1
|
Song J, Ok SM, Kwon EY, Kim HJ, Lee JY, Joo JY. Fatty Acid Binding Protein 4 Could Be a Linking Biomarker Between Periodontitis and Systemic Diseases. Biomedicines 2025; 13:402. [PMID: 40002815 PMCID: PMC11853709 DOI: 10.3390/biomedicines13020402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: This study aims to investigate the relationship between serum fatty acid-binding protein 4 (FABP4) levels and the severity of periodontitis in systemically healthy individuals. Additionally, the study examines whether non-surgical periodontal treatment can reduce FABP4 levels, establishing its potential as a biomarker linking periodontitis to systemic diseases. Methods: A total of 89 participants with stage I, II, or III periodontitis were recruited, excluding individuals with systemic diseases. Clinical parameters such as clinical attachment level (CAL), probing depth (PD), and gingival index (GI) were recorded. Serum FABP4 levels and Porphyromonas gingivalis (P. gingivalis) antibody titers were measured before and after periodontal treatment using ELISA kits. Statistical analysis included t-tests, correlation analysis, and multiple linear regression to assess changes in FABP4 levels and their association with clinical parameters. Results: FABP4 and P. gingivalis antibody titers significantly increased with the severity of periodontitis (p < 0.001). After non-surgical periodontal treatment, FABP4 levels significantly decreased across all stages of periodontitis. Moderate positive correlations were observed between FABP4 and CAL, PD, GI, and P. gingivalis antibody titers (p < 0.05). Multiple linear regression showed that FABP4 levels increased significantly with the progression of periodontitis, independent of age and sex. Conclusions: The study indicates that FABP4 is a potential biomarker for linking periodontitis to systemic conditions such as cardiovascular diseases and diabetes. Non-surgical periodontal treatment reduced FABP4 levels, potentially contributing to the improvement of systemic conditions associated with elevated FABP4. Further research should explore the role of FABP4 in patients with periodontitis and systemic diseases to strengthen its clinical relevance.
Collapse
Affiliation(s)
- Jiwon Song
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Yangsan 50612, Republic of Korea; (J.S.); (H.-J.K.); (J.-Y.L.)
| | - Soo-Min Ok
- Department of Oral Medicine, Dental Research Institute, Pusan National University Dental Hospital, Yangsan 50612, Republic of Korea;
| | - Eun-Young Kwon
- Department of Periodontology, Dental Clinic Center, Pusan National University Hospital, Busan 49241, Republic of Korea;
| | - Hyun-Joo Kim
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Yangsan 50612, Republic of Korea; (J.S.); (H.-J.K.); (J.-Y.L.)
- Department of Periodontology, Periodontal Disease Signaling Network Center, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ju-Youn Lee
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Yangsan 50612, Republic of Korea; (J.S.); (H.-J.K.); (J.-Y.L.)
| | - Ji-Young Joo
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Yangsan 50612, Republic of Korea; (J.S.); (H.-J.K.); (J.-Y.L.)
- Department of Periodontology, Periodontal Disease Signaling Network Center, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
2
|
Zhang Y, Hu B, Guan S, Li P, Guo Y, Xu P, Niu Y, Li Y, Feng Y, Du J, Xu J, Guan X, Gu J, Sun H, Huang M. Activation of pregnane X receptor sensitizes alcoholic steatohepatitis by transactivating fatty acid binding protein 4. Acta Pharm Sin B 2024; 14:4776-4788. [PMID: 39664417 PMCID: PMC11628830 DOI: 10.1016/j.apsb.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/17/2024] [Accepted: 07/25/2024] [Indexed: 12/13/2024] Open
Abstract
Alcoholic steatohepatitis (ASH) is a liver disease characterized by steatosis, inflammation, and necrosis of the liver tissue as a result of excessive alcohol consumption. Pregnane X receptor (PXR) is a xenobiotic nuclear receptor best known for its function in the transcriptional regulation of drug metabolism and disposition. Clinical reports suggested that the antibiotic rifampicin, a potent human PXR activator, is a contraindication in alcoholics, but the mechanism was unclear. In this study, we showed that the hepatic expression of fatty acid binding protein 4 (FABP4) was uniquely elevated in ASH patients and a mouse model of ASH. Pharmacological inhibiting FABP4 attenuated ASH in mice. Furthermore, treatment of mice with the mouse PXR agonist pregnenolon-16α-carbonitrile (PCN) induced the hepatic and circulating levels of FABP4 and exacerbated ASH in a PXR-dependent manner. Our mechanism study established FABP4 as a transcriptional target of PXR. Treatment with andrographolide, a natural compound and dual inhibitor of PXR and FABP4, alleviated mice from ASH. In summary, our results showed that the PXR-FABP4 gene regulatory axis plays an important role in the progression of ASH, which may have accounted for the contraindication of rifampicin in patients of alcoholic liver disease. Pharmacological inhibition of PXR and/or FABP4 may have its promise in the clinical management of ASH.
Collapse
Affiliation(s)
- Yiwen Zhang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Bingfang Hu
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shaoxing Guan
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Pan Li
- Department of Electrical Engineering and Computer Science, Case School of Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yingjie Guo
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130015, China
| | - Pengfei Xu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430072, China
| | - Yongdong Niu
- Department of Pharmacology, Shantou University Medical College, Shantou 515031, China
| | - Yujin Li
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Ye Feng
- Department of Endocrinology and Metabolic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiewen Du
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xiuchen Guan
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100069, China
| | - Jingkai Gu
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130015, China
| | - Haiyan Sun
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China
| | - Min Huang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
3
|
Correale J, Marrodan M. Multiple sclerosis and obesity: The role of adipokines. Front Immunol 2022; 13:1038393. [PMID: 36457996 PMCID: PMC9705772 DOI: 10.3389/fimmu.2022.1038393] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/28/2022] [Indexed: 11/24/2023] Open
Abstract
Multiple Sclerosis (MS), a chronic inflammatory disease of the central nervous system that leads to demyelination and neurodegeneration has been associated with various environmental and lifestyle factors. Population-based studies have provided evidence showing the prevalence of MS is increasing worldwide. Because a similar trend has been observed for obesity and metabolic syndrome, interest has grown in possible underlying biological mechanisms shared by both conditions. Adipokines, a family of soluble factors produced by adipose tissue that participate in a wide range of biological functions, contribute to a low state of chronic inflammation observed in obesity, and influence immune function, metabolism, and nutritional state. In this review, we aim to describe epidemiological and biological factors common to MS and obesity, as well as provide an update on current knowledge of how different pro- and anti-inflammatory adipokines participate as immune response mediators in MS, as well as in the animal model for MS, namely, experimental autoimmune encephalomyelitis (EAE). Multiple Sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) leading to demyelination, and neurodegeneration. Although its pathogenesis is not yet fully understood, there is considerable evidence to suggest MS arises from complex interactions between individual genetic susceptibility and external environmental factors. In recent decades, population-based studies have provided evidence indicating the prevalence of MS is increasing worldwide, in parallel with the rise in obesity and metabolic syndrome. This synchronous increment in the incidence of both MS and obesity has led to a search for potential biological mechanisms linking both conditions. Notably, a large number of studies have established significant correlation between obesity and higher prevalence, or worse prognosis, of several immune-mediated conditions. Fat tissue has been found to produce a variety of soluble factors named adipokines. These mediators, secreted by both adipocytes as well as diverse immune cells, participate in a wide range of biological functions, further strengthening the concept of a link between immune function, metabolism, and nutritional state. Because obesity causes overproduction of pro-inflammatory adipokines (namely leptin, resistin and visfatin) and reduction of anti-inflammatory adipokines (adiponectin and apelin), adipose tissue dysregulation would appear to contribute to a state of chronic, low-grade inflammation favoring the development of disease. In this review, we present a summary of current knowledge related to the pathological effects of different adipokines, prevalent in obese MS patients.
Collapse
Affiliation(s)
- Jorge Correale
- Departamento de Neurología, Fleni, Buenos Aires, Argentina
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Universidad de Buenos Aires/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | |
Collapse
|
4
|
Fu X, Almenglo C, Fernandez ÁL, Martínez-Cereijo JM, Iglesias-Alvarez D, Duran-Muñoz D, García-Caballero T, Gonzalez-Juanatey JR, Rodriguez-Mañero M, Eiras S. The Effect of Mineralocorticoid Receptor 3 Antagonists on Anti-Inflammatory and Anti-Fatty Acid Transport Profile in Patients with Heart Failure. Cells 2022; 11:1264. [PMID: 35455943 PMCID: PMC9027091 DOI: 10.3390/cells11081264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 02/01/2023] Open
Abstract
Epicardial fat thickness is associated with cardiovascular disease. Mineralocorticoid receptor antagonist (MRA), a pharmaceutical treatment for CVD, was found to have an effect on adipose tissue. Our aim was to analyse the main epicardial fat genesis and inflammation-involved cell markers and their regulation by risk factors and MRA. We included blood and epicardial or subcutaneous fat (EAT or SAT) from 71 patients undergoing heart surgery and blood from 66 patients with heart failure. Cell types (transcripts or proteins) were analysed by real-time polymerase chain reaction or immunohistochemistry. Plasma proteins were analysed by Luminex technology or enzyme-linked immunoassay. Our results showed an upregulation of fatty acid transporter levels after aldosterone-induced genesis. The MRA intake was the main factor associated with lower levels in epicardial fat. On the contrary, MRA upregulated the levels and its secretion of the anti-inflammatory marker intelectin 1 and reduced the proliferation of epicardial fibroblasts. Our results have shown the local MRA intake effect on fatty acid transporters and anti-inflammatory marker levels and the proliferation rate on epicardial fat fibroblasts. They suggest the role of MRA on epicardial fat genesis and remodelling in patients with cardiovascular disease. Translational perspective: the knowledge of epicardial fat genesis and its modulation by drugs might be useful for improving the treatments of cardiovascular disease.
Collapse
Affiliation(s)
- Xiaoran Fu
- Translational Cardiology Group, Health Research Institute, 15706 Santiago de Compostela, Spain;
| | - Cristina Almenglo
- Cardiology Group, Health Research Institute, University Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain; (C.A.); (D.I.-A.); (J.R.G.-J.)
| | - Ángel Luis Fernandez
- Heart Surgery Department, University Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain; (Á.L.F.); (J.M.M.-C.); (D.D.-M.)
- CIBERCV Madrid, Department of Morphological Sciences, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - José Manuel Martínez-Cereijo
- Heart Surgery Department, University Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain; (Á.L.F.); (J.M.M.-C.); (D.D.-M.)
| | - Diego Iglesias-Alvarez
- Cardiology Group, Health Research Institute, University Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain; (C.A.); (D.I.-A.); (J.R.G.-J.)
| | - Darío Duran-Muñoz
- Heart Surgery Department, University Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain; (Á.L.F.); (J.M.M.-C.); (D.D.-M.)
- CIBERCV Madrid, Department of Morphological Sciences, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Tomás García-Caballero
- Morphological Sciences Department, Medicine Faculty, University of Santiago de Compostela and Pathology Department, University Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Jose Ramón Gonzalez-Juanatey
- Cardiology Group, Health Research Institute, University Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain; (C.A.); (D.I.-A.); (J.R.G.-J.)
- CIBERCV Madrid, Department of Morphological Sciences, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Cardiology Department, University Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Moises Rodriguez-Mañero
- Translational Cardiology Group, Health Research Institute, 15706 Santiago de Compostela, Spain;
- CIBERCV Madrid, Department of Morphological Sciences, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Cardiology Department, University Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Sonia Eiras
- Translational Cardiology Group, Health Research Institute, 15706 Santiago de Compostela, Spain;
- CIBERCV Madrid, Department of Morphological Sciences, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| |
Collapse
|
5
|
Unveiling the Role of the Fatty Acid Binding Protein 4 in the Metabolic-Associated Fatty Liver Disease. Biomedicines 2022; 10:biomedicines10010197. [PMID: 35052876 PMCID: PMC8773613 DOI: 10.3390/biomedicines10010197] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 02/04/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD), the main cause of chronic liver disease worldwide, is a progressive disease ranging from fatty liver to steatohepatitis (metabolic-associated steatohepatitis; MASH). Nevertheless, it remains underdiagnosed due to the lack of effective non-invasive methods for its diagnosis and staging. Although MAFLD has been found in lean individuals, it is closely associated with obesity-related conditions. Adipose tissue is the main source of liver triglycerides and adipocytes act as endocrine organs releasing a large number of adipokines and pro-inflammatory mediators involved in MAFLD progression into bloodstream. Among the adipocyte-derived molecules, fatty acid binding protein 4 (FABP4) has been recently associated with fatty liver and additional features of advanced stages of MAFLD. Additionally, emerging data from preclinical studies propose FABP4 as a causal actor involved in the disease progression, rather than a mere biomarker for the disease. Therefore, the FABP4 regulation could be considered as a potential therapeutic strategy to MAFLD. Here, we review the current knowledge of FABP4 in MAFLD, as well as its potential role as a therapeutic target for this disease.
