randomized crossover design. The placebo powder was provided on the first test day and the blueberry powder doses were randomized with a 2-wk washout period. Plasma concentrations of lipids, glucose, and cytokines were determined. To determine whether FFAs derived from TGRL stimulate monocyte activation, and whether this is inhibited by blueberry intake, whole blood was treated with lipoprotein lipase (LPL). RESULTS The median concentrations of FFAs and cytokines [tumor necrosis factor-α, interleukin (IL)-6 and IL-8] in postprandial plasma (3.5 h) decreased compared with fasting plasma regardless of the blueberry intake (P < 0.001 for FFAs and P < 0.05 for cytokines). However, concentrations of FFAs and cytokines including IL-1β increased in LPL-treated whole blood compared with untreated blood samples from participants who consumed the placebo powder. Blueberry intake suppressed IL-1β and IL-6 production in LPL-treated postprandial blood compared with the placebo control when fasting changes were used as a covariate. CONCLUSIONS The plasma FFA concentration may be an important determinant affecting inflammatory cytokine production in blood. Supplementation with blueberry powder did not affect plasma FFA and cytokine concentrations; however, it attenuated the cytokine production induced by ex vivo treatment of whole blood with LPL. This trial was registered at clinicaltrials.gov as NCT01594008.
Collapse
Affiliation(s)
- Kikumi D Ono-Moore
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA;,Department of Nutrition and
| | - Ryan G Snodgrass
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA;,Department of Nutrition and
| | - Shurong Huang
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA
| | - Shamsher Singh
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA
| | - Tammy L Freytag
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA
| | - Dustin J Burnett
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA
| | | | - Leslie R Woodhouse
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA
| | - Susan J Zunino
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA;,Department of Nutrition and
| | - Janet M Peerson
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA;,Department of Nutrition and
| | - Joo Young Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, South Korea
| | - John C Rutledge
- Department of Internal Medicine, School of Medicine, University of California, Davis, CA; and
| | - Daniel H Hwang
- Western Human Nutrition Research Center, Agricultural Research Service/USDA, Davis, CA; Department of Nutrition and
| |
Collapse
|
28
|
Zhu R, Zheng J, Chen L, Gu B, Huang S. Astragaloside IV facilitates glucose transport in C2C12 myotubes through the IRS1/AKT pathway and suppresses the palmitate-induced activation of the IKK/IκBα pathway. Int J Mol Med 2016; 37:1697-1705. [PMID: 27082050 DOI: 10.3892/ijmm.2016.2555] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/29/2016] [Indexed: 11/05/2022] Open
Abstract
Astragaloside IV is a monomer isolated from Astragalus membranaceus (Fisch.) Bunge, which is one of the most widely used plant-derived drugs in traditional Chinese medicine for diabetes therapy. In the present study, we aimed to examine the effects of astragaloside IV on glucose in C2C12 myotubes and the underlying molecular mechanisms responsible for these effects. Four-day differentiated C2C12 myotubes were exposed to palmitate for 16 h in order to establish a model of insulin resistance and 3H glucose uptake, using 2-Deoxy‑D‑[1,2-3H(N)]-glucose (radiolabeled 2-DG), was detected. Astragaloside IV was added 2 h prior to palmitate exposure. The translocation of glucose transporter 4 (GLUT4) was evaluated by subcellular fractionation, and the expression of insulin signaling molecules such as insulin receptor β (IRβ), insulin receptor substrate (IRS)1/protein kinase B (AKT) and inhibitory κB kinase (IKK)/inhibitor-κBα (IκBα), which are associated with insulin signal transduction, were assessed in the basal or the insulin‑stimulated state using western blot analysis or RT-PCR. We also examined the mRNA expression of monocyte chemotactic protein 1 (MCP-1), interleukin 6 (IL-6), tumor necrosis factor α (TNFα) and Toll‑like receptor 4 (TLR4). Taken together, these findings demonstrated that astragaloside IV facilitates glucose transport in C2C12 myotubes through a mechanism involving the IRS1/AKT pathway, and suppresses the palmitate-induced activation of the IKK/IκBα pathway.
Collapse
Affiliation(s)
- Rongfeng Zhu
- Department of Internal Medicine, The 95th Hospital of Chinese People's Liberation Army, Putian, Fujian 351100, P.R. China
| | - Jianjun Zheng
- Department of Internal Medicine, The 95th Hospital of Chinese People's Liberation Army, Putian, Fujian 351100, P.R. China
| | - Lizhen Chen
- Department of Internal Medicine, The 95th Hospital of Chinese People's Liberation Army, Putian, Fujian 351100, P.R. China
| | - Bin Gu
- Department of Internal Medicine, The 95th Hospital of Chinese People's Liberation Army, Putian, Fujian 351100, P.R. China
| | - Shengli Huang
- Department of Internal Medicine, The 95th Hospital of Chinese People's Liberation Army, Putian, Fujian 351100, P.R. China
| |
Collapse
|
29
|
Mechanisms for the activation of Toll-like receptor 2/4 by saturated fatty acids and inhibition by docosahexaenoic acid. Eur J Pharmacol 2016; 785:24-35. [PMID: 27085899 DOI: 10.1016/j.ejphar.2016.04.024] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 04/09/2016] [Accepted: 04/12/2016] [Indexed: 02/06/2023]
Abstract
Saturated fatty acids can activate Toll-like receptor 2 (TLR2) and TLR4 but polyunsaturated fatty acids, particularly docosahexaenoic acid (DHA) inhibit the activation. Lipopolysaccharides (LPS) and lipopetides, ligands for TLR4 and TLR2, respectively, are acylated by saturated fatty acids. Removal of these fatty acids results in loss of their ligand activity suggesting that the saturated fatty acyl moieties are required for the receptor activation. X-ray crystallographic studies revealed that these saturated fatty acyl groups of the ligands directly occupy hydrophobic lipid binding domains of the receptors (or co-receptor) and induce the dimerization which is prerequisite for the receptor activation. Saturated fatty acids also induce the dimerization and translocation of TLR4 and TLR2 into lipid rafts in plasma membrane and this process is inhibited by DHA. Whether saturated fatty acids induce the dimerization of the receptors by interacting with these lipid binding domains is not known. Many experimental results suggest that saturated fatty acids promote the formation of lipid rafts and recruitment of TLRs into lipid rafts leading to ligand independent dimerization of the receptors. Such a mode of ligand independent receptor activation defies the conventional concept of ligand induced receptor activation; however, this may enable diverse non-microbial molecules with endogenous and dietary origins to modulate TLR-mediated immune responses. Emerging experimental evidence reveals that TLRs play a key role in bridging diet-induced endocrine and metabolic changes to immune responses.
