1
|
Ren X, Jin C, Li Q, Fu C, Fang Y, Xu Z, Liang Z, Wang T. Fatty acid binding proteins-mediated mitochondrial dysfunction in the development of age-related diseases: A review. Int J Biol Macromol 2025; 309:142913. [PMID: 40203912 DOI: 10.1016/j.ijbiomac.2025.142913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Fatty acid-binding proteins (FABPs) act as lipid chaperones and play a role in the pathological processes of various lipid signaling pathways. Mitochondria are crucial for the regulation of lipid metabolism. As an aging marker, lipid-mediated mitochondrial dysfunction has been observed in the etiology of numerous diseases, including neurodegenerative diseases, metabolic syndromes, cardiovascular diseases, and tumorigenesis. Members of the FABP family have been identified to regulate mitochondrial function. Targeting FABPs specifically may provide a promising approach to improve mitochondrial function and treat age-related diseases. This review summarizes the connection between FABPs and mitochondrial function and highlights certain FABPs involved in age-related diseases that hold significant therapeutic promise.
Collapse
Affiliation(s)
- Xingxing Ren
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Chaoyuan Jin
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Qilin Li
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200125, China
| | - Congyi Fu
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200125, China
| | - Yu Fang
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200125, China
| | - Zihang Xu
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200125, China
| | - Zi Liang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Tianshi Wang
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201207, China.
| |
Collapse
|
2
|
Tawfeeq HR, Lackey AI, Zhou Y, Diolintzi A, Zacharisen SM, Lau YH, Quadro L, Storch J. Tissue-Specific Ablation of Liver Fatty Acid-Binding Protein Induces a Metabolically Healthy Obese Phenotype in Female Mice. Nutrients 2025; 17:753. [PMID: 40077623 PMCID: PMC11901660 DOI: 10.3390/nu17050753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Obesity is associated with numerous metabolic complications including insulin resistance, dyslipidemia, and a reduced capacity for physical activity. Whole-body ablation of liver fatty acid-binding protein (LFABP) in mice was shown to alleviate several of these metabolic complications; high-fat (HF)-fed LFABP knockout (LFABP-/-) mice developed higher fat mass than their wild-type (WT) counterparts but displayed a metabolically healthy obese (MHO) phenotype with normoglycemia, normoinsulinemia, and reduced hepatic steatosis compared with WT. Since LFABP is expressed in both liver and intestine, in the present study, we generated LFABP conditional knockout (cKO) mice to determine the contributions of LFABP specifically within the liver or within the intestine, to the whole-body phenotype of the global knockout. Methods: Female liver-specific LFABP knockout (LFABPliv-/-), intestine-specific LFABP knockout (LFABPint-/-), and "floxed" LFABP (LFABPfl/fl) control mice were fed a 45% Kcal fat semipurified HF diet for 12 weeks. Results: While not as dramatic as found for whole-body LFABP-/- mice, both LFABPliv-/- and LFABPint-/- mice had significantly higher body weights and fat mass compared with LFABPfl/fl control mice. As with the global LFABP nulls, both LFABPliv-/- and LFABPint-/- mice remained normoglycemic and normoinsulinemic. Despite their greater fat mass, the LFABPliv-/- mice did not develop hepatic steatosis. Additionally, LFABPliv-/- and LFABPint-/- mice had higher endurance exercise capacity when compared with LFABPfl/fl control mice. Conclusions: The results suggest, therefore, that either liver-specific or intestine-specific ablation of LFABP in female mice is sufficient to induce, at least in part, the MHO phenotype observed following whole-body ablation of LFABP.
Collapse
Affiliation(s)
- Hiba Radhwan Tawfeeq
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901, USA
- Rutgers Center for Lipid Research, New Brunswick, NJ 08901, USA
| | - Atreju I. Lackey
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901, USA
- Rutgers Center for Lipid Research, New Brunswick, NJ 08901, USA
| | - Yinxiu Zhou
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| | - Anastasia Diolintzi
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901, USA
- Rutgers Center for Lipid Research, New Brunswick, NJ 08901, USA
| | - Sophia M. Zacharisen
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| | - Yin Hei Lau
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| | - Loredana Quadro
- Rutgers Center for Lipid Research, New Brunswick, NJ 08901, USA
- Department of Food Science, Rutgers University, New Brunswick, NJ 07102, USA
| | - Judith Storch
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901, USA
- Rutgers Center for Lipid Research, New Brunswick, NJ 08901, USA
| |
Collapse
|
3
|
Tawfeeq HR, Lackey AI, Zhou Y, Diolointzi A, Zacharisen S, Lau YH, Quadro L, Storch J. Tissue-Specific Ablation of Liver Fatty Acid-Binding Protein Induces a Metabolically Healthy Obese Phenotype in Female Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.02.631082. [PMID: 39803463 PMCID: PMC11722216 DOI: 10.1101/2025.01.02.631082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Background/Objectives Obesity is associated with numerous metabolic complications including insulin resistance, dyslipidemia, and a reduced capacity for physical activity. Whole-body ablation of liver fatty acid-binding protein (LFABP) in mice was shown to alleviate several of these metabolic complications; high fat (HF) fed LFABP knockout (LFABP-/-) mice developed higher fat mass than their wild-type (WT) counterparts but displayed a metabolically healthy obese (MHO) phenotype with normoglycemia, normoinsulinemia, and reduced hepatic steatosis compared with WT. LFABP is expressed in both liver and intestine, thus in the present study, LFABP conditional knockout (cKO) mice were generated to determine the contributions of LFABP specifically within the liver or the intestine to the whole body phenotype of the global knockout. Methods Female liver-specific LFABP knockout (LFABPliv-/-), intestine-specific LFABP knockout (LFABPint-/-), and floxed LFABP (LFABPfl/fl) control mice were fed a 45% Kcal fat semipurified HF diet for 12 weeks. Results While not as dramatic as found for whole-body LFABP-/- mice, both LFABPliv-/- and LFABPint-/- mice had significantly higher body weights and fat mass compared with LFABPfl/fl control mice. As with the global LFABP nulls, both LFABPliv-/- and LFABPint-/- mice remained normoglycemic and normoinsulinemic. Despite their greater fat mass, the LFABPliv-/- mice did not develop hepatic steatosis. Additionally, LFABPliv-/- and LFABPint-/- mice had higher endurance exercise capacity when compared with LFABPfl/fl control mice. Conclusions The results suggest, therefore, that either liver-specific or intestine-specific ablation of LFABP in female mice is sufficient to induce, at least in part, the MHO phenotype observed following whole-body ablation of LFABP.