Collapse
|
6
|
Ren Y, Zhao H, Yin C, Lan X, Wu L, Du X, Griffiths HR, Gao D. Adipokines, Hepatokines and Myokines: Focus on Their Role and Molecular Mechanisms in Adipose Tissue Inflammation. Front Endocrinol (Lausanne) 2022; 13:873699. [PMID: 35909571 PMCID: PMC9329830 DOI: 10.3389/fendo.2022.873699] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/17/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic low-grade inflammation in adipose tissue (AT) is a hallmark of obesity and contributes to various metabolic disorders, such as type 2 diabetes and cardiovascular diseases. Inflammation in ATs is characterized by macrophage infiltration and the activation of inflammatory pathways mediated by NF-κB, JNK, and NLRP3 inflammasomes. Adipokines, hepatokines and myokines - proteins secreted from AT, the liver and skeletal muscle play regulatory roles in AT inflammation via endocrine, paracrine, and autocrine pathways. For example, obesity is associated with elevated levels of pro-inflammatory adipokines (e.g., leptin, resistin, chemerin, progranulin, RBP4, WISP1, FABP4, PAI-1, Follistatin-like1, MCP-1, SPARC, SPARCL1, and SAA) and reduced levels of anti-inflammatory adipokines such as adiponectin, omentin, ZAG, SFRP5, CTRP3, vaspin, and IL-10. Moreover, some hepatokines (Fetuin A, DPP4, FGF21, GDF15, and MANF) and myokines (irisin, IL-6, and DEL-1) also play pro- or anti-inflammatory roles in AT inflammation. This review aims to provide an updated understanding of these organokines and their role in AT inflammation and related metabolic abnormalities. It serves to highlight the molecular mechanisms underlying the effects of these organokines and their clinical significance. Insights into the roles and mechanisms of these organokines could provide novel and potential therapeutic targets for obesity-induced inflammation.
Collapse
Affiliation(s)
- Yakun Ren
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
| | - Hao Zhao
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xi Lan
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Litao Wu
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Xiaojuan Du
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Helen R. Griffiths
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Dan Gao
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
- *Correspondence: Dan Gao,
| |
Collapse
|
7
|
Fahmi RM, Kamel AE, Elsayed DA, Zidan AA, Sarhan NT. Serum levels of leptin and adiponectin in patients with multiple sclerosis. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2021. [DOI: 10.1186/s41983-021-00369-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The role of adipokines such as leptin and adiponectin in regulating the immunity has been documented, however data concerning their consequence on multiple sclerosis (MS) Egyptian patients are deficient. The aim of this study is to demonstrate the serum levels of leptin and adiponectin in MS patients and to assess their association with disease disability and severity. A case–control study including 60 subjects (30 MS patients and 30 age, sex and body mass index-matched healthy controls) was performed.
Results
Serum leptin level was significantly higher among MS patients than controls (P < 0.001) while adiponectin was not significantly elevated in MS patients (P = 0.24). There was a significant positive correlation between leptin levels with MS disability (Expanded Disability Status Scale) (r = 0.678; P < 0.001), severity (Multiple Sclerosis Severity Score) (r = 0.631; P < 0.001) and progression (progression index) (r = 0.461; P = 0.01). There was no statistically significant correlation between adiponectin with disease disability, severity or progression.
Conclusions
MS patients had significantly higher serum leptin levels and insignificant adiponectin levels compared to controls. Leptin has a potential role in multiple sclerosis disability and severity. However, adiponectin is not useful as a biomarker of MS disease, disability and severity.
Collapse
|
8
|
Tasinov O, Dincheva I, Badjakov I, Kiselova-Kaneva Y, Galunska B, Nogueiras R, Ivanova D. Phytochemical Composition, Anti-Inflammatory and ER Stress-Reducing Potential of Sambucus ebulus L. Fruit Extract. PLANTS (BASEL, SWITZERLAND) 2021; 10:plants10112446. [PMID: 34834808 PMCID: PMC8623228 DOI: 10.3390/plants10112446] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 05/09/2023]
Abstract
Sambucus ebulus L. (SE) fruits are used for their immunostimulation, hematopoietic and antiviral potential. Recently, we focused on analyzing the mechanism underlying SE fruit aqueous extract's (FAE) immunomodulation and anti-inflammatory activities, with attention to its endoplasmic reticulum (ER) stress-reducing potential. J774A.1 macrophages were treated with SE FAE alone or in conditions of lipopolysaccharides (LPS) stimulation. Using GC-MS and LC-MS/MS, its phytochemical composition was analyzed. To measure transcription and protein levels, we used qPCR and Western blot, respectively. The prevailing phytochemicals in SE FAE were hydroxycinnamic acids, proanthocyanidins and anthocyanins. The content of some amino acids, organic acids, alcohols, fatty acids and esters were newly reported. Extracts exerted an immunostimulation potential by stimulating IL-6, TNFα, Ccl2, COX2 and iNOS transcription, without inducing ER stress. SE FAE suppressed the LPS-induced transcription of inflammation related genes (IL-1β, IL-6, TNFα, Ccl2, Icam-1, Fabp4, COX2, iNOS, Noxo1, IL-1ra, Sirt-1) and reduced the protein levels of iNOS, peIF2α, ATF6α and CHOP. The effects were comparable to that of salicylic acid. SE suppresses LPS-stimulated inflammatory markers on the transcription and translation levels. Targeting ER stress is possibly another mechanism underlying its anti-inflammatory potential. These findings reveal the potential of SE fruits as a beneficial therapeutic of inflammation and ER stress-related pathological conditions.
Collapse
Affiliation(s)
- Oskan Tasinov
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical University of Varna, 84B Tzar Osvoboditel Blvd., 9002 Varna, Bulgaria; (Y.K.-K.); (B.G.); (D.I.)
- Correspondence: ; Tel.: +359-896-036961
| | - Ivayla Dincheva
- AgroBioInstitute, Agricultural Academy, 8 Dr. Tsankov Blvd., 1164 Sofia, Bulgaria; (I.D.); (I.B.)
| | - Ilian Badjakov
- AgroBioInstitute, Agricultural Academy, 8 Dr. Tsankov Blvd., 1164 Sofia, Bulgaria; (I.D.); (I.B.)
| | - Yoana Kiselova-Kaneva
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical University of Varna, 84B Tzar Osvoboditel Blvd., 9002 Varna, Bulgaria; (Y.K.-K.); (B.G.); (D.I.)
| | - Bistra Galunska
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical University of Varna, 84B Tzar Osvoboditel Blvd., 9002 Varna, Bulgaria; (Y.K.-K.); (B.G.); (D.I.)
| | - Ruben Nogueiras
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Diana Ivanova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical University of Varna, 84B Tzar Osvoboditel Blvd., 9002 Varna, Bulgaria; (Y.K.-K.); (B.G.); (D.I.)
| |
Collapse
|
9
|
Soubiya, Madaiah H, Tarannum F, Faizuddin M. Association of adipocyte fatty acid-binding protein and tumor necrosis factor alpha with periodontal health and disease: A cross-sectional investigation. Dent Res J (Isfahan) 2021; 18:79. [PMID: 34760070 PMCID: PMC8543096 DOI: 10.4103/1735-3327.326652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 01/30/2021] [Accepted: 03/17/2021] [Indexed: 11/04/2022] Open
Abstract
Background Adipocyte fatty acid binding protein (A-FABP) is a novel biomarker of inflammation for various chronic systemic diseases. Since periodontitis is a chronic inflammatory disease, this study explores the association of A-FABP with periodontal disease parameters and tumor necrosis factor-alpha (TNF-α) levels in gingival crevicular fluid in periodontal health and disease. Materials and Methods This original research article describes a cross-sectional study conducted at the Department of Periodontics, M. R. Ambedkar Dental College and Hospital, Bangalore, India. This cross-sectional investigation was conducted on sixty subjects which were divided into three groups of twenty subjects each - healthy, gingivitis, and chronic periodontitis. Clinical parameters - plaque index, bleeding index, probing depth, and clinical attachment loss were recorded. Gingival crevicular fluid samples were analyzed for A-FABP and TNF-α levels using ELISA. One-way analysis of variance was used to find the significance of study parameters on a continuous scale between three groups. Pearson's correlation has been used to find the relationship between Gingival crevicular fluid concentration of markers and periodontal parameters. Multiple linear regression analysis was applied to the study. The statistical significance was considered at P < 0.05. Results Mean concentration of A-FABP (6.43 ± 2.51) and TNF-α (3454.82 ± 1566.44) was highest in the periodontitis group, and the difference among the groups was statistically significant (P < 0.05). A positive correlation was observed between clinical attachment loss and the two markers among all groups. The correlation between A-FABP and TNF-α in periodontitis groups was positive and statistically significant (P < 0.05). Multiple linear regression model was statistically significant (P < 0.05) indicating that there is a significant relationship between the set of predictors and the clinical attachment loss. Conclusion A-FABP and TNF-α levels in GCF were significantly elevated in the presence of inflammation. A-FABP has a probable stimulatory effect on TNF-α; however, its role needs to be explored. A-FABP could serve as a novel inflammatory biomarker of periodontitis and the scope of using A-FABP inhibition as a treatment modality could be investigated with interventional studies.
Collapse
Affiliation(s)
- Soubiya
- Department of Periodontics, M. R. Ambedkar Dental College and Hospital, Bengaluru, Karnataka, India
| | - Hemalata Madaiah
- Department of Periodontics, M. R. Ambedkar Dental College and Hospital, Bengaluru, Karnataka, India
| | - Fouzia Tarannum
- Department of Periodontics, M. R. Ambedkar Dental College and Hospital, Bengaluru, Karnataka, India
| | - Mohamed Faizuddin
- Department of Periodontics, M. R. Ambedkar Dental College and Hospital, Bengaluru, Karnataka, India
| |
Collapse
|
10
|
A-FABP in Metabolic Diseases and the Therapeutic Implications: An Update. Int J Mol Sci 2021; 22:ijms22179386. [PMID: 34502295 PMCID: PMC8456319 DOI: 10.3390/ijms22179386] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022] Open
Abstract
Adipocyte fatty acid-binding protein (A-FABP), which is also known as ap2 or FABP4, is a fatty acid chaperone that has been further defined as a fat-derived hormone. It regulates lipid homeostasis and is a key mediator of inflammation. Circulating levels of A-FABP are closely associated with metabolic syndrome and cardiometabolic diseases with imminent diagnostic and prognostic significance. Numerous animal studies have elucidated the potential underlying mechanisms involving A-FABP in these diseases. Recent studies demonstrated its physiological role in the regulation of adaptive thermogenesis and its pathological roles in ischemic stroke and liver fibrosis. Due to its implication in various diseases, A-FABP has become a promising target for the development of small molecule inhibitors and neutralizing antibodies for disease treatment. This review summarizes the clinical and animal findings of A-FABP in the pathogenesis of cardio-metabolic diseases in recent years. The underlying mechanism and its therapeutic implications are also highlighted.
Collapse
|
11
|
Petkevicius K, Bidault G, Virtue S, Jenkins B, van Dierendonck XAMH, Dugourd A, Saez-Rodriguez J, Stienstra R, Koulman A, Vidal-Puig A. Norepinephrine promotes triglyceride storage in macrophages via beta2-adrenergic receptor activation. FASEB J 2021; 35:e21266. [PMID: 33484195 PMCID: PMC7898725 DOI: 10.1096/fj.202001101r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 11/11/2020] [Accepted: 11/30/2020] [Indexed: 01/02/2023]
Abstract
Tissue‐resident macrophages are required for homeostasis, but also contribute to tissue dysfunction in pathophysiological states. The sympathetic neurotransmitter norepinephrine (NE) induces an anti‐inflammatory and tissue‐reparative phenotype in macrophages. As NE has a well‐established role in promoting triglyceride lipolysis in adipocytes, and macrophages accumulate triglyceride droplets in various physiological and disease states, we investigated the effect of NE on primary mouse bone marrow‐derived macrophage triglyceride metabolism. Surprisingly, our data show that in contrast to the canonical role of NE in stimulating lipolysis, NE acting via beta2‐adrenergic receptors (B2ARs) in macrophages promotes extracellular fatty acid uptake and their storage as triglycerides and reduces free fatty acid release from triglyceride‐laden macrophages. We demonstrate that these responses are mediated by a B2AR activation‐dependent increase in Hilpda and Dgat1 gene expression and activity. We further show that B2AR activation favors the storage of extracellular polyunsaturated fatty acids. Finally, we present evidence that macrophages isolated from hearts after myocardial injury, for which survival critically depends on leukocyte B2ARs, have a transcriptional signature indicative of a transient triglyceride accumulation. Overall, we describe a novel and unexpected role of NE in promoting triglyceride storage in macrophages that could have potential implications in multiple diseases.