Collapse
|
30
|
Blockade of Toll-Like Receptors (TLR2, TLR4) Attenuates Pain and Potentiates Buprenorphine Analgesia in a Rat Neuropathic Pain Model. Neural Plast 2015; 2016:5238730. [PMID: 26962463 PMCID: PMC4709736 DOI: 10.1155/2016/5238730] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 09/07/2015] [Accepted: 09/20/2015] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence indicates that microglial TLR2 and TLR4 play a significant role in nociception. Experiments were conducted to evaluate the contribution of TLR2 and TLR4 and their adaptor molecules to neuropathy and their ability to amplify opioid effectiveness. Behavioral tests (von Frey's and cold plate) and biochemical (Western blot and qRT-PCR) analysis of spinal cord and DRG tissue were conducted after chronic constriction injury (CCI) to the sciatic nerve. Repeated intrathecal administration of LPS-RS (TLR2 and TLR4 antagonist) and LPS-RS Ultrapure (TLR4 antagonist) attenuated allodynia and hyperalgesia. Biochemical analysis revealed time-dependent upregulation of mRNA and/or protein levels of TLR2 and TLR4 and MyD88 and TRIF adaptor molecules, which was paralleled by an increase in IBA-1/CD40-positive cells under neuropathy. LPS-RS and LPS-RS Ultrapure similarly influenced opioid analgesia by enhancing the effectiveness of buprenorphine but not morphine. Summing up, in light of their upregulation over the course of pain, both TLR2 and TLR4 may indeed play a significant role in neuropathy, which could be linked to the observed activation of IBA-1/CD40-positive cells. Blockade of TLR2 and TLR4 produced analgesia and enhanced buprenorphine's effectiveness, which suggests that they may be a putative target for future pharmacological pain relief tools, especially for opioid rotation, when the effect of morphine is tolerated.
Collapse
|
31
|
Philippe C, Wauquier F, Lyan B, Coxam V, Wittrant Y. GPR40, a free fatty acid receptor, differentially impacts osteoblast behavior depending on differentiation stage and environment. Mol Cell Biochem 2015; 412:197-208. [DOI: 10.1007/s11010-015-2626-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/08/2015] [Indexed: 11/25/2022]
|
32
|
Rocha DM, Caldas AP, Oliveira LL, Bressan J, Hermsdorff HH. Saturated fatty acids trigger TLR4-mediated inflammatory response. Atherosclerosis 2015; 244:211-5. [PMID: 26687466 DOI: 10.1016/j.atherosclerosis.2015.11.015] [Citation(s) in RCA: 343] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/16/2015] [Accepted: 11/16/2015] [Indexed: 12/11/2022]
Abstract
Toll-like receptors (TLR) mediate infection-induced inflammation and sterile inflammation by endogenous molecules. Among the TLR family, TLR4 is the best understood. However, while its downstream signaling pathways have been well defined, not all ligands of TLR4 are currently known. Current evidence suggests that saturated fatty acids (SFA) act as non-microbial TLR4 agonists, and trigger its inflammatory response. Thus, our present review provides a new perspective on the potential mechanism by which SFAs could modulate TLR4-induced inflammatory responses: (1) SFAs can be recognized by CD14-TLR4-MD2 complex and trigger inflammatory pathways, similar to lipopolysaccharide (LPS). (2) SFAs lead to modification of gut microbiota with an overproduction of LPS after a high-fat intake, enhancing this natural TLR4 ligand. (3) In addition, this metabolic endotoxemia leads to an oxidative stress thereby producing atherogenic lipids - oxLDL and oxidized phospholipids - which trigger CD36-TLR4-TLR6 inflammatory response. (4) Also, the high SFA consumption increases the lipemia and the mmLDL and oxLDL formation through oxidative modifications of LDL. The mmLDL, unlike oxLDL, is involved in activation of the CD14-TLR4-MD2 inflammatory pathway. Those molecules can induce TLR4 inflammatory response by MyD88-dependent and/or MyD88-independent pathways that, in turn, promotes the expression of proinflammatory transcript factors such as factor nuclear kappa B (NF-κB), which plays a crucial role in the induction of inflammatory mediators (cytokines, chemokines, or costimulatory molecules) implicated in the development and progression of many chronic diseases.