Collapse
Affiliation(s)
- Hiba Radhwan Tawfeeq
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey
- Rutgers Center for Lipid Research, New Brunswick, New Jersey
| | - Atreju I Lackey
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey
- Rutgers Center for Lipid Research, New Brunswick, New Jersey
| | - Yinxiu Zhou
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey
| | - Anastasia Diolointzi
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey
- Rutgers Center for Lipid Research, New Brunswick, New Jersey
| | - Sophia Zacharisen
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey
| | - Yin Hei Lau
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey
| | - Loredana Quadro
- Rutgers Center for Lipid Research, New Brunswick, New Jersey
- Department of Food Science, Rutgers University, New Brunswick, New Jersey
| | - Judith Storch
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey
- Rutgers Center for Lipid Research, New Brunswick, New Jersey
| |
Collapse
|
4
|
Białek W, Hryniewicz-Jankowska A, Czechowicz P, Sławski J, Collawn JF, Czogalla A, Bartoszewski R. The lipid side of unfolded protein response. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159515. [PMID: 38844203 DOI: 10.1016/j.bbalip.2024.159515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/16/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Although our current knowledge of the molecular crosstalk between the ER stress, the unfolded protein response (UPR), and lipid homeostasis remains limited, there is increasing evidence that dysregulation of either protein or lipid homeostasis profoundly affects the other. Most research regarding UPR signaling in human diseases has focused on the causes and consequences of disrupted protein folding. The UPR itself consists of very complex pathways that function to not only maintain protein homeostasis, but just as importantly, modulate lipid biogenesis to allow the ER to adjust and promote cell survival. Lipid dysregulation is known to activate many aspects of the UPR, but the complexity of this crosstalk remains a major research barrier. ER lipid disequilibrium and lipotoxicity are known to be important contributors to numerous human pathologies, including insulin resistance, liver disease, cardiovascular diseases, neurodegenerative diseases, and cancer. Despite their medical significance and continuous research, however, the molecular mechanisms that modulate lipid synthesis during ER stress conditions, and their impact on cell fate decisions, remain poorly understood. Here we summarize the current view on crosstalk and connections between altered lipid metabolism, ER stress, and the UPR.
Collapse
Affiliation(s)
- Wojciech Białek
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | | | - Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Aleksander Czogalla
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Rafał Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland.
| |
Collapse
|
5
|
Warren WG, Osborn M, Duffy P, Yates A, O'Sullivan SE. Potential safety implications of fatty acid-binding protein inhibition. Toxicol Appl Pharmacol 2024; 491:117079. [PMID: 39218163 DOI: 10.1016/j.taap.2024.117079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/15/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Fatty acid-binding proteins (FABPs) are small intracellular proteins that regulate fatty acid metabolism, transport, and signalling. There are ten known human isoforms, many of which are upregulated and involved in clinical pathologies. As such, FABP inhibition may be beneficial in disease states such as cancer, and those involving the cardiovascular system, metabolism, immunity, and cognition. Recently, a potent, selective FABP5 inhibitor (ART26.12), with 90-fold selectivity to FABP3 and 20-fold selectivity to FABP7, was found to be remarkably benign, with a no-observed-adverse-effect level of 1000 mg/kg in rats and dogs, showing no genotoxicity, cardiovascular, central, or respiratory toxicity. To understand the potential implication of FABP inhibition more fully, this review systematically assessed literature investigating genetic knockout, knockdown, and pharmacological inhibition of FABP3, FABP4, FABP5, or FABP7. Analysis of the literature revealed that animals bred not to express FABPs showed the most biological effects, suggesting key roles of these proteins during development. FABP ablation sometimes exacerbated symptoms of disease models, particularly those linked to metabolism, inflammatory and immune responses, cardiac contractility, neurogenesis, and cognition. However, FABP inhibition (genetic silencing or pharmacological) had a positive effect in many more disease conditions. Several polymorphisms of each FABP gene have also been linked to pathological conditions, but it was unclear how several polymorphisms affected protein function. Overall, analysis of the literature to date suggests that pharmacological inhibition of FABPs in adults is of low risk.
Collapse
Affiliation(s)
- William G Warren
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom.
| | - Myles Osborn
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom
| | - Paul Duffy
- Apconix Ltd., Alderley Park, Cheshire SK10 4TG, United Kingdom
| | - Andrew Yates
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom
| | | |
Collapse
|
6
|
Ohguro H, Watanabe M, Hikage F, Sato T, Nishikiori N, Umetsu A, Higashide M, Ogawa T, Furuhashi M. Fatty Acid-Binding Protein 4-Mediated Regulation Is Pivotally Involved in Retinal Pathophysiology: A Review. Int J Mol Sci 2024; 25:7717. [PMID: 39062961 PMCID: PMC11277531 DOI: 10.3390/ijms25147717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Fatty acid-binding proteins (FABPs), a family of lipid chaperone molecules that are involved in intracellular lipid transportation to specific cellular compartments, stimulate lipid-associated responses such as biological signaling, membrane synthesis, transcriptional regulation, and lipid synthesis. Previous studies have shown that FABP4, a member of this family of proteins that are expressed in adipocytes and macrophages, plays pivotal roles in the pathogenesis of various cardiovascular and metabolic diseases, including diabetes mellitus (DM) and hypertension (HT). Since significant increases in the serum levels of FABP4 were detected in those patients, FABP4 has been identified as a crucial biomarker for these systemic diseases. In addition, in the field of ophthalmology, our group found that intraocular levels of FABP4 (ioFABP4) and free fatty acids (ioFFA) were substantially elevated in patients with retinal vascular diseases (RVDs) including proliferative diabetic retinopathy (PDR) and retinal vein occlusion (RVO), for which DM and HT are also recognized as significant risk factors. Recent studies have also revealed that ioFABP4 plays important roles in both retinal physiology and pathogenesis, and the results of these studies have suggested potential molecular targets for retinal diseases that might lead to future new therapeutic strategies.