Collapse
Affiliation(s)
- Kasparas Petkevicius
- Institute of Metabolic Science, MDU MRC, University of Cambridge Metabolic Research Laboratories, Cambridge, United Kingdom
| | - Guillaume Bidault
- Institute of Metabolic Science, MDU MRC, University of Cambridge Metabolic Research Laboratories, Cambridge, United Kingdom
| | - Sam Virtue
- Institute of Metabolic Science, MDU MRC, University of Cambridge Metabolic Research Laboratories, Cambridge, United Kingdom
| | - Benjamin Jenkins
- Institute of Metabolic Science, MDU MRC, University of Cambridge Metabolic Research Laboratories, Cambridge, United Kingdom
| | - Xanthe A M H van Dierendonck
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands.,Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Aurelien Dugourd
- Joint Research Centre for Computational Biomedicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.,Institute for Computational Biomedicine, Faculty of Medicine & Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Joint Research Centre for Computational Biomedicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.,Institute for Computational Biomedicine, Faculty of Medicine & Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Rinke Stienstra
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands.,Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Albert Koulman
- Institute of Metabolic Science, MDU MRC, University of Cambridge Metabolic Research Laboratories, Cambridge, United Kingdom
| | - Antonio Vidal-Puig
- Institute of Metabolic Science, MDU MRC, University of Cambridge Metabolic Research Laboratories, Cambridge, United Kingdom.,Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
12
|
Xiao Y, Shu L, Wu X, Liu Y, Cheong LY, Liao B, Xiao X, Hoo RL, Zhou Z, Xu A. Fatty acid binding protein 4 promotes autoimmune diabetes by recruitment and activation of pancreatic islet macrophages. JCI Insight 2021; 6:141814. [PMID: 33690220 PMCID: PMC8119222 DOI: 10.1172/jci.insight.141814] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 02/18/2021] [Indexed: 12/25/2022] Open
Abstract
Both innate and adaptive immune cells are critical players in autoimmune destruction of insulin-producing β cells in type 1 diabetes. However, the early pathogenic events triggering the recruitment and activation of innate immune cells in islets remain obscure. Here we show that circulating fatty acid binding protein 4 (FABP4) level was significantly elevated in patients with type 1 diabetes and their first-degree relatives and positively correlated with the titers of several islet autoantibodies. In nonobese diabetic (NOD) mice, increased FABP4 expression in islet macrophages started from the neonatal period, well before the occurrence of overt diabetes. Furthermore, the spontaneous development of autoimmune diabetes in NOD mice was markedly reduced by pharmacological inhibition or genetic ablation of FABP4 or adoptive transfer of FABP4-deficient bone marrow cells. Mechanistically, FABP4 activated innate immune responses in islets by enhancing the infiltration and polarization of macrophages to proinflammatory M1 subtype, thus creating an inflammatory milieu required for activation of diabetogenic CD8+ T cells and shift of CD4+ helper T cells toward Th1 subtypes. These findings demonstrate FABP4 as a possible early mediator for β cell autoimmunity by facilitating crosstalk between innate and adaptive immune cells, suggesting that pharmacological inhibition of FABP4 may represent a promising therapeutic strategy for autoimmune diabetes.
Collapse
Affiliation(s)
- Yang Xiao
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lingling Shu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Hematologic Oncology, Cancer Center, Sun Yat-Sen University, Guangzhou, China
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Medicine, and
| | - Xiaoping Wu
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Yang Liu
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lai Yee Cheong
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Medicine, and
| | - Boya Liao
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Xiaoyu Xiao
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ruby L.C. Hoo
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Medicine, and
- Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
13
|
Lee CH, Lui DTW, Lam KSL. Adipocyte Fatty Acid-Binding Protein, Cardiovascular Diseases and Mortality. Front Immunol 2021; 12:589206. [PMID: 33815359 PMCID: PMC8017191 DOI: 10.3389/fimmu.2021.589206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 03/04/2021] [Indexed: 12/21/2022] Open
Abstract
It has been increasingly recognized that inflammation plays an important role in the pathogenesis of cardiovascular disease (CVD). In obesity, adipose tissue inflammation, especially in the visceral fat depots, contributes to systemic inflammation and promotes the development of atherosclerosis. Adipocyte fatty acid-binding protein (AFABP), a lipid chaperone abundantly secreted from the adipocytes and macrophages, is one of the key players mediating this adipose-vascular cross-talk, in part via its interaction with c-Jun NH2-terminal kinase (JNK) and activator protein-1 (AP-1) to form a positive feedback loop, and perpetuate inflammatory responses. In mice, selective JNK inactivation in the adipose tissue significantly reduced the expression of AFABP in their adipose tissue, as well as circulating AFABP levels. Importantly, fat transplant experiments showed that adipose-specific JNK inactivation in the visceral fat was sufficient to protect mice with apoE deficiency from atherosclerosis, with the beneficial effects attenuated by the continuous infusion of recombinant AFABP, supporting the role of AFABP as the link between visceral fat inflammation and atherosclerosis. In humans, raised circulating AFABP levels are associated with incident metabolic syndrome, type 2 diabetes and CVD, as well as non-alcoholic steatohepatitis, diabetic nephropathy and adverse renal outcomes, all being conditions closely related to inflammation and enhanced CV mortality. Collectively, these clinical data have provided support to AFABP as an important adipokine linking obesity, inflammation and CVD. This review will discuss recent findings on the role of AFABP in CVD and mortality, the possible underlying mechanisms, and pharmacological inhibition of AFABP as a potential strategy to combat CVD.
Collapse
Affiliation(s)
- Chi-Ho Lee
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, Hong Kong
| | - David T W Lui
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| | - Karen S L Lam
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
14
|
D’Anneo A, Bavisotto CC, Gammazza AM, Paladino L, Carlisi D, Cappello F, de Macario EC, Macario AJL, Lauricella M. Lipid chaperones and associated diseases: a group of chaperonopathies defining a new nosological entity with implications for medical research and practice. Cell Stress Chaperones 2020; 25:805-820. [PMID: 32856199 PMCID: PMC7591661 DOI: 10.1007/s12192-020-01153-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/16/2020] [Accepted: 08/10/2020] [Indexed: 02/08/2023] Open
Abstract
Fatty acid-binding proteins (FABPs) are lipid chaperones assisting in the trafficking of long-chain fatty acids with functions in various cell compartments, including oxidation, signaling, gene-transcription regulation, and storage. The various known FABP isoforms display distinctive tissue distribution, but some are active in more than one tissue. Quantitative and/or qualitative changes of FABPs are associated with pathological conditions. Increased circulating levels of FABPs are biomarkers of disorders such as obesity, insulin resistance, cardiovascular disease, and cancer. Deregulated expression and malfunction of FABPs can result from genetic alterations or posttranslational modifications and can be pathogenic. We have assembled the disorders with abnormal FABPs as chaperonopathies in a distinct nosological entity. This entity is similar but separate from that encompassing the chaperonopathies pertaining to protein chaperones. In this review, we discuss the role of FABPs in the pathogenesis of metabolic syndrome, cancer, and neurological diseases. We highlight the opportunities for improving diagnosis and treatment that open by encompassing all these pathological conditions within of a coherent nosological group, focusing on abnormal lipid chaperones as biomarkers of disease and etiological-pathogenic factors.
Collapse
Affiliation(s)
- Antonella D’Anneo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Laboratory of Biochemistry, University of Palermo, 90127 Palermo, Italy
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Anatomy, University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Anatomy, University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Letizia Paladino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Anatomy, University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Daniela Carlisi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy
| | - Francesco Cappello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Anatomy, University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Everly Conway de Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202 USA
| | - Alberto J. L. Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202 USA
| | - Marianna Lauricella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
15
|
Kim K, Ji P, Song M, Che TM, Bravo D, Pettigrew JE, Liu Y. Dietary plant extracts modulate gene expression profiles in alveolar macrophages of pigs experimentally infected with porcine reproductive and respiratory syndrome virus. J Anim Sci Biotechnol 2020; 11:74. [PMID: 32685145 PMCID: PMC7359597 DOI: 10.1186/s40104-020-00475-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/17/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Our previous study showed that 3 plant extracts enhanced the immune responses and growth efficiency of weaned pigs infected with porcine reproductive and respiratory syndrome virus (PRRSV), which is one of the most economically important disease in swine industry. However, each plant extract differently effected on growth efficiency and immune responses. Therefore, the objective of this study was conducted to characterize the effects and investigate the potential underlying mechanisms of 3 plant extracts on gene expression of alveolar macrophages in weaned pigs experimentally infected with PRRSV. RESULTS PRRSV infection altered (P < 0.05) the expression of 1,352 genes in pigs fed the control (CON; 755 up, 597 down). Compared with the infected CON, feeding capsicum (CAP), garlic botanical (GAR), or turmeric oleoresin (TUR) altered the expression of 46 genes (24 up, 22 down), 134 genes (59 up, 75 down), or 98 genes (55 up, 43 down) in alveolar macrophages of PRRSV-infected pigs, respectively. PRRSV infection up-regulated (P < 0.05) the expression of genes related to cell apoptosis, immune system process, and response to stimulus, but down-regulated (P < 0.05) the expression of genes involved in signaling transduction and innate immune response. Compared with the infected CON, feeding TUR or GAR reduced (P < 0.05) the expression of genes associated with antigen processing and presentation, feeding CAP up-regulated (P < 0.05) the expression of genes involved in antigen processing and presentation. Supplementation of CAP, GAR, or TUR also enhanced (P < 0.05) the expression of several genes related to amino acid metabolism, steroid hormone synthesis, or RNA degradation, respectively. CONCLUSIONS The results suggest that 3 plant extracts differently regulated the expression of genes in alveolar macrophages of PRRSV-infected pigs, especially altering genes involved in immunity.
Collapse
Affiliation(s)
- Kwangwook Kim
- Department of Animal Science, University of California, Davis, CA USA
| | - Peng Ji
- Department of Nutrition, University of California, Davis, CA USA
| | - Minho Song
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, South Korea
| | - Tung M. Che
- Department of Animal Production, Nong Lam University, Ho Chi Minh City, Vietnam
| | - David Bravo
- Pancosma SA, Geneva, Switzerland
- Current address: Land O’Lakes Inc., Arden Hills, MN USA
| | | | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA USA
| |
Collapse
|
16
|
Shi M, Ma L, Fu P. Role of Fatty Acid Binding Protein 4 (FABP4) in Kidney Disease. Curr Med Chem 2020; 27:3657-3664. [PMID: 30306857 DOI: 10.2174/0929867325666181008154622] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/12/2018] [Accepted: 09/26/2018] [Indexed: 02/08/2023]
Abstract
Accumulating evidences indicated that obesity and metabolic syndrome were independent risk factors for the development and progression of kidney diseases. Apart from inflammation, lipotoxicity, and hemodynamic factors, adipokines have been proposed to play crucial roles in the relationship between kidney diseases and metabolic disorders. As one of the key adipokines, fatty acid binding protein 4 (FABP4), which is mainly expressed in adipocytes and macrophages, has recently been shown to be associated with renal dysfunction and kidney damage. Both clinical and experimental studies have proposed circulating FABP4 as a novel predictor for renal injuries, and it might also be a predictor for cardiovascular events in patients with end stage renal disease (ESRD). FABP4 has also been detected in the glomerular cells and epithelial tubular cells in mouse and human kidneys, and the expression of FABP4 in these cells has been involved in the pathogenesis of kidney diseases. In addition, experimental studies suggested that inhibition of FABP4 had protective effects on renal damage. Here, we reviewed current knowledge regarding the role of FABP4 in pathophysiological insights as well as its potential function as a predictor and therapeutic target for kidney diseases.
Collapse
Affiliation(s)
- Min Shi
- Kidney Research Institute, Division of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Liang Ma
- Kidney Research Institute, Division of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ping Fu
- Kidney Research Institute, Division of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
17
|
Jaén RI, Val-Blasco A, Prieto P, Gil-Fernández M, Smani T, López-Sendón JL, Delgado C, Boscá L, Fernández-Velasco M. Innate Immune Receptors, Key Actors in Cardiovascular Diseases. JACC Basic Transl Sci 2020; 5:735-749. [PMID: 32760860 PMCID: PMC7393405 DOI: 10.1016/j.jacbts.2020.03.015] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the industrialized world. Most CVDs are associated with increased inflammation that arises mainly from innate immune system activation related to cardiac damage. Sustained activation of the innate immune system frequently results in maladaptive inflammatory responses that promote cardiovascular dysfunction and remodeling. Much research has focused on determining whether some mediators of the innate immune system are potential targets for CVD therapy. The innate immune system has specific receptors-termed pattern recognition receptors (PRRs)-that not only recognize pathogen-associated molecular patterns, but also sense danger-associated molecular signals. Activation of PRRs triggers the inflammatory response in different physiological systems, including the cardiovascular system. The classic PRRs, toll-like receptors (TLRs), and the more recently discovered nucleotide-binding oligomerization domain-like receptors (NLRs), have been recently proposed as key partners in the progression of several CVDs (e.g., atherosclerosis and heart failure). The present review discusses the key findings related to the involvement of TLRs and NLRs in the progression of several vascular and cardiac diseases, with a focus on whether some NLR subtypes (nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain-containing receptor 3 and nucleotide-binding oligomerization domain-containing protein 1) can be candidates for the development of new therapeutic strategies for several CVDs.