Collapse
Affiliation(s)
- D M Rocha
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil.
| | - A P Caldas
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil.
| | - L L Oliveira
- Department of General Biology, Universidade Federal de Viçosa, Viçosa, Brazil.
| | - J Bressan
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil.
| | - H H Hermsdorff
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil.
| |
Collapse
|
33
|
Hill AA, Reid Bolus W, Hasty AH. A decade of progress in adipose tissue macrophage biology. Immunol Rev 2015; 262:134-52. [PMID: 25319332 DOI: 10.1111/imr.12216] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
One decade has passed since seminal publications described macrophage infiltration into adipose tissue (AT) as a key contributor to inflammation and obesity-related insulin resistance. Currently, a PubMed search for 'adipose tissue inflammation' reveals over 3500 entries since these original reports. We now know that resident macrophages in lean AT are alternatively activated, M2-like, and play a role in AT homeostasis. In contrast, the macrophages in obese AT are dramatically increased in number and are predominantly classically activated, M1-like, and promote inflammation and insulin resistance. Mediators of AT macrophage (ATM) phenotype include adipokines and fatty acids secreted from adipocytes as well as cytokines secreted from other immune cells in AT. There are several mechanisms that could explain the large increase in ATMs in obesity. These include recruitment-dependent mechanisms such as adipocyte death, chemokine release, and lipolysis of fatty acids. Newer evidence also points to recruitment-independent mechanisms such as impaired apoptosis, increased proliferation, and decreased egress. Although less is known about the homeostatic function of M2-like resident ATMs, recent evidence suggests roles in AT expansion, thermoregulation, antigen presentation, and iron homeostasis. The field of immunometabolism has come a long way in the past decade, and many exciting new discoveries are bound to be made in the coming years that will expand our understanding of how AT stands at the junction of immune and metabolic co-regulation.
Collapse
Affiliation(s)
- Andrea A Hill
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | |
Collapse
|
34
|
Choi HW, Tian M, Song F, Venereau E, Preti A, Park SW, Hamilton K, Swapna GVT, Manohar M, Moreau M, Agresti A, Gorzanelli A, De Marchis F, Wang H, Antonyak M, Micikas RJ, Gentile DR, Cerione RA, Schroeder FC, Montelione GT, Bianchi ME, Klessig DF. Aspirin's Active Metabolite Salicylic Acid Targets High Mobility Group Box 1 to Modulate Inflammatory Responses. Mol Med 2015; 21:526-35. [PMID: 26101955 DOI: 10.2119/molmed.2015.00148] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 02/02/2023] Open
Abstract
Salicylic acid (SA) and its derivatives have been used for millennia to reduce pain, fever and inflammation. In addition, prophylactic use of acetylsalicylic acid, commonly known as aspirin, reduces the risk of heart attack, stroke and certain cancers. Because aspirin is rapidly de-acetylated by esterases in human plasma, much of aspirin's bioactivity can be attributed to its primary metabolite, SA. Here we demonstrate that human high mobility group box 1 (HMGB1) is a novel SA-binding protein. SA-binding sites on HMGB1 were identified in the HMG-box domains by nuclear magnetic resonance (NMR) spectroscopic studies and confirmed by mutational analysis. Extracellular HMGB1 is a damage-associated molecular pattern molecule (DAMP), with multiple redox states. SA suppresses both the chemoattractant activity of fully reduced HMGB1 and the increased expression of proinflammatory cytokine genes and cyclooxygenase 2 (COX-2) induced by disulfide HMGB1. Natural and synthetic SA derivatives with greater potency for inhibition of HMGB1 were identified, providing proof-of-concept that new molecules with high efficacy against sterile inflammation are attainable. An HMGB1 protein mutated in one of the SA-binding sites identified by NMR chemical shift perturbation studies retained chemoattractant activity, but lost binding of and inhibition by SA and its derivatives, thereby firmly establishing that SA binding to HMGB1 directly suppresses its proinflammatory activities. Identification of HMGB1 as a pharmacological target of SA/aspirin provides new insights into the mechanisms of action of one of the world's longest and most used natural and synthetic drugs. It may also provide an explanation for the protective effects of low-dose aspirin usage.
Collapse
Affiliation(s)
- Hyong Woo Choi
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| | - Miaoying Tian
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| | - Fei Song
- Center of Advanced Biotechnology and Medicine, Department of Molecular Biology and Biochemistry, and Northeast Structural Genomics Consortium, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Emilie Venereau
- Division of Genetics and Cell Biology, San Raffaele University and Research Institute, Milano, Italy
| | - Alessandro Preti
- Division of Genetics and Cell Biology, San Raffaele University and Research Institute, Milano, Italy
| | - Sang-Wook Park
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| | - Keith Hamilton
- Center of Advanced Biotechnology and Medicine, Department of Molecular Biology and Biochemistry, and Northeast Structural Genomics Consortium, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - G V T Swapna
- Center of Advanced Biotechnology and Medicine, Department of Molecular Biology and Biochemistry, and Northeast Structural Genomics Consortium, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Murli Manohar
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| | - Magali Moreau
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| | - Alessandra Agresti
- Division of Genetics and Cell Biology, San Raffaele University and Research Institute, Milano, Italy
| | - Andrea Gorzanelli
- Division of Genetics and Cell Biology, San Raffaele University and Research Institute, Milano, Italy
| | - Francesco De Marchis
- Division of Genetics and Cell Biology, San Raffaele University and Research Institute, Milano, Italy
| | - Huang Wang
- Center of Advanced Biotechnology and Medicine, Department of Molecular Biology and Biochemistry, and Northeast Structural Genomics Consortium, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Marc Antonyak
- Department of Chemistry and Chemical Biology, Department of Molecular Medicine, Cornell University, Ithaca, New York, United States of America
| | - Robert J Micikas
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| | - Daniel R Gentile
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| | - Richard A Cerione
- Department of Chemistry and Chemical Biology, Department of Molecular Medicine, Cornell University, Ithaca, New York, United States of America
| | - Frank C Schroeder
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| | - Gaetano T Montelione