Collapse
Affiliation(s)
- Hiroshi Ohguro
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Megumi Watanabe
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Fumihito Hikage
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Nami Nishikiori
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Araya Umetsu
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Megumi Higashide
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Toshifumi Ogawa
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.)
| |
Collapse
|
7
|
HAN M, YI X, YOU S, WU X, WANG S, HE D. Gehua Jiejiu Dizhi decoction ameliorates alcoholic fatty liver in mice by regulating lipid and bile acid metabolism and with exertion of antioxidant stress based on 4DLabel-free quantitative proteomic study. J TRADIT CHIN MED 2024; 44:277-288. [PMID: 38504534 PMCID: PMC10927405 DOI: 10.19852/j.cnki.jtcm.20231018.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 04/27/2023] [Indexed: 03/21/2024]
Abstract
OBJECTIVE To analyze the effect and molecular mechanism of Gehua Jiejiu Dizhi decoction (, GJDD) on alcoholic fatty live disease (AFLD) by using proteomic methods. METHODS The male C57BL/6J mouse were randomly divided into four groups: control group, model group, GJDD group and resveratrol group. After the AFLD model was successfully prepared by intragastric administration of alcohol once on the basis of the Lieber-DeCarli classical method, the GJDD group and resveratrol group were intragastrically administered with GJDD (4900 mg/kg) and resveratrol (400 mg/kg) respectively, once a day for 9 d. The fat deposition of liver tissue was observed and evaluated by oil red O (ORO) staining. 4DLabel-free quantitative proteome method was used to determine and quantify the protein expression in liver tissue of each experimental group. The differentially expressed proteins were screened according to protein expression differential multiples, and then analyzed by Gene ontology classification and Kyoto Encyclopedia of Genes and Genomes pathway enrichment. Finally, expression validation of the differentially co-expressed proteins from control group, model group and GJDD group were verified by targeted proteomics quantification techniques. RESULTS In semiquantitative analyses of ORO, all kinds of steatosis (ToS, MaS, and MiS) were evaluated higher in AFLD mice compared to those in GJDD or resveratrol-treated mice. 4DLabel-free proteomics analysis results showed that a total of 4513 proteins were identified, of which 3763 proteins were quantified and 946 differentially expressed proteins were screened. Compared with the control group, 145 proteins were up-regulated and 148 proteins were down-regulated in the liver tissue of model group. In addition, compared with the model group, 92 proteins were up-regulated and 135 proteins were down-regulated in the liver tissue of the GJDD group. 15 differentially co-expressed proteins were found between every two groups (model group vs control group, GJDD group vs model group and GJDD group vs control group), which were involved in many biological processes. Among them, 11 differentially co-expressed key proteins (Aox3, H1-5, Fabp5, Ces3a, Nudt7, Serpinb1a, Fkbp11, Rpl22l1, Keg1, Acss2 and Slco1a1) were further identified by targeted proteomic quantitative technology and their expression patterns were consistent with the results of 4D label-free proteomic analysis. CONCLUSIONS Our study provided proteomics-based evidence that GJDD alleviated AFLD by modulating liver protein expression, likely through the modulation of lipid metabolism, bile acid metabolism and with exertion of antioxidant stress.
Collapse
Affiliation(s)
- Min HAN
- 1 Guizhou University of Traditional Chinese Medicine, Graduate School, Guiyang 550025, China
| | - Xu YI
- 2 Department of Clinical medical laboratory, Department of Gastroenterology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Shaowei YOU
- 2 Department of Clinical medical laboratory, Department of Gastroenterology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Xueli WU
- 2 Department of Clinical medical laboratory, Department of Gastroenterology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Shuoshi WANG
- 2 Department of Clinical medical laboratory, Department of Gastroenterology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Diancheng HE
- 2 Department of Clinical medical laboratory, Department of Gastroenterology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| |
Collapse
|
8
|
Xu J, Zheng B, Xie C, Zhao Y, Wu H, Wang Y, Guan X, Lei X, Liu D, Lou X, Chen X, Huang Y. Inhibition of FABP5 attenuates inflammatory bowel disease by modulating macrophage alternative activation. Biochem Pharmacol 2024; 219:115974. [PMID: 38081366 DOI: 10.1016/j.bcp.2023.115974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023]
Abstract
Fatty acid binding protein 5 (FABP5) is an intracellular chaperone of fatty acid molecules that regulates lipid metabolism and cell growth. However, its role in intestinal inflammation remains enigmatic. Through examination of human tissue samples and single-cell data, we observed a significant upregulation of FABP5 within the mucosa of patients afflicted with ulcerative colitis (UC) and Crohn's disease (CD), predominantly localized in intestinal macrophages. Herein, we investigate the regulation of FABP5-IN-1, a FABP5 inhibitor, on various cells of the gut in an inflammatory environment. Our investigations confirmed that FABP5 ameliorates DSS-induced colitis in mice by impeding the differentiation of macrophages into M1 macrophages in vitro and in vivo. Furthermore, following FABP5-IN-1 intervention, we observed a notable restoration of intestinal goblet cells and tuft cells, even under inflammatory conditions. Additionally, FABP5-IN-1 exhibits a protective effect against DSS-induced colitis by promoting the polarization of macrophages towards the M2 phenotype in vivo. In summary, FABP5-IN-1 confers protection against DSS-induced acute colitis through a multifaceted approach, encompassing the reduction of inflammatory macrophage infiltration, macrophage polarization, regulating Th17/Treg cells to play an anti-inflammatory role in IBD. The implications for IBD are underscored by the comprehensive in vivo and in vitro experiments presented in this article, thereby positioning FABP5 as a promising and novel therapeutic target for the treatment of IBD.