Collapse
Key Words
- AMI, acute myocardial infarction
- CARD, caspase activation and recruitment domain
- CVD, cardiovascular disease
- Ca2+, calcium ion
- DAMPs, danger-associated molecular patterns
- DAP, D-glutamyl-meso-diaminopimelic acid
- ER, endoplasmic reticulum
- HF, heart failure
- I/R, ischemia/reperfusion
- IL, interleukin
- MAPK, mitogen-activated protein kinase
- NF-κB, nuclear factor κ-light-chain-enhancer of activated B cells
- NLR, nucleotide-binding oligomerization domain-like receptors
- NLRP, nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain-containing receptor
- NLRP3
- NOD, Nucleotide-binding oligomerization domain-containing protein
- NOD1
- PAMP, pathogen-associated molecular pattern
- ROS, reactive oxygen species
- SR, sarcoplasmic reticulum
- TLR, toll-like receptor
- cardiovascular disease
- innate immune system
- nucleotide-binding oligomerization domain-like receptors
- toll-like receptors
Collapse
Affiliation(s)
- Rafael I. Jaén
- Biomedical Research Institute “Alberto Sols” CSIC-UAM, Madrid, Spain
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
| | - Almudena Val-Blasco
- Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Patricia Prieto
- Biomedical Research Institute “Alberto Sols” CSIC-UAM, Madrid, Spain
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
- Pharmacology, Pharmacognosy and Botany department, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Marta Gil-Fernández
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
- Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Tarik Smani
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Sevilla, Spain
| | - José Luis López-Sendón
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
- Servicio de Cardiología, Hospital Universitario La Paz, Madrid, Spain
| | - Carmen Delgado
- Biomedical Research Institute “Alberto Sols” CSIC-UAM, Madrid, Spain
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
| | - Lisardo Boscá
- Biomedical Research Institute “Alberto Sols” CSIC-UAM, Madrid, Spain
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
| | - María Fernández-Velasco
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
- Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
18
|
Su H, Zou Y, Chen G, Dou H, Xie H, Yuan X, Zhang X, Zhang N, Li M, Xu Y. Exploration of Fragment Binding Poses Leading to Efficient Discovery of Highly Potent and Orally Effective Inhibitors of FABP4 for Anti-inflammation. J Med Chem 2020; 63:4090-4106. [DOI: 10.1021/acs.jmedchem.9b02107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Haixia Su
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Zou
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guofeng Chen
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huixia Dou
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hang Xie
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaojing Yuan
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianglei Zhang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Naixia Zhang
- Department of Analytical Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minjun Li
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yechun Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
19
|
Yan Y, Song D, Wu J, Wang J. Long Non-Coding RNAs Link Oxidized Low-Density Lipoprotein With the Inflammatory Response of Macrophages in Atherogenesis. Front Immunol 2020; 11:24. [PMID: 32082313 PMCID: PMC7003668 DOI: 10.3389/fimmu.2020.00024] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is characterized as a chronic inflammatory response to cholesterol deposition in arteries. Low-density lipoprotein (LDL), especially the oxidized form (ox-LDL), plays a crucial role in the occurrence and development of atherosclerosis by inducing endothelial cell (EC) dysfunction, attracting monocyte-derived macrophages, and promoting chronic inflammation. However, the mechanisms linking cholesterol accumulation with inflammation in macrophage foam cells are poorly understood. Long non-coding RNAs (lncRNAs) are a group of non-protein-coding RNAs longer than 200 nucleotides and are found to regulate the progress of atherosclerosis. Recently, many lncRNAs interfering with cholesterol deposition or inflammation were identified, which might help elucidate their underlying molecular mechanism or be used as novel therapeutic targets. In this review, we summarize and highlight the role of lncRNAs linking cholesterol (mainly ox-LDL) accumulation with inflammation in macrophages during the process of atherosclerosis.
Collapse
Affiliation(s)
- Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Dandan Song
- Department of Clinical Laboratory, Second Hospital of Jilin University, Changchun, China
| | - Junduo Wu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Korbecki J, Bajdak-Rusinek K. The effect of palmitic acid on inflammatory response in macrophages: an overview of molecular mechanisms. Inflamm Res 2019; 68:915-932. [PMID: 31363792 PMCID: PMC6813288 DOI: 10.1007/s00011-019-01273-5] [Citation(s) in RCA: 294] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
Palmitic acid is a saturated fatty acid whose blood concentration is elevated in obese patients. This causes inflammatory responses, where toll-like receptors (TLR), TLR2 and TLR4, play an important role. Nevertheless, palmitic acid is not only a TLR agonist. In the cell, this fatty acid is converted into phospholipids, diacylglycerol and ceramides. They trigger the activation of various signaling pathways that are common for LPS-mediated TLR4 activation. In particular, metabolic products of palmitic acid affect the activation of various PKCs, ER stress and cause an increase in ROS generation. Thanks to this, palmitic acid also strengthens the TLR4-induced signaling. In this review, we discuss the mechanisms of inflammatory response induced by palmitic acid. In particular, we focus on describing its effect on ER stress and IRE1α, and the mechanisms of NF-κB activation. We also present the mechanisms of inflammasome NLRP3 activation and the effect of palmitic acid on enhanced inflammatory response by increasing the expression of FABP4/aP2. Finally, we focus on the consequences of inflammatory responses, in particular, the effect of TNF-α, IL-1β and IL-6 on insulin resistance. Due to the high importance of macrophages and the production of proinflammatory cytokines by them, this work mainly focuses on these cells.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Molecular Biology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18 St., 40-752, Katowice, Poland.
| | - Karolina Bajdak-Rusinek
- Department of Medical Genetics, School of Medicine in Katowice, Medical University of Silesia, Medyków 18 St., 40-752, Katowice, Poland
| |
Collapse
|
21
|
Wang CP, Hsu CC, Hung WC, Yu TH, Wu CC, Tsai IT, Tang WH, Chung FM, Houng JY, Lee YJ, Lu YC. Plasma fatty acid-binding protein 4 (FABP4) level is associated with abnormal QTc interval in patients with stable angina and chronic kidney disease. BMC Cardiovasc Disord 2019; 19:153. [PMID: 31234795 PMCID: PMC6591904 DOI: 10.1186/s12872-019-1134-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 06/12/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Fatty acid-binding protein 4 (FABP4) (also known as adipocyte FABP or adipocyte P2) is expressed in adipocytes, macrophages, and capillary endothelial cells. Previous studies have shown associations among plasma FABP4, insulin resistance, metabolic syndrome, diabetes mellitus, greater coronary plaque burden, coronary artery disease, heart failure, and mortality. However, little is known about the relationship between FABP4 level and prolonged QT interval. The aim of this study was to investigate whether plasma FABP4 level is associated with a prolonged QT interval by analyzing 12-lead electrocardiograms (ECGs) in patients with stable angina and chronic kidney disease (CKD). METHODS This study included 397 consecutive patients with stable angina and CKD who were enrolled in a disease management program. Plasma FABP4 concentrations were measured using enzyme-linked immunosorbent assays. A 12-lead ECG recording was obtained from each patient. We assessed the relationships between FABP4 levels (both as a continuous variable and stratified by tertile) at admission and corrected QT (QTc) prolongation. RESULTS Patients with an abnormal QTc interval had higher median plasma FABP4 levels than those with borderline and normal QTc intervals (15.9 ng/mL vs. 10.2 ng/mL vs. 8.5 ng/mL, respectively, P < 0.0001). Statistically significant associations were observed between plasma FABP4 levels and QTc interval (β = 0.267, P < 0.0001). Using multivariate and trend analyses, a higher concentration of plasma FABP4 level was independently associated with QTc prolongation in patients with stable angina and CKD. CONCLUSION In this study, plasma FABP4 levels were significantly higher in the patients with an abnormal QTc interval and were correlated with QTc prolongation. Further studies are required to elucidate whether plasma FABP4 plays a role in the pathogenesis of QTc prolongation.
Collapse
Affiliation(s)
- Chao-Ping Wang
- Division of Cardiology, I-Shou University, Kaohsiung, 82445, Taiwan.,School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Chia-Chang Hsu
- Division of Gastroenterology and Hepatology, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Wei-Chin Hung
- Division of Cardiology, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Teng-Hung Yu
- Division of Cardiology, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Cheng-Ching Wu
- Division of Cardiology, I-Shou University, Kaohsiung, 82445, Taiwan
| | - I-Ting Tsai
- Department of Emergency, E-Da Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Wei-Hua Tang
- Division of Cardiology, Department of Internal Medicine, National Yang-Ming University Hospital, Yilan, 26058, Taiwan
| | - Fu-Mei Chung
- Division of Cardiology, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Jer-Yiing Houng
- Department of Nutrition, Institute of Biotechnology and Chemical Engineering, College of Medicine, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Yau-Jiunn Lee
- Lee's Endocrinologic Clinic, Pingtung, 90000, Taiwan
| | - Yung-Chuan Lu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, E-Da Hospital, I-Shou University, No. 1, Yi-Da Rd, Jiau-Shu Village, Yan-Chao District, Kaohsiung, 82445, Taiwan. .,School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, 82445, Taiwan.
| |
Collapse
|
22
|
Deng T, Wang Y, Wang C, Yan H. FABP4 silencing ameliorates hypoxia reoxygenation injury through the attenuation of endoplasmic reticulum stress-mediated apoptosis by activating PI3K/Akt pathway. Life Sci 2019; 224:149-156. [PMID: 30904493 DOI: 10.1016/j.lfs.2019.03.046] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/19/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Endoplasmic reticulum (ER) stress and subsequent apoptosis play a vital role in myocardial ischemia reperfusion (IR) injury. Fatty acid binding protein 4 (FABP4) may induce ER stress. The aim of this study was to investigate the mechanism and effect of FABP4 on IR injury in vitro. Rat H9c2 cells were exposed to hypoxia reoxygenation (HR) to create an IR model in vitro. FABP4 was overexpressed in HR-injured H9c2 cells. Transfection with FABP4 siRNA increased cell viability and decreased LDH upon HR stimulation. FABP4 cessation also suppressed apoptotic cells and caspase-3 activity after HR. Downregulation of FABP4 significantly inhibited ER stress by decreasing the protein expression of p-PERK, GRP78, and ATF6. FABP4 silencing also restrained the ER stress-mediated apoptotic pathway, as indicated by decreased pro-apoptotic proteins p-JNK, CHOP, Bax, and caspase-12, as well as upregulation of Bcl-2 during HR. Furthermore, FABP4 silencing activated the PI3K/Akt pathway. Blocking this pathway by the specific PI3K inhibitor-LY294002 restored HR-induced ER stress and subsequently reversed the protective effect of FABP4 silencing on HR injury. Taken together, our findings revealed that FABP4 silencing exerts protective effects against HR injury in H9c2 cells through inhibiting ER stress-induced cell apoptosis via activation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Tianming Deng
- Department of Cardiology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China.
| | - Yanhong Wang
- Department of Geriatrcs, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Chongchong Wang
- Department of Cardiology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Hua Yan
- Department of Cardiology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| |
Collapse
|
23
|
Trojnar M, Patro-Małysza J, Kimber-Trojnar Ż, Leszczyńska-Gorzelak B, Mosiewicz J. Associations between Fatty Acid-Binding Protein 4⁻A Proinflammatory Adipokine and Insulin Resistance, Gestational and Type 2 Diabetes Mellitus. Cells 2019; 8:cells8030227. [PMID: 30857223 PMCID: PMC6468522 DOI: 10.3390/cells8030227] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/03/2019] [Accepted: 03/03/2019] [Indexed: 12/12/2022] Open
Abstract
There is ample scientific evidence to suggest a link between the fatty acid-binding protein 4 (FABP4) and insulin resistance, gestational (GDM), and type 2 (T2DM) diabetes mellitus. This novel proinflammatory adipokine is engaged in the regulation of lipid metabolism at the cellular level. The molecule takes part in lipid oxidation, the regulation of transcription as well as the synthesis of membranes. An involvement of FABP4 in the pathogenesis of obesity and insulin resistance seems to be mediated via FABP4-dependent peroxisome proliferator-activated receptor γ (PPARγ) inhibition. A considerable number of studies have shown that plasma concentrations of FABP4 is increased in obesity and T2DM, and that circulating FABP4 levels are correlated with certain clinical parameters, such as body mass index, insulin resistance, and dyslipidemia. Since plasma-circulating FABP4 has the potential to modulate the function of several types of cells, it appears to be of extreme interest to try to develop potential therapeutic strategies targeting the pathogenesis of metabolic diseases in this respect. In this manuscript, representing a detailed review of the literature on FABP4 and the abovementioned metabolic disorders, various mechanisms of the interaction of FABP4 with insulin signaling pathways are thoroughly discussed. Clinical aspects of insulin resistance in diabetic patients, including women diagnosed with GDM, are analyzed as well.
Collapse
Affiliation(s)
- Marcin Trojnar
- Chair and Department of Internal Medicine, Medical University of Lublin, 20-081 Lublin, Poland.
| | - Jolanta Patro-Małysza
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland.
| | - Żaneta Kimber-Trojnar
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland.
| | | | - Jerzy Mosiewicz
- Chair and Department of Internal Medicine, Medical University of Lublin, 20-081 Lublin, Poland.
| |
Collapse
|
24
|
Abstract
Fatty acid-binding proteins (FABPs), a family of lipid chaperones, contribute to systemic metabolic regulation via several lipid signaling pathways. Fatty acid-binding protein 4 (FABP4), known as adipocyte FABP (A-FABP) or aP2, is mainly expressed in adipocytes and macrophages and plays important roles in the development of insulin resistance and atherosclerosis in relation to metabolically driven low-grade and chronic inflammation, referred to as ‘metaflammation’. FABP4 is secreted from adipocytes in a non-classical pathway associated with lipolysis and acts as an adipokine for the development of insulin resistance and atherosclerosis. Circulating FABP4 levels are associated with several aspects of metabolic syndrome and cardiovascular disease. Ectopic expression and function of FABP4 in cells and tissues are also related to the pathogenesis of several diseases. Pharmacological modification of FABP4 function by specific inhibitors, neutralizing antibodies or antagonists of unidentified receptors would be novel therapeutic strategies for several diseases, including obesity, diabetes mellitus, atherosclerosis and cardiovascular disease. Significant roles of FABP4 as a lipid chaperone in physiological and pathophysiological conditions and the possibility of FABP4 being a therapeutic target for metabolic and cardiovascular diseases are discussed in this review.