- Center of Advanced Biotechnology and Medicine, Department of Molecular Biology and Biochemistry, and Northeast Structural Genomics Consortium, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America.,Department of Biochemistry and Molecular Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Marco E Bianchi
- Division of Genetics and Cell Biology, San Raffaele University and Research Institute, Milano, Italy
| | - Daniel F Klessig
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| |
Collapse
|
35
|
Ferguson PJ, Laxer RM. New discoveries in CRMO: IL-1β, the neutrophil, and the microbiome implicated in disease pathogenesis in Pstpip2-deficient mice. Semin Immunopathol 2015; 37:407-12. [PMID: 25894861 DOI: 10.1007/s00281-015-0488-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 03/31/2015] [Indexed: 01/22/2023]
Abstract
Chronic non-bacterial osteomyelitis (CNO), chronic recurrent multifocal osteomyelitis (CRMO) and synovitis, acne, pustulosis, hyperostosis and osteitis (SAPHO) syndrome are autoinflammatory disorder(s) in which sterile osteomyelitis is frequently accompanied by inflammatory conditions of the joints, skin, or intestine. Patients with CRMO commonly have a personal or family history of psoriasis, inflammatory bowel disease, and inflammatory arthritis, suggesting shared disease pathogenesis. Work by our group and others has demonstrated that dysregulation of interleukin-1 (IL-1) signaling can drive sterile osteomyelitis in the two human monogenic forms of the disease. Recent work in the chronic multifocal osteomyelitis (cmo) mouse model demonstrates that the disease is IL-1-mediated, that neutrophils are critical effector cells and that both caspase-1 and caspase-8 play redundant roles in mediating the cleavage of pro-IL-1β into its biologically active form. Recent data in the cmo mouse demonstrate that dietary manipulation alters the cmo microbiome and can prevent the development of osteomyelitis. Further investigation is needed to determine the specific components of the diet that result in protection from disease and if this finding can be translated into a treatment for human CRMO.
Collapse
Affiliation(s)
- Polly J Ferguson
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA,
| | | |
Collapse
|
36
|
Wauquier F, Léotoing L, Philippe C, Spilmont M, Coxam V, Wittrant Y. Pros and cons of fatty acids in bone biology. Prog Lipid Res 2015; 58:121-45. [PMID: 25835096 DOI: 10.1016/j.plipres.2015.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/06/2015] [Accepted: 03/23/2015] [Indexed: 12/12/2022]
Abstract
Despite the growing interest in deciphering the causes and consequences of obesity-related disorders, the mechanisms linking fat intake to bone behaviour remain unclear. Since bone fractures are widely associated with increased morbidity and mortality, most notably in elderly and obese people, bone health has become a major social and economic issue. Consistently, public health system guidelines have encouraged low-fat diets in order to reduce associated complications. However, from a bone point of view, mechanisms linking fat intake to bone alteration remain quite controversial. Thus, after more than a decade of dedicated studies, this timely review offers a comprehensive overview of the relationships between bone and fatty acids. Using clinical evidences as a starting-point to more complex molecular elucidation, this work highlights the complexity of the system and reveals that bone alteration that cannot be solved simply by taking ω-3 pills. Fatty acid effects on bone metabolism can be both direct and indirect and require integrated investigations. Furthermore, even at the level of a single cell, one fatty acid is able to trigger several different independent pathways (receptors, metabolites…) which may all have a say in the final cellular metabolic response.
Collapse
Affiliation(s)
- Fabien Wauquier
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Laurent Léotoing
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Claire Philippe
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Mélanie Spilmont
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Véronique Coxam
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Yohann Wittrant
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France.
| |
Collapse
|
37
|
Suppressor of cytokine signalling (SOCS) proteins as guardians of inflammatory responses critical for regulating insulin sensitivity. Biochem J 2014; 461:177-88. [PMID: 24966052 DOI: 10.1042/bj20140143] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Overactivation of immune pathways in obesity is an important cause of insulin resistance and thus new approaches aimed to limit inflammation or its consequences may be effective for treating Type 2 diabetes. The SOCS (suppressors of cytokine signalling) are a family of proteins that play an essential role in mediating inflammatory responses in both immune cells and metabolic organs such as the liver, adipose tissue and skeletal muscle. In the present review we discuss the role of SOCS1 and SOCS3 in controlling immune cells such as macrophages and T-cells and the impact this can have on systemic inflammation and insulin resistance. We also dissect the mechanisms by which SOCS (1-7) regulate insulin signalling in different tissues including their impact on the insulin receptor and insulin receptor substrates. Lastly, we discuss the important findings from SOCS whole-body and tissue-specific null mice, which implicate an important role for these proteins in controlling insulin action and glucose homoeostasis in obesity.
Collapse
|
38
|
Talbot NA, Wheeler-Jones CP, Cleasby ME. Palmitoleic acid prevents palmitic acid-induced macrophage activation and consequent p38 MAPK-mediated skeletal muscle insulin resistance. Mol Cell Endocrinol 2014; 393:129-42. [PMID: 24973767 PMCID: PMC4148479 DOI: 10.1016/j.mce.2014.06.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/11/2014] [Accepted: 06/13/2014] [Indexed: 12/21/2022]
Abstract
Obesity and saturated fatty acid (SFA) treatment are both associated with skeletal muscle insulin resistance (IR) and increased macrophage infiltration. However, the relative effects of SFA and unsaturated fatty acid (UFA)-activated macrophages on muscle are unknown. Here, macrophages were treated with palmitic acid, palmitoleic acid or both and the effects of the conditioned medium (CM) on C2C12 myotubes investigated. CM from palmitic acid-treated J774s (palm-mac-CM) impaired insulin signalling and insulin-stimulated glycogen synthesis, reduced Inhibitor κBα and increased phosphorylation of p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase in myotubes. p38 MAPK inhibition or siRNA partially ameliorated these defects, as did addition of tumour necrosis factor-α blocking antibody to the CM. Macrophages incubated with both FAs generated CM that did not induce IR, while palmitoleic acid-mac-CM alone was insulin sensitising. Thus UFAs may improve muscle insulin sensitivity and counteract SFA-mediated IR through an effect on macrophage activation.