Collapse
Affiliation(s)
- Jingping Xu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Bolin Zheng
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, PR China
| | - Chunlan Xie
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen 361102, PR China
| | - Yao Zhao
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Hailun Wu
- First Affiliated Hospital, Nanchang University, Nanchang 330000, Jiangxi Province, PR China
| | - Yiting Wang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Xiaoli Guan
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Xintao Lei
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Dexin Liu
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, PR China
| | - Xiaoying Lou
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Xiaohui Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen 361003, PR China.
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China.
| |
Collapse
|
9
|
Gindri dos Santos B, Goedeke L. Macrophage immunometabolism in diabetes-associated atherosclerosis. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00032. [PMID: 37849988 PMCID: PMC10578522 DOI: 10.1097/in9.0000000000000032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/15/2023] [Indexed: 10/19/2023]
Abstract
Macrophages play fundamental roles in atherosclerotic plaque formation, growth, and regression. These cells are extremely plastic and perform different immune functions depending on the stimuli they receive. Initial in vitro studies have identified specific metabolic pathways that are crucial for the proper function of pro-inflammatory and pro-resolving macrophages. However, the plaque microenvironment, especially in the context of insulin resistance and type 2 diabetes, constantly challenges macrophages with several simultaneous inflammatory and metabolic stimuli, which may explain why atherosclerosis is accelerated in diabetic patients. In this mini review, we discuss how macrophage mitochondrial function and metabolism of carbohydrates, lipids, and amino acids may be affected by this complex plaque microenvironment and how risk factors associated with type 2 diabetes alter the metabolic rewiring of macrophages and disease progression. We also briefly discuss current challenges in assessing macrophage metabolism and identify future tools and possible strategies to alter macrophage metabolism to improve treatment options for diabetes-associated atherosclerosis.
Collapse
Affiliation(s)
- Bernardo Gindri dos Santos
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leigh Goedeke
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine (Endocrinology), The Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
10
|
Abstract
Fatty acid-binding proteins (FABPs) are small lipid-binding proteins abundantly expressed in tissues that are highly active in fatty acid (FA) metabolism. Ten mammalian FABPs have been identified, with tissue-specific expression patterns and highly conserved tertiary structures. FABPs were initially studied as intracellular FA transport proteins. Further investigation has demonstrated their participation in lipid metabolism, both directly and via regulation of gene expression, and in signaling within their cells of expression. There is also evidence that they may be secreted and have functional impact via the circulation. It has also been shown that the FABP ligand binding repertoire extends beyond long-chain FAs and that their functional properties also involve participation in systemic metabolism. This article reviews the present understanding of FABP functions and their apparent roles in disease, particularly metabolic and inflammation-related disorders and cancers.
Collapse
Affiliation(s)
- Judith Storch
- Department of Nutritional Sciences and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, United States;
| | - Betina Corsico
- Instituto de Investigaciones Bioquímicas de La Plata, CONICET-UNLP, Facultad de Ciencias Médicas, La Plata, Argentina;
| |
Collapse
|
11
|
Cui CY, Ferrucci L, Gorospe M. Macrophage Involvement in Aging-Associated Skeletal Muscle Regeneration. Cells 2023; 12:1214. [PMID: 37174614 PMCID: PMC10177543 DOI: 10.3390/cells12091214] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
The skeletal muscle is a dynamic organ composed of contractile muscle fibers, connective tissues, blood vessels and nerve endings. Its main function is to provide motility to the body, but it is also deeply involved in systemic metabolism and thermoregulation. The skeletal muscle frequently encounters microinjury or trauma, which is primarily repaired by the coordinated actions of muscle stem cells (satellite cells, SCs), fibro-adipogenic progenitors (FAPs), and multiple immune cells, particularly macrophages. During aging, however, the capacity of skeletal muscle to repair and regenerate declines, likely contributing to sarcopenia, an age-related condition defined as loss of muscle mass and function. Recent studies have shown that resident macrophages in skeletal muscle are highly heterogeneous, and their phenotypes shift during aging, which may exacerbate skeletal muscle deterioration and inefficient regeneration. In this review, we highlight recent insight into the heterogeneity and functional roles of macrophages in skeletal muscle regeneration, particularly as it declines with aging.