Collapse
Affiliation(s)
- Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine
| |
Collapse
|
25
|
Glycerol-3-phosphate acyltransferases 3 and 4 direct glycerolipid synthesis and affect functionality in activated macrophages. Biochem J 2019; 476:85-99. [DOI: 10.1042/bcj20180381] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 12/13/2022]
Abstract
AbstractMacrophage classical M1 activation via TLR4 triggers a variety of responses to achieve the elimination of foreign pathogens. During this process, there is also an increase in lipid droplets which contain large quantities of triacylglycerol (TAG) and phospholipid (PL). The functional consequences of this increment in lipid mass are poorly understood. Here, we studied the contribution of glycerolipid synthesis to lipid accumulation, focusing specifically on the first and rate-limiting enzyme of the pathway: glycerol-3-phosphate acyltransferase (GPAT). Using bone marrow-derived macrophages (BMDMs) treated with Kdo2-lipid A, we showed that glycerolipid synthesis is induced during macrophage activation. GPAT4 protein level and GPAT3/GPAT4 enzymatic activity increase during this process, and these two isoforms were required for the accumulation of cell TAG and PL. The phagocytic capacity of Gpat3−/− and Gpat4−/− BMDM was impaired. Additionally, inhibiting fatty acid β-oxidation reduced phagocytosis only partially, suggesting that lipid accumulation is not necessary for the energy requirements for phagocytosis. Finally, Gpat4−/− BMDM expressed and released more pro-inflammatory cytokines and chemokines after macrophage activation, suggesting a role for GPAT4 in suppressing inflammatory responses. Together, these results provide evidence that glycerolipid synthesis directed by GPAT4 is important for the attenuation of the inflammatory response in activated macrophages.
Collapse
|
26
|
Liu L, Cui HX, Zheng MQ, Zhao GP, Wen J. Comparative analysis of differentially expressed genes related to triglyceride metabolism between intramuscular fat and abdominal fat in broilers. Br Poult Sci 2018; 59:514-520. [DOI: 10.1080/00071668.2018.1483573] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- L. Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- State Key Laboratory of Animal Nutrition, Beijing, China
| | - H. X. Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- State Key Laboratory of Animal Nutrition, Beijing, China
| | - M. Q. Zheng
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- State Key Laboratory of Animal Nutrition, Beijing, China
| | - G. P. Zhao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- State Key Laboratory of Animal Nutrition, Beijing, China
| | - J. Wen
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- State Key Laboratory of Animal Nutrition, Beijing, China
| |
Collapse
|
27
|
Yan F, Liu X, Zhang S, Su J, Zhang Q, Chen J. Molecular Dynamics Exploration of Selectivity of Dual Inhibitors 5M7, 65X, and 65Z toward Fatty Acid Binding Proteins 4 and 5. Int J Mol Sci 2018; 19:ijms19092496. [PMID: 30142969 PMCID: PMC6164837 DOI: 10.3390/ijms19092496] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/14/2018] [Accepted: 08/18/2018] [Indexed: 12/11/2022] Open
Abstract
Designing highly selective inhibitors of fatty acid binding proteins 4 and 5 (FABP4 and FABP5) is of importance for treatment of some diseases related with inflammation, metabolism, and tumor growth. In this study, molecular dynamics (MD) simulations combined with molecular mechanics generalized Born surface area (MM-GBSA) method were performed to probe binding selectivity of three inhibitors (5M7, 65X, and 65Z) to FABP4/FABP5 with Ki values of 0.022/0.50 μM, 0.011/0.086 μM, and 0.016/0.12 μM, respectively. The results not only suggest that all inhibitors associate more tightly with FABP4 than FABP5, but also prove that the main forces driving the selective bindings of inhibitors to FABP4 and FABP5 stem from the difference in the van der Waals interactions and polar interactions of inhibitors with two proteins. Meanwhile, a residue-based free energy decomposition method was applied to reveal molecular basis that inhibitors selectively interact with individual residues of two different proteins. The calculated results show that the binding difference of inhibitors to the residues (Phe16, Phe19), (Ala33, Gly36), (Phe57, Leu60), (Ala75, Ala78), (Arg126, Arg129), and (Tyr128, Tyr131) in (FABP4, FABP5) drive the selectivity of inhibitors toward FABP4 and FABP5. This study will provide great help for further design of effective drugs to protect against a series of metabolic diseases, arteriosclerosis, and inflammation.
Collapse
Affiliation(s)
- Fangfang Yan
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, China.
| | - Xinguo Liu
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, China.
| | - Shaolong Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, China.
| | - Jing Su
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, China.
| | - Qinggang Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, China.
| | - Jianzhong Chen
- School of Science, Shandong Jiaotong University, Jinan 250357, China.
| |
Collapse
|
28
|
Thomas SS, Kim M, Lee SJ, Cha YS. Antiobesity Effects of Purple Perilla (Perilla frutescens var. acuta) on Adipocyte Differentiation and Mice Fed a High-fat Diet. J Food Sci 2018; 83:2384-2393. [PMID: 30070698 DOI: 10.1111/1750-3841.14288] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/31/2018] [Accepted: 06/24/2018] [Indexed: 02/06/2023]
Abstract
Purple perilla (PE) is a medicinal plant that has several health benefits. In this study, the antiobesity effect of PE was studied in 3T3-L1 preadipocytes and C57BL/6J mice fed high-fat diets. Triglyceride quantification and Oil Red O staining in matured adipocytes revealed that PE reduced lipid accumulation in differentiated adipocytes by downregulating adipogenic gene and upregulating lipolytic gene expressions. Mice were fed normal diet, high-fat diet and high-fat diet supplemented with different concentrations of PE. Treatment with PE significantly prevented body weight gain, improved serum lipids, hepatic lipids and reduced the epididymal fat. Furthermore, in the adipose tissue and liver, expression of genes related to lipolysis and fatty acid β-oxidation were upregulated in PE- treated mice. Thus, our results suggested that PE has antiobesity effects in rodents and can be effective in obesity management. PRACTICAL APPLICATION Purple perilla, rich in polyphenols such as rosmarinic acid, showed lipid lowering in adipocyte cells and prevented body weight gain in mice. Therefore we conclude that purple perilla may be a potential candidate for the development of functional foods or nutraceuticals in managing obesity in humans.
Collapse
Affiliation(s)
- Shalom Sara Thomas
- Dept. of Food Science and Human Nutrition, Chonbuk National Univ., Deokjin-gu, Jeonju, 54896, Republic of Korea
| | - Mina Kim
- Div. of Functional Food and Nutrition, Dept. of Agrofood Resources, National Inst. of Agricultural Science, Rural Development Administration, Wanju, 55365, Republic of Korea
| | - Seung Je Lee
- Research and Development Office, Jeonbuk Inst. for Food-Bioindustry, Jeonju, 54810, Republic of Korea
| | - Youn-Soo Cha
- Dept. of Food Science and Human Nutrition, Chonbuk Natl. Univ., Deokjin-gu, Jeonju, Jeollabuk-do, 54896, Republic of Korea
| |
Collapse
|
29
|
Intermittent High Glucose Exacerbates A-FABP Activation and Inflammatory Response through TLR4-JNK Signaling in THP-1 Cells. J Immunol Res 2018; 2018:1319272. [PMID: 29850615 PMCID: PMC5925160 DOI: 10.1155/2018/1319272] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/15/2018] [Accepted: 03/19/2018] [Indexed: 01/01/2023] Open
Abstract
Background Glucose fluctuation confers additional risks on diabetes-related vascular diseases, but the underlying mechanisms are unknown. Macrophage activation mediated by TLR4-JNK signaling plays an important role during the progress of diabetes. In the present study, we hypothesize that glucose fluctuation results in macrophage inflammation through TLR4-JNK signaling pathways. Methods THP-1 cells were treated with normal glucose (5 mM), constant high glucose (25 mM), and intermittent high glucose (rotation per 6 h in 5 mM or 25 mM) for 24 h. The mRNA and protein expression levels of TLR4, p-JNK, and adipocyte fatty acid-binding protein (A-FABP) were determined, and the proinflammatory cytokines TNF-α and IL-1β were quantified. Results In constant high glucose, TLR4 expression and JNK phosphorylation levels increased, and this effect was more pronounced in intermittent high glucose. Accordingly, the expression of A-FABP and the release of the proinflammatory cytokines TNF-α and IL-1β also increased in response to constant high glucose, an effect that also was more evident in intermittent high glucose. The inhibition of p-JNK by SP600125 did not attenuate TLR4 expression, but totally inhibited both A-FABP expression and the production of the proinflammatory cytokines TNF-α and IL-1β in both constant and intermittent high glucose. Conclusions Intermittent high glucose potentiates A-FABP activation and inflammatory responses via TLR4/p-JNK signaling in THP-1 cells. These findings suggest a more detrimental impact of glucose fluctuation on macrophage inflammation in diabetes-related vascular diseases than thus far generally assumed.
Collapse
|
30
|
Shi M, Huang R, Guo F, Li L, Feng Y, Wei Z, Zhou L, Ma L, Fu P. Pharmacological inhibition of fatty acid-binding protein 4 (FABP4) protects against renal ischemia-reperfusion injury. RSC Adv 2018; 8:15207-15214. [PMID: 35541316 PMCID: PMC9079974 DOI: 10.1039/c8ra00122g] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/18/2018] [Indexed: 02/05/2023] Open
Abstract
Fatty acid-binding protein 4 (FABP4) is a key mediator of endoplasmic reticulum (ER) stress and apoptosis in diabetes and atherosclerosis. Studies also confirmed that circulating FABP4 depended on renal function in chronic kidney disease (CKD) and acute kidney injury (AKI) patients. However, the function of FABP4 in AKI remains poorly understood and the aim of this study was to investigate the role of FABP4 in ischemia-reperfusion (I/R)-induced AKI. In the present study, renal I/R injury triggered the high expression of the FABP4 gene and protein in the nucleus and cytoplasm of tubular cells of mouse kidney tissue compared to that of Sham. Pretreatment with BMS309403, a highly selective inhibitor of FABP4 at a dose of 20 mg kg−1 d−1 for 4 d, significantly reduced serum creatinine levels to improve acute renal dysfunction and attenuated renal tubular damage in injured kidneys. Pharmacological inhibition of FABP4 also decreased the number of TdT-mediated dUTP nick-end labeling (TUNEL) positive apoptotic tubular cells, accompanied by the down-regulation of cleaved-caspase-3 expression. Furthermore, oral administration of FABP4 inhibitor resulted in a significant attenuation of ER stress indicated by its maker proteins expression of glucose-regulated protein 78 (GRP78), C/EBP homologous protein (CHOP), and caspase-12 in I/R injured kidneys. In vitro, the increased expression of FABP4 in the human renal proximal tubule cell line (HK-2 cell) was induced by hypoxia followed by reoxygenation (HR) and the FABP4 inhibitor resulted in a significant attenuation of cell apoptosis and ER stress in HR-induced HK-2 cells. In summary, these findings indicated that FABP4 contributed to the pathogenesis of I/R-induced AKI and suggested that the inhibition of FABP4 might be a promising therapeutic strategy for AKI treatment. FABP4 inhibition might attenuate I/R-induced AKI through reducing ER stress and apoptosis.![]()
Collapse
Affiliation(s)
- Min Shi
- Kidney Research Institute
- Division of Nephrology
- West China Hospital of Sichuan University
- Chengdu 610041
- China
| | - Rongshuang Huang
- Kidney Research Institute
- Division of Nephrology
- West China Hospital of Sichuan University
- Chengdu 610041
- China
| | - Fan Guo
- Kidney Research Institute
- Division of Nephrology
- West China Hospital of Sichuan University
- Chengdu 610041
- China
| | - Lingzhi Li
- Kidney Research Institute
- Division of Nephrology
- West China Hospital of Sichuan University
- Chengdu 610041
- China
| | - Yanhuan Feng
- Kidney Research Institute
- Division of Nephrology
- West China Hospital of Sichuan University
- Chengdu 610041
- China
| | | | - Li Zhou
- Kidney Research Institute
- Division of Nephrology
- West China Hospital of Sichuan University
- Chengdu 610041
- China
| | - Liang Ma
- Kidney Research Institute
- Division of Nephrology
- West China Hospital of Sichuan University
- Chengdu 610041
- China
| | - Ping Fu
- Kidney Research Institute
- Division of Nephrology
- West China Hospital of Sichuan University
- Chengdu 610041
- China
| |
Collapse
|
31
|
Hashimoto R, Kakigi R, Nakamura K, Itoh S, Daida H, Okada T, Katoh Y. LPS enhances expression of CD204 through the MAPK/ERK pathway in murine bone marrow macrophages. Atherosclerosis 2017; 266:167-175. [DOI: 10.1016/j.atherosclerosis.2017.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/09/2017] [Accepted: 10/05/2017] [Indexed: 01/08/2023]
|
32
|
Wiśniewska A, Olszanecki R, Totoń-Żurańska J, Kuś K, Stachowicz A, Suski M, Gębska A, Gajda M, Jawień J, Korbut R. Anti-Atherosclerotic Action of Agmatine in ApoE-Knockout Mice. Int J Mol Sci 2017; 18:ijms18081706. [PMID: 28777310 PMCID: PMC5578096 DOI: 10.3390/ijms18081706] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 07/31/2017] [Accepted: 08/01/2017] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is an inflammatory disease in which dysfunction of mitochondria play an important role, and disorders of lipid management intensify this process. Agmatine, an endogenous polyamine formed by decarboxylation of arginine, exerts a protective effect on mitochondria and modulates fatty acid metabolism. We investigated the effect of exogenous agmatine on the development of atherosclerosis and changes in lipid profile in apolipoprotein E knockout (apoE-/-) mice. Agmatine caused an approximate 40% decrease of atherosclerotic lesions, as estimated by en face and cross-section methods with an influence on macrophage but not on smooth muscle content in the plaques. Agmatine treatment did not changed gelatinase activity within the plaque area. What is more, the action of agmatine was associated with an increase in the number of high density lipoproteins (HDL) in blood. Real-Time PCR analysis showed that agmatine modulates liver mRNA levels of many factors involved in oxidation of fatty acid and cholesterol biosynthesis. Two-dimensional electrophoresis coupled with mass spectrometry identified 27 differentially expressed mitochondrial proteins upon agmatine treatment in the liver of apoE-/- mice, mostly proteins related to metabolism and apoptosis. In conclusion, prolonged administration of agmatine inhibits atherosclerosis in apoE-/- mice; however, the exact mechanisms linking observed changes and elevations of HDL plasma require further investigation.