Collapse
Affiliation(s)
- Nicola A Talbot
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK
| | - Caroline P Wheeler-Jones
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK
| | - Mark E Cleasby
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK.
| |
Collapse
|
39
|
Klawitter M, Hakozaki M, Kobayashi H, Krupkova O, Quero L, Ospelt C, Gay S, Hausmann O, Liebscher T, Meier U, Sekiguchi M, Konno SI, Boos N, Ferguson SJ, Wuertz K. Expression and regulation of toll-like receptors (TLRs) in human intervertebral disc cells. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2014; 23:1878-91. [PMID: 24997157 DOI: 10.1007/s00586-014-3442-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 06/10/2014] [Accepted: 06/25/2014] [Indexed: 12/15/2022]
Abstract
PURPOSE Although inflammatory processes play an essential role in painful intervertebral disc (IVD) degeneration, the underlying regulatory mechanisms are not well understood. This study was designed to investigate the expression, regulation and importance of specific toll-like receptors (TLRs)--which have been shown to play an essential role e.g. in osteoarthritis--during degenerative disc disease. METHODS The expression of TLRs in human IVDs was measured in isolated cells as well as in normal or degenerated IVD tissue. The role of IL-1β or TNF-α in regulating TLRs (expression/activation) as well as in regulating activity of down-stream pathways (NF-κB) and expression of inflammation-related genes (IL-6, IL-8, HSP60, HSP70, HMGB1) was analyzed. RESULTS Expression of TLR1/2/3/4/5/6/9/10 was detected in isolated human IVD cells, with TLR1/2/4/6 being dependent on the degree of IVD degeneration. Stimulation with IL-1β or TNF-α moderately increased TLR1/TLR4 mRNA expression (TNF-α only), and strongly increased TLR2 mRNA expression (IL-1β/TNF-α), with the latter being confirmed on the protein level. Stimulation with IL-1β, TNF-α or Pam3CSK4 (a TLR2-ligand) stimulated IL-6 and IL-8, which was inhibited by a TLR2 neutralizing antibody for Pam3CSK4; IL-1β and TNF-α caused NF-κB activation. HSP60, HSP70 and HMGB1 did not increase IL-6 or IL-8 and were not regulated by IL-1β/TNF-α. CONCLUSION We provide evidence that several TLRs are expressed in human IVD cells, with TLR2 possibly playing the most crucial role. As TLRs mediate catabolic and inflammatory processes, increased levels of TLRs may lead to aggravated disc degeneration, chronic inflammation and pain development. Especially with the identification of more endogenous TLR ligands, targeting these receptors may hold therapeutic promise.
Collapse
Affiliation(s)
- Marina Klawitter
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bhavaraju L, Shannahan J, William A, McCormick R, McGee J, Kodavanti U, Madden M. Diesel and biodiesel exhaust particle effects on rat alveolar macrophages with in vitro exposure. CHEMOSPHERE 2014; 104:126-33. [PMID: 24268344 PMCID: PMC3962714 DOI: 10.1016/j.chemosphere.2013.10.080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 10/23/2013] [Accepted: 10/30/2013] [Indexed: 05/23/2023]
Abstract
Combustion emissions from diesel engines emit particulate matter which deposits within the lungs. Alveolar macrophages (AMs) encounter the particles and attempt to engulf the particles. Emissions particles from diesel combustion engines have been found to contain diverse biologically active components including metals and polyaromatic hydrocarbons which cause adverse health effects. However little is known about AM response to particles from the incorporation of biodiesel. The objective of this study was to examine the toxicity in Wistar Kyoto rat AM of biodiesel blend (B20) and low sulfur petroleum diesel (PDEP) exhaust particles. Particles were independently suspended in media at a range of 1-500μgmL(-1). Results indicated B20 and PDEP initiated a dose dependent increase of inflammatory signals from AM after exposure. After 24h exposure to B20 and PDEP gene expression of cyclooxygenase-2 (COX-2) and macrophage inflammatory protein 2 (MIP-2) increased. B20 exposure resulted in elevated prostaglandin E2 (PGE2) release at lower particle concentrations compared to PDEP. B20 and PDEP demonstrated similar affinity for sequestration of PGE2 at high concentrations, suggesting detection is not impaired. Our data suggests PGE2 release from AM is dependent on the chemical composition of the particles. Particle analysis including measurements of metals and ions indicate B20 contains more of select metals than PDEP. Other particle components generally reduced by 20% with 20% incorporation of biodiesel into original diesel. This study shows AM exposure to B20 results in increased production of PGE2in vitro relative to diesel.
Collapse
Affiliation(s)
- Laya Bhavaraju
- Currciculum in Toxicology, University of North Carolina, Chapel Hill, NC, United States
| | | | - Aaron William
- National Renewable Energy Laboratory, Golden, CO, United States
| | | | - John McGee
- EPHD, NHEERL, US EPA, Research Triangle Park, NC, United States
| | | | - Michael Madden
- EPHD, NHEERL, US EPA, Research Triangle Park, NC, United States.