Collapse
Affiliation(s)
- Chang-Yi Cui
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
12
|
Liu H, Hallauer Hastings M, Kitchen R, Xiao C, Baldovino Guerra JR, Kuznetsov A, Rosenzweig A. Beneficial Effects of Moderate Hepatic Activin A Expression on Metabolic Pathways, Inflammation, and Atherosclerosis. Arterioscler Thromb Vasc Biol 2023; 43:330-349. [PMID: 36453275 DOI: 10.1161/atvbaha.122.318138] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND Atherosclerosis is an inflammatory vascular disease marked by hyperlipidemia and hematopoietic stem cell expansion. Activin A, a member of the Activin/GDF/TGFβ/BMP (growth/differentiation factor/transforming growth factor beta/bone morphogenetic protein) family is broadly expressed and increases in human atherosclerosis, but its functional effects in vivo in this context remain unclear. METHODS We studied LDLR-/- mice on a Western diet for 12 weeks and used adeno-associated viral vectors with a liver-specific TBG (thyroxine-binding globulin) promoter to express Activin A or GFP (control). Atherosclerotic lesions were analyzed by oil red staining. Blood lipid profiling was performed by high-performance liquid chromatography, and immune cells were evaluated by flow cytometry. Liver RNA-sequencing was performed to explore the underlying mechanisms. RESULTS Activin A expression decreased in both livers and aortae from LDLR-/- mice fed a Western diet compared with standard laboratory diet. Adenoassociated virus-TBG-Activin A increased Activin A hepatic expression ≈10-fold at 12 weeks; P<0.001) and circulating Activin A levels ≈2000 pg/ml versus ≈50 pg/ml; P<0.001, compared with controls). Hepatic Activin A expression decreased plasma total and LDL (low-density lipoprotein) cholesterol ≈60% and ≈40%, respectively), reduced inflammatory cells in aortae and proliferating hematopoietic stem cells in bone marrow, and reduced atherosclerotic lesion and necrotic core area in aortae. Activin A also attenuated liver steatosis and expression of the lipogenesis genes, Srebp1 and Srebp2. RNA sequencing revealed Activin A not only blocked expression of genes involved in hepatic de novo lipogenesis but also fatty acid uptake and liver inflammation. In addition, Activin A expressed in the liver also reduced white fat tissue accumulation, decreased adipocyte size, and improved glucose tolerance. CONCLUSIONS Our studies reveal hepatic Activin A expression reduces inflammation, hematopoietic stem cell expansion, liver steatosis, circulating cholesterol, and fat accumulation, which likely all contribute to the observed protection against atherosclerosis. The reduced Activin A observed in LDLR-/- mice on a Western diet seems maladaptive and deleterious for atherogenesis.
Collapse
Affiliation(s)
- Huan Liu
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | | | - Robert Kitchen
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | - Chunyang Xiao
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | | | - Alexandra Kuznetsov
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | - Anthony Rosenzweig
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| |
Collapse
|
13
|
Hou Y, Wei D, Bossila EA, Zhang Z, Li S, Bao J, Xu H, Zhang L, Zhao Y. FABP5 Deficiency Impaired Macrophage Inflammation by Regulating AMPK/NF-κB Signaling Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2181-2191. [PMID: 36426981 DOI: 10.4049/jimmunol.2200182] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022]
Abstract
Fatty acid binding protein 5 (FABP5) is mainly involved in the uptake, transport, and metabolism of fatty acid in the cytoplasm, and its role in immune cells has been recognized in recent years. However, the role of FABP5 in macrophage inflammation and its underlying mechanisms were not fully addressed. In our study, the acute liver injury and sepsis mouse models were induced by i.p. injection of LPS and cecal contents, respectively. Oleic acid (0.6 g/kg) was injected four times by intragastric administration every week, and this lasted for 1 wk before the LPS or cecal content challenge. We found that myeloid-specific deletion of FABP5 mitigated LPS-induced acute liver injury with reduced mortality of mice, histological liver damage, alanine aminotransferase, and proinflammatory factor levels. Metabolic analysis showed that FABP5 deletion increased the intracellular unsaturated fatty acids, especially oleic acid, in LPS-induced macrophages. The addition of oleic acid also decreased LPS-stimulated macrophage inflammation in vitro and reduced acute liver injury in LPS-induced or cecal content-induced sepsis mice. RNA-sequencing and molecular mechanism studies showed that FABP5 deletion or oleic acid supplementation increased the AMP/ATP ratio and AMP-activated protein kinase (AMPK) activation and inhibited the NF-κB pathway during the inflammatory response to LPS stimulation of macrophages. Inhibiting AMPK activation or expression by chemical or genetic approaches significantly rescued the decreased NF-κB signaling pathway and inflammatory response in LPS-treated FABP5-knockout macrophages. Our present study indicated that inhibiting FABP5 or supplementation of oleic acid might be used for the treatment of sepsis-caused acute liver injury.
Collapse
Affiliation(s)
- Yangxiao Hou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Dong Wei
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Elhusseny A Bossila
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Biotechnology Department, Faculty of Agriculture Al-Azhar University, Cairo, Egypt
| | - Zhaoqi Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Sihong Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiaming Bao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Huawen Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lianfeng Zhang
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; and
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regeneration, Beijing, China
| |
Collapse
|
14
|
FABP5 controls macrophage alternative activation and allergic asthma by selectively programming long-chain unsaturated fatty acid metabolism. Cell Rep 2022; 41:111668. [DOI: 10.1016/j.celrep.2022.111668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/13/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
|
15
|
Krasniewski LK, Chakraborty P, Cui CY, Mazan-Mamczarz K, Dunn C, Piao Y, Fan J, Shi C, Wallace T, Nguyen C, Rathbun IA, Munk R, Tsitsipatis D, De S, Sen P, Ferrucci L, Gorospe M. Single-cell analysis of skeletal muscle macrophages reveals age-associated functional subpopulations. eLife 2022; 11:e77974. [PMID: 36259488 PMCID: PMC9629833 DOI: 10.7554/elife.77974] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 10/10/2022] [Indexed: 11/30/2022] Open
Abstract
Tissue-resident macrophages represent a group of highly responsive innate immune cells that acquire diverse functions by polarizing toward distinct subpopulations. The subpopulations of macrophages that reside in skeletal muscle (SKM) and their changes during aging are poorly characterized. By single-cell transcriptomic analysis with unsupervised clustering, we found 11 distinct macrophage clusters in male mouse SKM with enriched gene expression programs linked to reparative, proinflammatory, phagocytic, proliferative, and senescence-associated functions. Using a complementary classification, membrane markers LYVE1 and MHCII identified four macrophage subgroups: LYVE1-/MHCIIhi (M1-like, classically activated), LYVE1+/MHCIIlo (M2-like, alternatively activated), and two new subgroups, LYVE1+/MHCIIhi and LYVE1-/MHCIIlo. Notably, one new subgroup, LYVE1+/MHCIIhi, had traits of both M2 and M1 macrophages, while the other new subgroup, LYVE1-/MHCIIlo, displayed strong phagocytic capacity. Flow cytometric analysis validated the presence of the four macrophage subgroups in SKM and found that LYVE1- macrophages were more abundant than LYVE1+ macrophages in old SKM. A striking increase in proinflammatory markers (S100a8 and S100a9 mRNAs) and senescence-related markers (Gpnmb and Spp1 mRNAs) was evident in macrophage clusters from older mice. In sum, we have identified dynamically polarized SKM macrophages and propose that specific macrophage subpopulations contribute to the proinflammatory and senescent traits of old SKM.