Collapse
Affiliation(s)
- Anna Wiśniewska
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Rafał Olszanecki
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Justyna Totoń-Żurańska
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Katarzyna Kuś
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Aneta Stachowicz
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Maciej Suski
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Anna Gębska
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Mariusz Gajda
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland.
| | - Jacek Jawień
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| | - Ryszard Korbut
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland.
| |
Collapse
|
33
|
Antonopoulos AS, Sanna F, Sabharwal N, Thomas S, Oikonomou EK, Herdman L, Margaritis M, Shirodaria C, Kampoli AM, Akoumianakis I, Petrou M, Sayeed R, Krasopoulos G, Psarros C, Ciccone P, Brophy CM, Digby J, Kelion A, Uberoi R, Anthony S, Alexopoulos N, Tousoulis D, Achenbach S, Neubauer S, Channon KM, Antoniades C. Detecting human coronary inflammation by imaging perivascular fat. Sci Transl Med 2017; 9:eaal2658. [PMID: 28701474 DOI: 10.1126/scitranslmed.aal2658] [Citation(s) in RCA: 639] [Impact Index Per Article: 79.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/30/2017] [Accepted: 05/30/2017] [Indexed: 12/11/2022]
Abstract
Early detection of vascular inflammation would allow deployment of targeted strategies for the prevention or treatment of multiple disease states. Because vascular inflammation is not detectable with commonly used imaging modalities, we hypothesized that phenotypic changes in perivascular adipose tissue (PVAT) induced by vascular inflammation could be quantified using a new computerized tomography (CT) angiography methodology. We show that inflamed human vessels release cytokines that prevent lipid accumulation in PVAT-derived preadipocytes in vitro, ex vivo, and in vivo. We developed a three-dimensional PVAT analysis method and studied CT images of human adipose tissue explants from 453 patients undergoing cardiac surgery, relating the ex vivo images with in vivo CT scan information on the biology of the explants. We developed an imaging metric, the CT fat attenuation index (FAI), that describes adipocyte lipid content and size. The FAI has excellent sensitivity and specificity for detecting tissue inflammation as assessed by tissue uptake of 18F-fluorodeoxyglucose in positron emission tomography. In a validation cohort of 273 subjects, the FAI gradient around human coronary arteries identified early subclinical coronary artery disease in vivo, as well as detected dynamic changes of PVAT in response to variations of vascular inflammation, and inflamed, vulnerable atherosclerotic plaques during acute coronary syndromes. Our study revealed that human vessels exert paracrine effects on the surrounding PVAT, affecting local intracellular lipid accumulation in preadipocytes, which can be monitored using a CT imaging approach. This methodology can be implemented in clinical practice to noninvasively detect plaque instability in the human coronary vasculature.
Collapse
Affiliation(s)
- Alexios S Antonopoulos
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Fabio Sanna
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Nikant Sabharwal
- Cardiothoracic Directorate, Oxford University Hospitals National Health System (NHS) Foundation Trust, Oxford, UK
| | - Sheena Thomas
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Evangelos K Oikonomou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Laura Herdman
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Marios Margaritis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre of Research Excellence, British Heart Foundation, Oxford, UK
| | - Cheerag Shirodaria
- Cardiothoracic Directorate, Oxford University Hospitals National Health System (NHS) Foundation Trust, Oxford, UK
| | - Anna-Maria Kampoli
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Mario Petrou
- Department of Cardiothoracic Surgery, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Rana Sayeed
- Department of Cardiothoracic Surgery, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - George Krasopoulos
- Department of Cardiothoracic Surgery, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Constantinos Psarros
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Patricia Ciccone
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Carl M Brophy
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Janet Digby
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew Kelion
- Cardiothoracic Directorate, Oxford University Hospitals National Health System (NHS) Foundation Trust, Oxford, UK
| | - Raman Uberoi
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Suzan Anthony
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Dimitris Tousoulis
- 1st Department of Cardiology, Athens University Medical School, Athens, Greece
| | - Stephan Achenbach
- Medizinische Klinik 2, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre of Research Excellence, British Heart Foundation, Oxford, UK
- Oxford Biomedical Research Centre, National Institute of Health Research, Oxford, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre of Research Excellence, British Heart Foundation, Oxford, UK
- Oxford Biomedical Research Centre, National Institute of Health Research, Oxford, UK
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Oxford Centre of Research Excellence, British Heart Foundation, Oxford, UK
- Oxford Biomedical Research Centre, National Institute of Health Research, Oxford, UK
| |
Collapse
|
34
|
Rodríguez-Calvo R, Girona J, Alegret JM, Bosquet A, Ibarretxe D, Masana L. Role of the fatty acid-binding protein 4 in heart failure and cardiovascular disease. J Endocrinol 2017; 233:R173-R184. [PMID: 28420707 DOI: 10.1530/joe-17-0031] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 04/18/2017] [Indexed: 01/05/2023]
Abstract
Obesity and ectopic fat accumulation in non-adipose tissues are major contributors to heart failure (HF) and cardiovascular disease (CVD). Adipocytes act as endocrine organs by releasing a large number of bioactive molecules into the bloodstream, which participate in a communication network between white adipose tissue and other organs, including the heart. Among these molecules, fatty acid-binding protein 4 (FABP4) has recently been shown to increase cardiometabolic risk. Both clinical and experimental evidence have identified FABP4 as a relevant player in atherosclerosis and coronary artery disease, and it has been directly related to cardiac alterations such as left ventricular hypertrophy (LVH) and both systolic and diastolic cardiac dysfunction. The available interventional studies preclude the establishment of a direct causal role of this molecule in CVD and HF and propose FABP4 as a biomarker rather than as an aetiological factor. However, several experimental reports have suggested that FABP4 may act as a direct contributor to cardiac metabolism and physiopathology, and the pharmacological targeting of FABP4 may restore some of the metabolic alterations that are conducive to CVD and HF. Here, we review the current knowledge regarding FABP4 in the context of HF and CVD as well as the molecular basis by which this protein participates in the regulation of cardiac function.
Collapse
Affiliation(s)
- Ricardo Rodríguez-Calvo
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Josefa Girona
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Josep M Alegret
- Department of CardiologyCardiovascular Research Group, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Reus, Spain
| | - Alba Bosquet
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Daiana Ibarretxe
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Lluís Masana
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| |
Collapse
|
35
|
Wang Q, Shi G, Teng Y, Li X, Xie J, Shen Q, Zhang C, Ni S, Tang Z. Successful reduction of inflammatory responses and arachidonic acid-cyclooxygenase 2 pathway in human pulmonary artery endothelial cells by silencing adipocyte fatty acid-binding protein. JOURNAL OF INFLAMMATION-LONDON 2017; 14:8. [PMID: 28331434 PMCID: PMC5359915 DOI: 10.1186/s12950-017-0155-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/16/2017] [Indexed: 01/12/2023]
Abstract
Background Adipocyte fatty acid-binding protein, also known as aP2 or fatty acid-binding protein 4 (FABP4), plays an important role in inflammatory and metabolic responses in adipocytes and macrophages. Recent work has demonstrated that macrophage FABP4 integrates inflammatory and lipid metabolic responses, thereby contributing to the development of insulin resistance and atherosclerosis. However, it is not known whether FABP4 in human pulmonary artery endothelial cells(HPAECs) modulates inflammation. Results Here, we demonstrate that FABP4 and inflammatory cytokines are upregulated in lipopolysaccharide(LPS)-stimulated HPAECs. In addition, LPS increases the expression of molecules in the arachidonic acid(AA)–cyclooxygenase (COX) 2 signaling pathway in FABP4-expressing, but not FABP4-deficient, HPAECs. Conclusions Our findings demonstrate that silencing FABP4 could decrease inflammatory cytokines, which were reported to be expressed via the AA–COX2 pathway, in HPAECs. In addition, silencing FABP4 could inhibit the expression of molecules in the AA–COX2 pathways. So we speculate silencing FABP4 could decrease the inflammatory response in HPAECs, which involves in the AA–COX2 signaling pathway. Our study suggests that FABP4 could be a potential biomarker and intervention point for the inflammation-related disease in HPAECs such as pulmonary thromboembolism.
Collapse
Affiliation(s)
- Qian Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu China
| | - Guanglin Shi
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu China
| | - Ying Teng
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu China
| | - Xia Li
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu China
| | - Jin Xie
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu China
| | - Qin Shen
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu China
| | - Caixin Zhang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu China
| | - Songshi Ni
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu China
| | - Zhiyuan Tang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu China
| |
Collapse
|
36
|
Hu X, Ma X, Luo Y, Xu Y, Xiong Q, Pan X, Bao Y, Jia W. Contribution of serum adipocyte fatty acid-binding protein levels to the presence of microalbuminuria in a Chinese hyperglycemic population. J Diabetes Investig 2017; 8:582-589. [PMID: 27990767 PMCID: PMC5497028 DOI: 10.1111/jdi.12611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/30/2016] [Accepted: 12/11/2016] [Indexed: 01/04/2023] Open
Abstract
AIMS/INTRODUCTION Individuals with type 2 diabetes mellitus are vulnerable to micro- and macrovascular complications in the presence of microalbuminuria. Adipocyte fatty acid-binding protein (A-FABP) was proposed as an indicator for albuminuria in patients with diabetes. The present study aimed to explore the associations between serum A-FABP levels and microalbuminuria in the hyperglycemic population. MATERIALS AND METHODS Serum A-FABP levels were detected using sandwich enzyme-linked immunosorbent assay. Microalbuminuria was identified by urinary albumin-to-creatinine ratio (UACR), when the value was between 30-300 mg/g. The participants were divided into the subgroups based on sex and the status of impaired glucose regulation or newly diagnosed type 2 diabetes mellitus. RESULTS A total of 939 participants, consisting of 436 men and 503 women, were enrolled. Serum levels of A-FABP were much higher in participants with microalbuminuria than those without microalbuminuria. This result held true for all subgroups (all P < 0.05). For Spearman's correlation analyses, serum A-FABP levels showed a positive relationship with the UACR in men and women (both P < 0.01). Multiple stepwise regression analysis showed that serum A-FABP levels were independently and positively correlated with UACR in both sexes (men: standardized β = 0.256, P < 0.001; women: standardized β = 0.155, P = 0.001). This relationship remained significant in every subgroup (all P < 0.01). CONCLUSIONS For hyperglycemic individuals, serum A-FABP levels increased in the presence of microalbuminuria. Serum A-FABP levels were identified as an independent factor positively associated with the UACR.