| |
Collapse
|
41
|
Li J, Chen L, Zhang Y, Zhang WJ, Xu W, Qin Y, Xu J, Zou D. TLR4 is required for the obesity-induced pancreatic beta cell dysfunction. Acta Biochim Biophys Sin (Shanghai) 2013; 45:1030-8. [PMID: 23985305 DOI: 10.1093/abbs/gmt092] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Obesity is an important inducing factor for type 2 diabetes. However, the mechanism underlying high-fat-(HF) diet-induced obesity in pancreatic beta cell dysfunction is still unclear. Toll-like receptor-4 (TLR4) is a key mediator of innate immunity. To investigate the effects of TLR4 in obesity-induced pancreatic beta cell dysfunction, we used male diabetic (db/db), obese (ob/ob) mice, TLR4-wild type (WT), and TLR4-knockout mice that were fed with normal diet or HF diet for 24 weeks. Immunostaining of TLR4 and TLR4 mRNA level in pancreatic islet were assessed. The results from biological characteristics, glucose tolerance test, insulin tolerance test, and insulin release test showed that the function of pancreatic islet was impaired in HF-fed TLR4 WT mice, but was protected in HF-fed TLR4 deficient (TLR4(-/-)) mice. By electron microscope detection, we observed that beta cell insulin secretory vesicles increased in HF-fed TLR4 WT mice. Ultrastructure of beta cell in HF-fed TLR4(-/-) mice was similar to that in normal chow diet-fed TLR4 WT mice. Then, glucose-stimulated insulin secretion assay by using primary pancreatic islet showed that the secretion function of pancreatic islet in HF-fed TLR4(-/-) mice was better than that in HF-fed TLR4 WT mice. Furthermore, in HF-fed TLR4(-/-) mice, the mRNA levels of IL-6, TNF-α, and MCP-1 genes in pancreatic islet were significantly lower than those in HF-fed TLR4 WT mice. Consistent with the change in gene expression, HF-fed TLR4 WT mice but not HF-fed TLR4(-/-) mice exhibited macrophage invasion in pancreatic island. Taken together, our data indicated that HF diet-induced obesity can stimulate the up-regulation of TLR4 locating on the surface of pancreatic beta cell, and subsequently lead to the recruitment of macrophage into pancreatic islet, which finally results in pancreatic beta cell dysfunction. This process is a possible mechanism involved in obesity-induced pancreatic beta cell dysfunction.
Collapse
Affiliation(s)
- Juan Li
- Department of Clinical Nutrition, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
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: 185] [Impact Index Per Article: 15.4] [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
|
43
|
Li X, Yu Y, Funk CD. Cyclooxygenase‐2 induction in macrophages is modulated by docosahexaenoic acid
via
interactions with free fatty acid receptor 4 (FFA4). FASEB J 2013; 27:4987-97. [DOI: 10.1096/fj.13-235333] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Xinzhi Li
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| | - Ying Yu
- Key Laboratory of Nutrition and MetabolismInstitute for Nutritional SciencesShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghaiChina
| | - Colin D. Funk
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| |
Collapse
|
44
|
Lee JH, Jung NH, Lee BH, Kim SH, Jun JH. Suppression of Heme Oxygenase-1 by Prostaglandin E2-Protein Kinase A-A-Kinase Anchoring Protein Signaling Is Central for Augmented Cyclooxygenase-2 Expression in Lipopolysaccharide-Stimulated RAW 264.7 Macrophages. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2013; 5:329-36. [PMID: 24003391 PMCID: PMC3756181 DOI: 10.4168/aair.2013.5.5.329] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 12/13/2012] [Accepted: 12/26/2012] [Indexed: 12/21/2022]
Abstract
PURPOSE Prostaglandin (PG) E2 is an immunomodulatory lipid mediator generated mainly via the cyclooxygenase-2 (COX-2) pathway from arachidonic acid at sites of infection and inflammation. A positive feedback loop of PGE2 on COX-2 expression is critical for homeostasis during toll-like receptor (TLR)-mediated inflammatory processes. The mechanism of PGE2-regulated COX-2 expression remains poorly understood. The low-molecular-weight stress protein heme oxygenase-1 (HO-1) contributes to the anti-inflammatory, anti-oxidant and anti-apoptotic response against environmental stress. METHODS We explored the involvement of HO-1 on PGE2 regulation of LPS-induced COX-2 expression in RAW 264.7 macrophages. RESULTS LPS-induced COX-2 expression in RAW 264.7 macrophages was enhanced by exogenous PGE2 or cyclic AMP (cAMP) analogue and was suppressed by a COX inhibitor (indomethacin), a protein kinase A (PKA) inhibitor (KT5720), and A kinase anchoring protein (AKAP) disruptors (Ht31 and RIAD). This result suggests that the stimulatory effects of endogenous and exogenous PGE2 on COX-2 expression are mediated by a cAMP-PKA-AKAP-dependent pathway. The induction of HO-1 was observed in LPS-stimulated RAW 264.7 macrophages. This induction was suppressed by exogenous PGE2 and enhanced by blockage of the endogenous PGE2 effect by the PKA inhibitor or AKAP disruptors. In addition, HO-1 induction by the HO activator copper protoporphyrin suppressed LPS-induced COX-2 expression, which was restored by the addition of exogenous PGE2. The induction of HO-1 inhibited LPS-induced NF-κB p-65 nuclear expression and translocation. CONCLUSIONS AKAP plays an important role in PGE2 regulation of COX-2 expression, and the suppression of HO-1 by PGE2-cAMP-PKA-AKAP signaling helps potentiate the LPS-induced COX-2 expression through a positive feedback loop in RAW 264.7 macrophages.