Collapse
Affiliation(s)
- Linda K Krasniewski
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Papiya Chakraborty
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Chang-Yi Cui
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Krystyna Mazan-Mamczarz
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Christopher Dunn
- Flow Cytometry Core, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Yulan Piao
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Jinshui Fan
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Changyou Shi
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Tonya Wallace
- Flow Cytometry Core, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Cuong Nguyen
- Flow Cytometry Core, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Isabelle A Rathbun
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Dimitrios Tsitsipatis
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Supriyo De
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Payel Sen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of HealthBaltimoreUnited States
| |
Collapse
|
16
|
Basak S, Mallick R, Banerjee A, Pathak S, Duttaroy AK. Cytoplasmic fatty acid-binding proteins in metabolic diseases and cancers. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 132:143-174. [PMID: 36088074 DOI: 10.1016/bs.apcsb.2022.05.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytoplasmic fatty acid-binding proteins (FABPs) are multipurpose proteins that can modulate lipid fluxes, trafficking, signaling, and metabolism. FABPs regulate metabolic and inflammatory pathways, its inhibition can improve type 2 diabetes mellitus and atherosclerosis. In addition, FABPs are involved in obesity, metabolic disease, cardiac dysfunction, and cancers. FABPs are promising tissue biomarkers in solid tumors for diagnostic and/or prognostic targets for novel therapeutic strategies. The signaling responsive elements of FABPs and determinants of FABP-mediated functions may be exploited in preventing or treating these diseases.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
17
|
Zhang T, Shen HH, Qin XY, Li MQ. The metabolic characteristic of decidual immune cells and their unique properties in pregnancy loss. Immunol Rev 2022; 308:168-186. [PMID: 35582842 DOI: 10.1111/imr.13085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022]
Abstract
Maternal tolerance to semi- or fully allograft conceptus is a prerequisite for the maintenance of pregnancy. Once this homeostasis is disrupted, it may result in pregnancy loss. As a potential approach to prevent pregnancy loss, targeting decidual immune cells (DICs) at the maternal-fetal interface has been suggested. Although the phenotypic features and functions of DIC have been extensively profiled, the regulatory pathways for this unique immunological adaption have yet to be elucidated. In recent years, a pivotal mechanism has been highlighted in the area of immunometabolism, by which the changes in intracellular metabolic pathways in DIC and interaction with the adjacent metabolites in the microenvironment can alter their phenotypes and function. More inspiringly, the manipulation of metabolic profiling in DIC provides a novel avenue for the prevention and treatment of pregnancy loss. Herein, this review highlights the major metabolic programs (specifically, glycolysis, ATP-adenosine metabolism, lysophosphatidic acid metabolism, and amino acid metabolism) in multiple immune cells (including decidual NK cells, macrophages, and T cells) and their integrations with the metabolic microenvironment in normal pregnancy. Importantly, this perspective may help to provide a potential therapeutic strategy for reducing pregnancy loss via targeting this interplay.
Collapse
Affiliation(s)
- Tao Zhang
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Hui-Hui Shen
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Xue-Yun Qin
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai, China.,Shanghai Medical School, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Mysore V, Tahir S, Furuhashi K, Arora J, Rosetti F, Cullere X, Yazbeck P, Sekulic M, Lemieux ME, Raychaudhuri S, Horwitz BH, Mayadas TN. Monocytes transition to macrophages within the inflamed vasculature via monocyte CCR2 and endothelial TNFR2. J Exp Med 2022; 219:e20210562. [PMID: 35404389 PMCID: PMC9006314 DOI: 10.1084/jem.20210562] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 11/16/2021] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Monocytes undergo phenotypic and functional changes in response to inflammatory cues, but the molecular signals that drive different monocyte states remain largely undefined. We show that monocytes acquire macrophage markers upon glomerulonephritis and may be derived from CCR2+CX3CR1+ double-positive monocytes, which are preferentially recruited, dwell within glomerular capillaries, and acquire proinflammatory characteristics in the nephritic kidney. Mechanistically, the transition to immature macrophages begins within the vasculature and relies on CCR2 in circulating cells and TNFR2 in parenchymal cells, findings that are recapitulated in vitro with monocytes cocultured with TNF-TNFR2-activated endothelial cells generating CCR2 ligands. Single-cell RNA sequencing of cocultures defines a CCR2-dependent monocyte differentiation path associated with the acquisition of immune effector functions and generation of CCR2 ligands. Immature macrophages are detected in the urine of lupus nephritis patients, and their frequency correlates with clinical disease. In conclusion, CCR2-dependent functional specialization of monocytes into macrophages begins within the TNF-TNFR2-activated vasculature and may establish a CCR2-based autocrine, feed-forward loop that amplifies renal inflammation.