Collapse
Affiliation(s)
- Xiang Hu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Clinical Center for Diabetes, Shanghai, China.,Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China.,Shanghai Diabetes Institute, Shanghai, China.,Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Xiaojing Ma
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Clinical Center for Diabetes, Shanghai, China.,Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China.,Shanghai Diabetes Institute, Shanghai, China.,Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Yuqi Luo
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Clinical Center for Diabetes, Shanghai, China.,Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China.,Shanghai Diabetes Institute, Shanghai, China.,Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Yiting Xu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Clinical Center for Diabetes, Shanghai, China.,Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China.,Shanghai Diabetes Institute, Shanghai, China.,Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Qin Xiong
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Clinical Center for Diabetes, Shanghai, China.,Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China.,Shanghai Diabetes Institute, Shanghai, China.,Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Xiaoping Pan
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Clinical Center for Diabetes, Shanghai, China.,Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China.,Shanghai Diabetes Institute, Shanghai, China.,Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Yuqian Bao
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Clinical Center for Diabetes, Shanghai, China.,Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China.,Shanghai Diabetes Institute, Shanghai, China.,Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Clinical Center for Diabetes, Shanghai, China.,Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China.,Shanghai Diabetes Institute, Shanghai, China.,Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| |
Collapse
|
37
|
Ochoa-Martínez ÁC, Ruíz-Vera T, Pruneda-Álvarez LG, González-Palomo AK, Almendarez-Reyna CI, Pérez-Vázquez FJ, Pérez-Maldonado IN. Serum adipocyte-fatty acid binding protein (FABP4) levels in women from Mexico exposed to polycyclic aromatic hydrocarbons (PAHs). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:1862-1870. [PMID: 27796996 DOI: 10.1007/s11356-016-7971-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 10/24/2016] [Indexed: 06/06/2023]
Abstract
Recent studies indicate that exposure to polycyclic aromatic hydrocarbons (PAHs) is a very important risk factor for the development of cardiovascular diseases (CVDs). Correspondingly, adipocyte-fatty acid binding protein (FABP4, also known as aP2 and AFABP) has been proposed as a new, meaningful and useful biomarker to predict metabolic and cardiovascular diseases. Therefore, the aim of this study was to evaluate serum FABP4 levels in Mexican women exposed to PAHs. Urinary 1-hydroxypyrene ((1-OHP), exposure biomarker for PAHs) levels were quantified using a high-performance liquid chromatography (HPLC) technique, and serum FABP4 concentrations were analyzed using a commercially available ELISA kit. The mean urinary 1-OHP level found in women participating in this study was 1.30 ± 1.10 μmol/mol creatinine (2.45 ± 2.10 μg/g creatinine). Regarding serum FABP4 concentrations, the levels ranged from 3.80 to 62.5 ng/mL in the assessed population. Moreover, a significant association (p < 0.001) was found between urinary 1-OHP levels and serum FABP4 concentrations in women after adjusting for potential confounding variables. The presented data in this study can be considered only as a starting point for further studies. Then, in order to elucidate whether FABP4 represents a risk factor for CVD disease in humans exposed to air contaminants (such as PAHs), large epidemiological studies are necessary.
Collapse
Affiliation(s)
- Ángeles C Ochoa-Martínez
- Laboratorio de Toxicología Molecular, Centro de Investigación Aplicada en Ambiente y Salud (CIAAS), Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Tania Ruíz-Vera
- Laboratorio de Toxicología Molecular, Centro de Investigación Aplicada en Ambiente y Salud (CIAAS), Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Lucia G Pruneda-Álvarez
- Laboratorio de Toxicología Molecular, Centro de Investigación Aplicada en Ambiente y Salud (CIAAS), Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Ana K González-Palomo
- Laboratorio de Toxicología Molecular, Centro de Investigación Aplicada en Ambiente y Salud (CIAAS), Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Claudia I Almendarez-Reyna
- Laboratorio de Toxicología Molecular, Centro de Investigación Aplicada en Ambiente y Salud (CIAAS), Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Francisco J Pérez-Vázquez
- Laboratorio de Toxicología Molecular, Centro de Investigación Aplicada en Ambiente y Salud (CIAAS), Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Iván N Pérez-Maldonado
- Laboratorio de Toxicología Molecular, Centro de Investigación Aplicada en Ambiente y Salud (CIAAS), Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
- Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
- Unidad Académica Multidisciplinaria Zona Media, Universidad Autónoma de San Luis Potosí, Rio-verde, San Luis Potosí, Mexico.
- , Avenida Sierra Leona No. 550, Colonia Lomas Segunda Sección, 78210, San Luis Potosí, SLP, Mexico.
| |
Collapse
|
38
|
Multiple Sclerosis and Obesity: Possible Roles of Adipokines. Mediators Inflamm 2016; 2016:4036232. [PMID: 27721574 PMCID: PMC5046034 DOI: 10.1155/2016/4036232] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/22/2016] [Accepted: 08/08/2016] [Indexed: 12/21/2022] Open
Abstract
Multiple Sclerosis (MS) is an autoimmune disorder of the Central Nervous System that has been associated with several environmental factors, such as diet and obesity. The possible link between MS and obesity has become more interesting in recent years since the discovery of the remarkable properties of adipose tissue. Once MS is initiated, obesity can contribute to increased disease severity by negatively influencing disease progress and treatment response, but, also, obesity in early life is highly relevant as a susceptibility factor and causally related risk for late MS development. The aim of this review was to discuss recent evidence about the link between obesity, as a chronic inflammatory state, and the pathogenesis of MS as a chronic autoimmune and inflammatory disease. First, we describe the main cells involved in MS pathogenesis, both from neural tissue and from the immune system, and including a new participant, the adipocyte, focusing on their roles in MS. Second, we concentrate on the role of several adipokines that are able to participate in the mediation of the immune response in MS and on the possible cross talk between the latter. Finally, we explore recent therapy that involves the transplantation of adipocyte precursor cells for the treatment of MS.
Collapse
|
39
|
Interaction between gut microbiota and toll-like receptor: from immunity to metabolism. J Mol Med (Berl) 2016; 95:13-20. [PMID: 27639584 PMCID: PMC5225216 DOI: 10.1007/s00109-016-1474-4] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/15/2016] [Accepted: 09/08/2016] [Indexed: 02/06/2023]
Abstract
The human gut contains trillions of commensal bacteria, and similar to pathogenic bacteria, the gut microbes and their products can be recognized by toll-like receptors (TLRs). It is well acknowledged that the interaction between gut microbiota and the local TLRs help to maintain the homeostasis of intestinal immunity. High-fat intake or obesity can weaken gut integrity leading to the penetration of gut microbiota or their bacterial products into the circulation, leading to the activation of TLRs on immune cells and subsequently low-grade systemic inflammation in host. Metabolic cells including hepatocytes and adipocytes also express TLRs. Although they are able to produce and secrete inflammatory molecules, the effectiveness remains low compared with the immune cells embedded in the liver and adipose tissue. The interaction of TLRs in these metabolic cells or organs with gut microbiota remains unclear, but a few studies have suggested that the functions of these TLRs are related to metabolism. Alteration of the gut microbiota is associated with body weight change and adiposity in human, and the interaction between the commensal gut microbiota and TLRs may possibly involve both metabolic and immunological regulation. In this review, we will summarize the current findings on the relationship between TLRs and gut microbiota with a focus on metabolic regulation and discuss how such interaction participates in host metabolism.
Collapse
|
40
|
Pei H, Xie C, Liu Y, Shao M, Chen J, Li D, Ma L, Chen L. Therapeutic potential of a synthetic FABP4 inhibitor 8g on atherosclerosis in ApoE-deficient mice: the inhibition of lipid accumulation and inflammation. RSC Adv 2016. [DOI: 10.1039/c6ra05637g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We discovered a synthetic FABP4 inhibitor that ameliorated the symptoms of atherosclerosis and suppressed lipid accumulation.
Collapse
Affiliation(s)
- Heying Pei
- State Key Laboratory of Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- Chengdu
| | - Caifeng Xie
- Institute of Translational Medicine
- Nanchang University
- Nanchang
- P. R. China
| | - Yibin Liu
- State Key Laboratory of Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- Chengdu
| | - Mingfeng Shao
- State Key Laboratory of Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- Chengdu
| | - Jinying Chen
- State Key Laboratory of Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- Chengdu
| | - Dan Li
- State Key Laboratory of Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- Chengdu
| | - Liang Ma
- Division of Nephrology
- Kidney Research Institute
- West China Hospital
- West China Medical School
- Sichuan University
| | - Lijuan Chen
- State Key Laboratory of Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- Chengdu
| |
Collapse
|
41
|
Goulopoulou S, McCarthy CG, Webb RC. Toll-like Receptors in the Vascular System: Sensing the Dangers Within. Pharmacol Rev 2016; 68:142-67. [PMID: 26721702 PMCID: PMC4709508 DOI: 10.1124/pr.114.010090] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Toll-like receptors (TLRs) are components of the innate immune system that respond to exogenous infectious ligands (pathogen-associated molecular patterns, PAMPs) and endogenous molecules that are released during host tissue injury/death (damage-associated molecular patterns, DAMPs). Interaction of TLRs with their ligands leads to activation of downstream signaling pathways that induce an immune response by producing inflammatory cytokines, type I interferons (IFN), and other inflammatory mediators. TLR activation affects vascular function and remodeling, and these molecular events prime antigen-specific adaptive immune responses. Despite the presence of TLRs in vascular cells, the exact mechanisms whereby TLR signaling affects the function of vascular tissues are largely unknown. Cardiovascular diseases are considered chronic inflammatory conditions, and accumulating data show that TLRs and the innate immune system play a determinant role in the initiation and development of cardiovascular diseases. This evidence unfolds a possibility that targeting TLRs and the innate immune system may be a novel therapeutic goal for these conditions. TLR inhibitors and agonists are already in clinical trials for inflammatory conditions such as asthma, cancer, and autoimmune diseases, but their study in the context of cardiovascular diseases is in its infancy. In this article, we review the current knowledge of TLR signaling in the cardiovascular system with an emphasis on atherosclerosis, hypertension, and cerebrovascular injury. Furthermore, we address the therapeutic potential of TLR as pharmacological targets in cardiovascular disease and consider intriguing research questions for future study.
Collapse
Affiliation(s)
- Styliani Goulopoulou
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| | - Cameron G McCarthy
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| | - R Clinton Webb
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| |
Collapse
|
42
|
Luo H, Wang J, Qiao C, Ma N, Liu D, Zhang W. Pycnogenol attenuates atherosclerosis by regulating lipid metabolism through the TLR4-NF-κB pathway. Exp Mol Med 2015; 47:e191. [PMID: 26492950 PMCID: PMC4673476 DOI: 10.1038/emm.2015.74] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/10/2015] [Accepted: 06/16/2015] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is a leading cause of death worldwide and is characterized by lipid-laden foam cell formation. Recently, pycnogenol (PYC) has drawn much attention because of its prominent effect on cardiovascular disease (CVD). However, its protective effect against atherosclerosis and the underlying mechanism remains undefined. Here PYC treatment reduced areas of plaque and lipid deposition in atherosclerotic mice, concomitant with decreases in total cholesterol and triglyceride levels and increases in HDL cholesterol levels, indicating a potential antiatherosclerotic effect of PYC through the regulation of lipid levels. Additionally, PYC preconditioning markedly decreased foam cell formation and lipid accumulation in lipopolysaccharide (LPS)-stimulated human THP-1 monocytes. A mechanistic analysis indicated that PYC decreased the lipid-related protein expression of adipose differentiation-related protein (ADRP) and adipocyte lipid-binding protein (ALBP/aP2) in a dose-dependent manner. Further analysis confirmed that PYC attenuated LPS-induced lipid droplet formation via ADRP and ALBP expression through the Toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) pathway, because pretreatment with anti-TLR4 antibody or a specific inhibitor of NF-κB (PDTC) strikingly mitigated the LPS-induced increase in ADRP and ALBP. Together, our results provide insight into the ability of PYC to attenuate bacterial infection-triggered pathological processes associated with atherosclerosis. Thus PYC may be a potential lead compound for the future development of antiatherosclerotic CVD therapy.
Collapse
Affiliation(s)
- Hong Luo
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Jing Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Chenhui Qiao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Ning Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Donghai Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Weihua Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Henan, China
| |
Collapse
|
43
|
Hu B, Guo Y, Garbacz WG, Jiang M, Xu M, Huang H, Tsung A, Billiar TR, Ramakrishnan SK, Shah YM, Lam KSL, Huang M, Xie W. Fatty acid binding protein-4 (FABP4) is a hypoxia inducible gene that sensitizes mice to liver ischemia/reperfusion injury. J Hepatol 2015; 63:855-62. [PMID: 26070408 PMCID: PMC4867094 DOI: 10.1016/j.jhep.2015.05.030] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 04/28/2015] [Accepted: 05/21/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Fatty acid binding protein 4 (FABP4) has been known as a mediator of inflammatory response in the macrophages and adipose tissue, but its hepatic function is poorly understood. The goal of this study is to investigate the role of FABP4 in liver ischemia/reperfusion (I/R), a clinical condition that involves both hypoxia and inflammation. METHODS To examine the I/R regulation of FABP4, mice were subjected to I/R surgery before being measured for FABP4 gene expression. Both loss-of-function (by using a pharmacological FABP4 inhibitor) and gain-of-function (by adenoviral overexpression of FABP4) were used to determine the functional relevance of FABP4 expression and its regulation during I/R. To determine the hypoxia responsive regulation of FABP4, primary mouse hepatocytes were exposed to hypoxia. The FABP4 gene promoter was cloned and its regulation by hypoxia inducible factor 1α (HIF-1α) was characterized by luciferase reporter gene, electrophoretic mobility shift, and chromatin immunoprecipitation assays. RESULTS We found that the hepatic expression of FABP4 was markedly induced by I/R. At the functional level, pharmacological inhibition of FABP4 alleviated the I/R injury, whereas adenoviral overexpression of FABP4 sensitized mice to I/R injury. We also showed that exposure of primary hepatocytes to hypoxia or transgenic overexpression of HIF-1α in the mouse liver was sufficient to induce the expression of FABP4. Our promoter analysis established FABP4 as a novel transcriptional target of HIF-1α. CONCLUSIONS FABP4 is a hypoxia inducible gene that sensitizes mice to liver I/R injury. FABP4 may represent a novel therapeutic target, and FABP4 inhibitors may be used as therapeutic agents to manage hepatic I/R injury.