Collapse
Affiliation(s)
- Jae-Hyoung Lee
- Department of Internal Medicine, Eulji Hospital, Eulji University School of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
45
|
Zhu X, Chung S, Bi X, Chuang CC, Brown AL, Liu M, Seo J, Cuffe H, Gebre AK, Boudyguina E, Parks JS. Myeloid cell-specific ABCA1 deletion does not worsen insulin resistance in HF diet-induced or genetically obese mouse models. J Lipid Res 2013; 54:2708-17. [PMID: 23894207 DOI: 10.1194/jlr.m038943] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Obesity-associated low-grade chronic inflammation plays an important role in the development of insulin resistance. The membrane lipid transporter ATP-binding cassette transporter A1 (ABCA1) promotes formation of nascent HDL particles. ABCA1 also dampens macrophage inflammation by reducing cellular membrane cholesterol and lipid raft content. We tested the hypothesis that myeloid-specific ABCA1 deletion may exacerbate insulin resistance by increasing the obesity-associated chronic low-grade inflammation. Myeloid cell-specific ABCA1 knockout (MSKO) and wild-type (WT) mice developed obesity, insulin resistance, mild hypercholesterolemia, and hepatic steatosis to a similar extent with a 45% high-fat (HF) diet feeding or after crossing into the ob/ob background. Resident peritoneal macrophages and stromal vascular cells from obese MSKO mice accumulated significantly more cholesterol. Relative to chow, HF diet markedly induced macrophage infiltration and inflammatory cytokine expression to a similar extent in adipose tissue of WT and MSKO mice. Among pro-inflammatory cytokines examined, only IL-6 was highly upregulated in MSKO-ob/ob versus ob/ob mouse peritoneal macrophages, indicating a nonsignificant effect of myeloid ABCA1 deficiency on obesity-associated chronic inflammation. In conclusion, myeloid-specific ABCA1 deficiency does not exacerbate obesity-associated low-grade chronic inflammation and has minimal impact on the pathogenesis of insulin resistance in both HF diet-induced and genetically obese mouse models.
Collapse
Affiliation(s)
- Xuewei Zhu
- Department of Pathology/Lipid Sciences and Wake Forest School of Medicine, Winston-Salem, NC; and
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hashimoto Y, Yamada K, Tsushima H, Miyazawa D, Mori M, Nishio K, Ohkubo T, Hibino H, Ohara N, Okuyama H. Three dissimilar high fat diets differentially regulate lipid and glucose metabolism in obesity-resistant Slc:Wistar/ST rats. Lipids 2013; 48:803-15. [PMID: 23807365 DOI: 10.1007/s11745-013-3805-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 05/15/2013] [Indexed: 12/25/2022]
Abstract
Epidemiologic and ecologic studies suggest that dietary fat plays an important role in the development of obesity. Certain Wistar rat strains do not become obese when fed high-fat diets unlike others. In a preliminary study, we confirmed that Slc:Wistar/ST rats did not become obese when fed high-fat diets. The mechanisms governing the response of hepatic lipid-metabolizing enzymes to large quantities of dietary lipids consumed by obesity-resistant animals are unknown. The aim of the present study is to examine how obesity-resistant animals metabolize various types of high-fat diets and why they do not become obese. For this purpose, male Slc:Wistar/ST rats were fed a control low-fat diet (LS) or a high-fat diet containing fish oil (HF), soybean oil (HS), or lard (HL) for 4 weeks. We observed their phenotypes and determined lipid profiles in plasma and liver as well as mRNA expression levels in liver of genes related to lipid and glucose metabolism using DNA microarray and quantitative reverse transcriptase polymerase chain analyses. The body weights of all dietary groups were similar due to isocaloric intakes, whereas the weight of white adipose tissues in the LS group was significantly lower. The HF diet lowered plasma lipid levels by accelerated lipolysis in the peroxisomes and suppressed levels of very-low-density lipoprotein (VLDL) secretion. The HS diet promoted hepatic lipid accumulation by suppressed lipolysis in the peroxisomes and normal levels of VLDL secretion. The lipid profiles of rats fed the LS or HL diet were similar. The HL diet accelerated lipid and glucose metabolism.
Collapse
Affiliation(s)
- Yoko Hashimoto
- Department of Biochemistry, School of Dentistry, Aichi-Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya 464-8650, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Oral administration of alkylglycerols differentially modulates high-fat diet-induced obesity and insulin resistance in mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:834027. [PMID: 23864898 PMCID: PMC3705780 DOI: 10.1155/2013/834027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 06/05/2013] [Indexed: 12/31/2022]
Abstract
Alkylglycerols (AKGs) from shark liver oil (SLO) were demonstrated to have strong potency to stimulate immune response. However, no study has been conducted on the effects of AKGs on diet-induced obesity and metabolic inflammatory disorder. The purpose of the present study was to investigate the effect of two AKGs isoforms on obesity and insulin resistance in mice fed high-fat (HF) diet. Forty-eight C57BL/6 mice were divided into normal, HF, HF + 20 mg/kg selachyl alcohol (SA), HF + 200 mg/kg SA, HF + 20 mg/kg batyl alcohol (BA), and HF + 200 mg/kg BA groups. Body weight, fasting glucose, lipids, insulin and leptin levels, serum IL-1β, and TNF-α levels were compared among different groups. Our results showed that high-dose SA decreased body weight, serum triglyceride, cholesterol, fasting glucose level, insulin level, and serum leptin level of the HF fed mice, while high-dose BA increased fasting insulin level of the HF fed mice. Pretreatment of primary adipocytes with 10 μM SA or BA differentially modulates LPS-mediated MAPK and NF-κB signaling. Our study demonstrated that oral administration of AKGs has differential effects on HF-induced obesity and metabolic inflammatory disorder in mice.