Collapse
Affiliation(s)
- Vijayashree Mysore
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Suhail Tahir
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Kazuhiro Furuhashi
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Jatin Arora
- Center for Data Sciences, Brigham and Women’s Hospital, Boston, MA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Xavier Cullere
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Pascal Yazbeck
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Miroslav Sekulic
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | | | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women’s Hospital, Boston, MA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Centre for Genetics and Genomics Versus Arthritis, The University of Manchester, Manchester, UK
| | - Bruce H. Horwitz
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA
| | - Tanya N. Mayadas
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
19
|
Xu B, Chen L, Zhan Y, Marquez KNS, Zhuo L, Qi S, Zhu J, He Y, Chen X, Zhang H, Shen Y, Chen G, Gu J, Guo Y, Liu S, Xie T. The Biological Functions and Regulatory Mechanisms of Fatty Acid Binding Protein 5 in Various Diseases. Front Cell Dev Biol 2022; 10:857919. [PMID: 35445019 PMCID: PMC9013884 DOI: 10.3389/fcell.2022.857919] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
In recent years, fatty acid binding protein 5 (FABP5), also known as fatty acid transporter, has been widely researched with the help of modern genetic technology. Emerging evidence suggests its critical role in regulating lipid transport, homeostasis, and metabolism. Its involvement in the pathogenesis of various diseases such as metabolic syndrome, skin diseases, cancer, and neurological diseases is the key to understanding the true nature of the protein. This makes FABP5 be a promising component for numerous clinical applications. This review has summarized the most recent advances in the research of FABP5 in modulating cellular processes, providing an in-depth analysis of the protein's biological properties, biological functions, and mechanisms involved in various diseases. In addition, we have discussed the possibility of using FABP5 as a new diagnostic biomarker and therapeutic target for human diseases, shedding light on challenges facing future research.
Collapse
Affiliation(s)
- Binyue Xu
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Yu Zhan
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Karl Nelson S. Marquez
- Clinical Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hankou, China
| | - Lvjia Zhuo
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Shasha Qi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Jinyu Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Ying He
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Xudong Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Hao Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Yingying Shen
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Gongxing Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Jianzhong Gu
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yong Guo
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuiping Liu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
20
|
Higashide M, Furuhashi M, Watanabe M, Itoh K, Suzuki S, Umetsu A, Tsugeno Y, Ida Y, Hikage F, Ohguro H. Fatty Acid-Binding Proteins 4 and 5 Are Involved in the Pathogenesis of Retinal Vascular Diseases in Different Manners. Life (Basel) 2022; 12:life12040467. [PMID: 35454958 PMCID: PMC9025502 DOI: 10.3390/life12040467] [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: 03/03/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/16/2022] Open
Abstract
This study reports on the pathological significance of the vitreous fatty acid-binding protein (Vt-FABP) 4 and 5, and vascular endothelial growth factor A (Vt-VEGFA) in patients with retinal vascular diseases (RVDs) including proliferative diabetic retinopathy (PDR) and retinal vein occlusion (RVO). Subjects with PDR (n = 20), RVO (n = 10), and controls (epiretinal membrane, n = 18) who had undergone vitrectomies were enrolled in this study. The levels of Vt-FABP4, Vt-FABP5, and Vt-VEGFA were evaluated by enzyme-linked immunosorbent assays (ELISA). Retinal circulation levels were measured by a laser-speckle flow analyzer (LSFA) and other relevant data were collected. The Vt-FABP5 levels were significantly (p < 0.05) elevated in patients with RVDs compared to control patients. This elevation was more evident in patients with RVO than with PDR. Log Vt-FABP5 was significantly correlated negatively or positively with all the LSFA retinal circulation indexes or Log triglycerides (r = 0.31, p = 0.031), respectively. However, the elevations in the Vt-FABP4 and Vt-VEGFA levels were more evident in the PDR group (p < 0.05) and these factors were correlated positively with Log fasting glucose and negatively with some of the LSFA retinal circulation indexes. Multivariable regression analyses indicated that the LSFA blood flows of the optic disc at baseline was an independent effector with Log Vt-FABP5 other than several possible factors including age, gender, Log triglycerides, Log Vt-FABP4 and Log Vt-VEGFA. These current findings suggest that Vt-FABP5 is involved in the pathogenesis of RVD in a manner that is different from that for Vt-FABP4 and Vt-VEGFA, presumably by regulating retinal circulation.
Collapse
Affiliation(s)
- Megumi Higashide
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo 060-8556, Japan; (M.H.); (M.W.); (K.I.); (S.S.); (A.U.); (Y.T.); (Y.I.); (F.H.)
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan;
| | - Megumi Watanabe
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo 060-8556, Japan; (M.H.); (M.W.); (K.I.); (S.S.); (A.U.); (Y.T.); (Y.I.); (F.H.)
| | - Kaku Itoh
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo 060-8556, Japan; (M.H.); (M.W.); (K.I.); (S.S.); (A.U.); (Y.T.); (Y.I.); (F.H.)
| | - Soma Suzuki
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo 060-8556, Japan; (M.H.); (M.W.); (K.I.); (S.S.); (A.U.); (Y.T.); (Y.I.); (F.H.)
| | - Araya Umetsu
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo 060-8556, Japan; (M.H.); (M.W.); (K.I.); (S.S.); (A.U.); (Y.T.); (Y.I.); (F.H.)
| | - Yuri Tsugeno
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo 060-8556, Japan; (M.H.); (M.W.); (K.I.); (S.S.); (A.U.); (Y.T.); (Y.I.); (F.H.)
| | - Yosuke Ida
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo 060-8556, Japan; (M.H.); (M.W.); (K.I.); (S.S.); (A.U.); (Y.T.); (Y.I.); (F.H.)
| | - Fumihito Hikage
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo 060-8556, Japan; (M.H.); (M.W.); (K.I.); (S.S.); (A.U.); (Y.T.); (Y.I.); (F.H.)
| | - Hiroshi Ohguro
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo 060-8556, Japan; (M.H.); (M.W.); (K.I.); (S.S.); (A.U.); (Y.T.); (Y.I.); (F.H.)