Collapse
Affiliation(s)
- Bingfang Hu
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China; Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yan Guo
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wojciech G Garbacz
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mengxi Jiang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Meishu Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hai Huang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sadeesh K Ramakrishnan
- Department of Molecular & Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yatrik M Shah
- Department of Molecular & Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Karen S L Lam
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Min Huang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China.
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
44
|
Furuhashi M, Saitoh S, Shimamoto K, Miura T. Fatty Acid-Binding Protein 4 (FABP4): Pathophysiological Insights and Potent Clinical Biomarker of Metabolic and Cardiovascular Diseases. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2015; 8:23-33. [PMID: 25674026 PMCID: PMC4315049 DOI: 10.4137/cmc.s17067] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/16/2014] [Accepted: 12/16/2014] [Indexed: 12/13/2022]
Abstract
Over the past decade, evidences of an integration of metabolic and inflammatory pathways, referred to as metaflammation in several aspects of metabolic syndrome, have been accumulating. Fatty acid-binding protein 4 (FABP4), also known as adipocyte FABP (A-FABP) or aP2, is mainly expressed in adipocytes and macrophages and plays an important role in the development of insulin resistance and atherosclerosis in relation to metaflammation. Despite lack of a typical secretory signal peptide, FABP4 has been shown to be released from adipocytes in a non-classical pathway associated with lipolysis, possibly acting as an adipokine. Elevation of circulating FABP4 levels is associated with obesity, insulin resistance, diabetes mellitus, hypertension, cardiac dysfunction, atherosclerosis, and cardiovascular events. Furthermore, ectopic expression and function of FABP4 in several types of cells and tissues have been recently demonstrated. Here, we discuss both the significant role of FABP4 in pathophysiological insights and its usefulness as a biomarker of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shigeyuki Saitoh
- Department of Nursing, Division of Medical and Behavioral Subjects, Sapporo Medical University School of Health Sciences, Sapporo, Japan
| | | | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
45
|
Hao Y, Ma X, Luo Y, Shen Y, Dou J, Pan X, Bao Y, Jia W. Serum adipocyte fatty acid binding protein levels are positively associated with subclinical atherosclerosis in Chinese pre- and postmenopausal women with normal glucose tolerance. J Clin Endocrinol Metab 2014; 99:4321-7. [PMID: 25127012 DOI: 10.1210/jc.2014-1832] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
CONTEXT Recent studies highlight a critical interaction between adipocyte fatty acid binding protein (A-FABP) and cardiovascular disorders. However, associations of A-FABP with subclinical atherosclerosis in a population with normal glucose tolerance remain unknown. OBJECTIVE The study aimed to evaluate the relationship between A-FABP and carotid intima-media thickness (C-IMT) in a Chinese population with normal glucose tolerance. DESIGN AND SETTING A cross-sectional analysis was conducted of 2253 cardiovascular disease-free normal glucose tolerance subjects (835 men, 1418 women; 20-78 years old) from the Shanghai Obesity Study. MAIN OUTCOME AND MEASURES C-IMT was measured by B-mode ultrasound and used to assess subclinical atherosclerosis. Serum A-FABP levels were quantified by a sandwich ELISA. RESULTS The median serum level for A-FABP was 4.0 ng/mL (interquartile range: 2.6-6.0 ng/mL), and significantly higher in women than men (P < .001). After adjusting for age and body mass index (BMI), a partial correlation analysis showed that A-FABP levels correlated with C-IMT in men, premenopausal, and postmenopausal women (P = .024, .006, and .016, respectively). Furthermore, C-IMT increased along with quartile A-FABP values (all P for trend <.001). Regression analyses demonstrated that A-FABP was associated with C-IMT only in women (P = .044 and .001 for pre- and postmenopausal, respectively). Moreover, A-FABP was identified as a risk factor for C-IMT in pre- and postmenopausal women with a normal BMI (P < .001 and P = .012, respectively). CONCLUSIONS Serum A-FABP levels independently and positively correlate with subclinical atherosclerosis in pre- and postmenopausal Chinese women with normal glucose tolerance after adjustments for the traditional risk factors.
Collapse
Affiliation(s)
- Yaping Hao
- Shanghai Key Laboratory of Diabetes Mellitus; Department of Endocrinology and Metabolism; Shanghai Clinical Center for Diabetes; Shanghai Key Clinical Center for Metabolic Disease; Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | | | | | | | | | | | | | | |
Collapse
|
46
|
NADPH oxidases: an overview from structure to innate immunity-associated pathologies. Cell Mol Immunol 2014; 12:5-23. [PMID: 25263488 DOI: 10.1038/cmi.2014.89] [Citation(s) in RCA: 677] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 08/18/2014] [Accepted: 08/18/2014] [Indexed: 12/11/2022] Open
Abstract
Oxygen-derived free radicals, collectively termed reactive oxygen species (ROS), play important roles in immunity, cell growth, and cell signaling. In excess, however, ROS are lethal to cells, and the overproduction of these molecules leads to a myriad of devastating diseases. The key producers of ROS in many cells are the NOX family of NADPH oxidases, of which there are seven members, with various tissue distributions and activation mechanisms. NADPH oxidase is a multisubunit enzyme comprising membrane and cytosolic components, which actively communicate during the host responses to a wide variety of stimuli, including viral and bacterial infections. This enzymatic complex has been implicated in many functions ranging from host defense to cellular signaling and the regulation of gene expression. NOX deficiency might lead to immunosuppression, while the intracellular accumulation of ROS results in the inhibition of viral propagation and apoptosis. However, excess ROS production causes cellular stress, leading to various lethal diseases, including autoimmune diseases and cancer. During the later stages of injury, NOX promotes tissue repair through the induction of angiogenesis and cell proliferation. Therefore, a complete understanding of the function of NOX is important to direct the role of this enzyme towards host defense and tissue repair or increase resistance to stress in a timely and disease-specific manner.
Collapse
|
47
|
Meana C, Peña L, Lordén G, Esquinas E, Guijas C, Valdearcos M, Balsinde J, Balboa MA. Lipin-1 integrates lipid synthesis with proinflammatory responses during TLR activation in macrophages. THE JOURNAL OF IMMUNOLOGY 2014; 193:4614-22. [PMID: 25252959 DOI: 10.4049/jimmunol.1400238] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Lipin-1 is a Mg(2+)-dependent phosphatidic acid phosphatase involved in the de novo synthesis of phospholipids and triglycerides. Using macrophages from lipin-1-deficient animals and human macrophages deficient in the enzyme, we show in this work that this phosphatase acts as a proinflammatory mediator during TLR signaling and during the development of in vivo inflammatory processes. After TLR4 stimulation lipin-1-deficient macrophages showed a decreased production of diacylglycerol and activation of MAPKs and AP-1. Consequently, the generation of proinflammatory cytokines like IL-6, IL-12, IL-23, or enzymes like inducible NO synthase and cyclooxygenase 2, was reduced. In addition, animals lacking lipin-1 had a faster recovery from endotoxin administration concomitant with a reduced production of harmful molecules in spleen and liver. These findings demonstrate an unanticipated role for lipin-1 as a mediator of macrophage proinflammatory activation and support a critical link between lipid biosynthesis and systemic inflammatory responses.
Collapse
Affiliation(s)
- Clara Meana
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, 47003 Valladolid, Spain; and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Lucía Peña
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, 47003 Valladolid, Spain; and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Gema Lordén
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, 47003 Valladolid, Spain; and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Esperanza Esquinas
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, 47003 Valladolid, Spain; and
| | - Carlos Guijas
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, 47003 Valladolid, Spain; and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Martín Valdearcos
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, 47003 Valladolid, Spain; and
| | - Jesús Balsinde
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, 47003 Valladolid, Spain; and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - María A Balboa
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid, 47003 Valladolid, Spain; and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| |
Collapse
|
48
|
Wu G, Li H, Zhou M, Fang Q, Bao Y, Xu A, Jia W. Mechanism and clinical evidence of lipocalin-2 and adipocyte fatty acid-binding protein linking obesity and atherosclerosis. Diabetes Metab Res Rev 2014; 30:447-56. [PMID: 24214285 DOI: 10.1002/dmrr.2493] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 10/29/2013] [Indexed: 12/19/2022]
Abstract
Obesity is considered to be a chronic inflammatory state in which the dysfunction of adipose tissue plays a central role. The adipokines, which are cytokines secreted by adipose tissue, are key links between obesity and related diseases such as metabolic syndrome and atherosclerosis. LCN2 and A-FABP, both of which are major adipokines predominantly produced in adipose tissue, have recently been shown to be pivotal modulators of vascular function. However, different adipokines modulate the development of atherosclerosis in distinctive manners, which are partly attributable to their unique regulatory mechanisms and functions. This review highlights recent advances in the understanding of the role of two adipokines in mediating chronic inflammation and the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Guangyu Wu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, China; Department of Medicine, Medical School of Soochow University, Suzhou, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Effect of toll-like receptor agonists on the formation of macrophage/foam cells upon acute peritonitis in mice. Bull Exp Biol Med 2014; 156:49-52. [PMID: 24319727 DOI: 10.1007/s10517-013-2275-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We studied effects of zymosan, double-stranded RNA, LPS of E. coli and bacterial CpG DNA, agonists of toll-like receptor TLR2, TLR3, TLR4 and TLR9, respectively, on the formation of macrophage/foam cells 24 h after induction of acute peritonitis. Administration of agonists led to transformation of peritoneal macrophages into foam cells and significant activation of cell biosynthesis and increased the content of triglycerides and cholesterol esters in the absence of LDL and irrespective of the capacity of TLR agonists to stimulate neutrophil infiltration and TNF-α production in the peritoneal cavity.
Collapse
|
50
|
Kajimoto K, Minami Y, Harashima H. Cytoprotective role of the fatty acid binding protein 4 against oxidative and endoplasmic reticulum stress in 3T3-L1 adipocytes. FEBS Open Bio 2014; 4:602-10. [PMID: 25161868 PMCID: PMC4141204 DOI: 10.1016/j.fob.2014.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 06/20/2014] [Accepted: 06/30/2014] [Indexed: 01/27/2023] Open
Abstract
Oxidative stress in 3T3-L1 adipocytes was elevated by silencing of FABP4. FABP4 silencing did not alter levels of glutathione or superoxide dismutase. The recombinant FABP4 significantly reduced levels of hydrogen peroxide. The resistance of adipocytes to oxidative stress was decreased by FABP4 knockdown. Silencing of FABP4 elevated the endoplasmic reticulum stress in adipocytes.
The fatty acid binding protein 4 (FABP4), one of the most abundant proteins in adipocytes, has been reported to have a proinflammatory function in macrophages. However, the physiological role of FABP4, which is constitutively expressed in adipocytes, has not been fully elucidated. Previously, we demonstrated that FABP4 was involved in the regulation of interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF) production in 3T3-L1 adipocytes. In this study, we examined the effects of FABP4 silencing on the oxidative and endoplasmic reticulum (ER) stress in 3T3-L1 adipocytes. We found that the cellular reactive oxygen species (ROS) and 8-nitro-cyclic GMP levels were significantly elevated in the differentiated 3T3-L1 adipocytes transfected with a small interfering RNA (siRNA) against Fabp4, although the intracellular levels or enzyme activities of antioxidants including reduced glutathione (GSH), superoxide dismutase (SOD) and glutathione S-transferase A4 (GSTA4) were not altered. An in vitro evaluation using the recombinant protein revealed that FABP4 itself functions as a scavenger protein against hydrogen peroxide (H2O2). FABP4-knockdown resulted in a significant lowering of cell viability of 3T3-L1 adipocytes against H2O2 treatment. Moreover, four kinds of markers related to the ER stress response including the endoplasmic reticulum to nucleus signaling 1 (Ern1), the signal sequence receptor α (Ssr1), the ORM1-like 3 (Ormdl3), and the spliced X-box binding protein 1 (Xbp1s), were all elevated as the result of the knockdown of FABP4. Consequently, FABP4 might have a new role as an antioxidant protein against H2O2 and contribute to cytoprotection against oxidative and ER stress in adipocytes.
Collapse
Key Words
- Adipocyte
- Antioxidant
- ER stress
- ER, endoplasmic reticulum
- Ern1, endoplasmic reticulum to nucleus signaling 1
- FABP, fatty acid binding protein
- FABP4
- GSH, reduced glutathione
- GSTA4, glutathione S-transferase A4
- H2O2, hydrogen peroxide
- Ormdl3, ORM1-like 3
- Oxidative stress
- ROS, reactive oxygen species
- SOD, superoxide dismutase
- Ssr1, signal sequence receptor α
- UPR, unfolded protein response
- VEGF, vascular endothelial growth factor
- Xbp1, X-box binding protein 1.
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Kazuaki Kajimoto
- Corresponding author. Tel.: +81 11 706 2197; fax: +81 11 706 4879.
| | | | | |
Collapse
|