Collapse
|
48
|
Harrison LM, Balan KV, Babu US. Dietary fatty acids and immune response to food-borne bacterial infections. Nutrients 2013; 5:1801-22. [PMID: 23698167 PMCID: PMC3708349 DOI: 10.3390/nu5051801] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 04/22/2013] [Accepted: 04/27/2013] [Indexed: 01/18/2023] Open
Abstract
Functional innate and acquired immune responses are required to protect the host from pathogenic bacterial infections. Modulation of host immune functions may have beneficial or deleterious effects on disease outcome. Different types of dietary fatty acids have been shown to have variable effects on bacterial clearance and disease outcome through suppression or activation of immune responses. Therefore, we have chosen to review research across experimental models and food sources on the effects of commonly consumed fatty acids on the most common food-borne pathogens, including Salmonella sp., Campylobacter sp., Shiga toxin-producing Escherichia coli, Shigella sp., Listeria monocytogenes, and Staphylococcus aureus. Altogether, the compilation of literature suggests that no single fatty acid is an answer for protection from all food-borne pathogens, and further research is necessary to determine the best approach to improve disease outcomes.
Collapse
Affiliation(s)
- Lisa M Harrison
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708, USA.
| | | | | |
Collapse
|
49
|
Absence of TLR4 reduces neurovascular unit and secondary inflammatory process after traumatic brain injury in mice. PLoS One 2013; 8:e57208. [PMID: 23555560 PMCID: PMC3610903 DOI: 10.1371/journal.pone.0057208] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 01/21/2013] [Indexed: 11/19/2022] Open
Abstract
Background Traumatic brain injury (TBI) initiates a neuroinflammatory cascade that contributes to neuronal damage and behavioral impairment. Toll-like receptors (TLRs) are signaling receptors in the innate immune system, although emerging evidence indicates their role in brain injury. We have therefore investigated the role played by TLR4 signaling pathway in the development of mechanisms of secondary inflammatory process in traumatic brain injury (TBI) differ in mice that lack a functional TLR4 signaling pathway. Methods/Principal Findings Controlled cortical impact injury was performed on TLR4 knockout (KO) mice (C57BL/10ScNJ) and wild-type (WT) mice (C57BL/10ScNJ). TBI outcome was evaluated by determination of infarct volume and assessment of neurological scores. Brains were collected at 24 h after TBI. When compared to WT mice, TLR4 KO mice had lower infarct volumes and better outcomes in neurological and behavioral tests (evaluated by EBST and rotarod test). Mice that lacked TLR4 had minor expression of TBI-induced GFAP, Chymase, Tryptase, IL-1β, iNOS, PARP and Nitrotyrosine mediators implicated in brain damage. The translocation of expression of p-JNK, IκB-α and NF-κB pathway were also lower in brains from TLR4 KO mice. When compared to WT mice, resulted in significant augmentation of all the above described parameters. In addition, apoptosis levels in TLR4 KO mice had minor expression of Bax while on the contrary with Bcl-2. Conclusions/Significance Our results clearly demonstrated that absence of TLR4 reduces the development of neuroinflammation, tissues injury events associated with brain trauma and may play a neuroprotective role in TBI in mice.
Collapse
|
50
|
Hattar K, Savai R, Subtil FSB, Wilhelm J, Schmall A, Lang DS, Goldmann T, Eul B, Dahlem G, Fink L, Schermuly RT, Banat GA, Sibelius U, Grimminger F, Vollmer E, Seeger W, Grandel U. Endotoxin induces proliferation of NSCLC in vitro and in vivo: role of COX-2 and EGFR activation. Cancer Immunol Immunother 2013; 62:309-20. [PMID: 22923191 PMCID: PMC3569588 DOI: 10.1007/s00262-012-1341-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 08/11/2012] [Indexed: 12/16/2022]
Abstract
Lung cancer is frequently complicated by pulmonary infections which may impair prognosis of this disease. Therefore, we investigated the effect of bacterial lipopolysaccharides (LPS) on tumor proliferation in vitro in the non-small cell lung cancer (NSCLC) cell line A549, ex vivo in a tissue culture model using human NSCLC specimens and in vivo in the A549 adenocarcinoma mouse model. LPS induced a time- and dose-dependent increase in proliferation of A549 cells as quantified by MTS activity and cell counting. In parallel, an increased expression of the proliferation marker Ki-67 and cyclooxygenase (COX)-2 was detected both in A549 cells and in ex vivo human NSCLC tissue. Large amounts of COX-2-derived prostaglandin (PG)E(2) were secreted from LPS-stimulated A549 cells. Pharmacological interventions revealed that the proliferative effect of LPS was dependent on CD14 and Toll-like receptor (TLR)4. Moreover, blocking of the epidermal growth factor receptor (EGFR) also decreased LPS-induced proliferation of A549 cells. Inhibition of COX-2 activity in A549 cells severely attenuated both PGE(2) release and proliferation in response to LPS. Synthesis of PGE(2) was also reduced by inhibiting CD14, TLR4 and EGFR in A549 cells. The proliferative effect of LPS on A549 cells could be reproduced in the A549 adenocarcinoma mouse model with enhancement of tumor growth and Ki-67 expression in implanted tumors. In summary, LPS induces proliferation of NSCLC cells in vitro, ex vivo in human NSCLC specimen and in vivo in a mouse model of NSCLC. Pulmonary infection may thus directly induce tumor progression in NSCLC.
Collapse
Affiliation(s)
- Katja Hattar
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Rajkumar Savai
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Jochen Wilhelm
- Department of Internal Medicine II, Biostatistics Group, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Anja Schmall
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Dagmar S. Lang
- Clinical and Experimental Pathology, Research Center Borstel, Borstel, Germany
| | - Torsten Goldmann
- Clinical and Experimental Pathology, Research Center Borstel, Borstel, Germany
| | - Bastian Eul
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Gabriele Dahlem
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Ludger Fink
- Department of Pathology, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Ralph-Theo Schermuly
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Gamal-Andre Banat
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Ulf Sibelius
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Ekkehard Vollmer
- Clinical and Experimental Pathology, Research Center Borstel, Borstel, Germany
| | - Werner Seeger
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ulrich Grandel
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| |
Collapse
|