- Correspondence: ; Tel.: +81-61-12-111
| |
Collapse
|
21
|
Wculek SK, Dunphy G, Heras-Murillo I, Mastrangelo A, Sancho D. Metabolism of tissue macrophages in homeostasis and pathology. Cell Mol Immunol 2022; 19:384-408. [PMID: 34876704 PMCID: PMC8891297 DOI: 10.1038/s41423-021-00791-9] [Citation(s) in RCA: 212] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/25/2021] [Indexed: 02/06/2023] Open
Abstract
Cellular metabolism orchestrates the intricate use of tissue fuels for catabolism and anabolism to generate cellular energy and structural components. The emerging field of immunometabolism highlights the importance of cellular metabolism for the maintenance and activities of immune cells. Macrophages are embryo- or adult bone marrow-derived leukocytes that are key for healthy tissue homeostasis but can also contribute to pathologies such as metabolic syndrome, atherosclerosis, fibrosis or cancer. Macrophage metabolism has largely been studied in vitro. However, different organs contain diverse macrophage populations that specialize in distinct and often tissue-specific functions. This context specificity creates diverging metabolic challenges for tissue macrophage populations to fulfill their homeostatic roles in their particular microenvironment and conditions their response in pathological conditions. Here, we outline current knowledge on the metabolic requirements and adaptations of macrophages located in tissues during homeostasis and selected diseases.
Collapse
Affiliation(s)
- Stefanie K Wculek
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain.
| | - Gillian Dunphy
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Ignacio Heras-Murillo
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Annalaura Mastrangelo
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain.
| |
Collapse
|
22
|
Dolfi B, Gallerand A, Haschemi A, Guinamard RR, Ivanov S. Macrophage metabolic regulation in atherosclerotic plaque. Atherosclerosis 2021; 334:1-8. [PMID: 34450556 DOI: 10.1016/j.atherosclerosis.2021.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/02/2021] [Accepted: 08/05/2021] [Indexed: 12/18/2022]
Abstract
Metabolism plays a key role in controlling immune cell functions. In this review, we will discuss the diversity of plaque resident myeloid cells and will focus on their metabolic demands that could reflect on their particular intraplaque localization. Defining the metabolic configuration of plaque resident myeloid cells according to their topologic distribution could provide answers to key questions regarding their functions and contribution to disease development.
Collapse
Affiliation(s)
| | | | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Rodolphe R Guinamard
- Université Côte D'Azur, Laboratoire de PhysioMédecine Moléculaire, CNRS, Nice, France
| | | |
Collapse
|
23
|
Development of mode of action networks related to the potential role of PPARγ in respiratory diseases. Pharmacol Res 2021; 172:105821. [PMID: 34403731 DOI: 10.1016/j.phrs.2021.105821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 11/30/2022]
Abstract
The peroxisome proliferator-activated receptor γ (PPARγ) is a key transcription factor, operating at the intercept of metabolic control and immunomodulation. It is ubiquitously expressed in multiple tissues and organs, including lungs. There is a growing body of information supporting the role of PPARγ signalling in respiratory diseases. The aim of the present study was to develop mode of action (MoA) networks reflecting the relationships between PPARγ signalling and the progression/alleviation of a spectrum of lung pathologies. Data mining was performed using the resources of the NIH PubMed and PubChem information systems. By linking available data on pathological/therapeutic effects of PPARγ modulation, knowledge-based MoA networking at different levels of biological organization (molecular, cellular, tissue, organ, and system) was performed. Multiple MoA networks were developed to relate PPARγ modulation to the progress or the alleviation of pulmonary disorders, triggered by diverse pathogenic, genetic, chemical, or mechanical factors. Pharmacological targeting of PPARγ signalling was discussed with regard to ligand- and cell type-specific effects in the context of distinct disease inductor- and disease stage-dependent patterns. The proposed MoA networking analysis allows for a better understanding of the potential role of PPARγ modulation in lung pathologies. It presents a mechanistically justified basis for further computational, experimental, and clinical monitoring studies on the dynamic control of PPARγ signalling in respiratory diseases.
Collapse
|
24
|
Jin R, Hao J, Yi Y, Sauter E, Li B. Regulation of macrophage functions by FABP-mediated inflammatory and metabolic pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158964. [PMID: 33984518 PMCID: PMC8169605 DOI: 10.1016/j.bbalip.2021.158964] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022]
Abstract
Macrophages are almost everywhere in the body, where they serve pivotal functions in maintaining tissue homeostasis, remodeling, and immunoregulation. Macrophages are traditionally thought to differentiate from bone marrow-derived hematopoietic stem cells (HSCs). Emerging studies suggest that some tissue macrophages at steady state originate from embryonic precursors in the yolk sac or fetal liver and are maintained in situ by self-renewal, but bone marrow-derived monocytes can give rise to tissue macrophages in pathogenic settings, such as inflammatory injuries and cancer. Macrophages are popularly classified as Th1 cytokine (e.g. IFNγ)-activated M1 macrophages (the classical activation) or Th2 cytokine (e.g. IL-4)-activated M2 macrophages (the alternative activation). However, given the myriad arrays of stimuli macrophages may encounter from local environment, macrophages exhibit notorious heterogeneity in their phenotypes and functions. Determining the underlying metabolic pathways engaged during macrophage activation is critical for understanding macrophage phenotypic and functional adaptivity under different disease settings. Fatty acid binding proteins (FABPs) represent a family of evolutionarily conserved proteins facilitating lipid transport, metabolism and responses inside cells. More specifically, adipose-FABP (A-FABP) and epidermal-FABP (E-FABP) are highly expressed in macrophages and play a central role in integrating metabolic and inflammatory pathways. In this review we highlight how A-FABP and E-FABP are respectively upregulated in different subsets of activated macrophages and provide a unique perspective in defining macrophage phenotypic and functional heterogeneity through FABP-regulated lipid metabolic and inflammatory pathways.
Collapse
Affiliation(s)
- Rong Jin
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA; Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiaqing Hao
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Yanmei Yi
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA; School of Basic Medical Sciences, Guangdong Medical University, Zhanjiang, China
| | - Edward Sauter
- Division of Cancer Prevention, NIH/NCI, Bethesda, MD, USA
| | - Bing Li
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
25
|
Umbarawan Y, Enoura A, Ogura H, Sato T, Horikawa M, Ishii T, Sunaga H, Matsui H, Yokoyama T, Kawakami R, Maeno T, Setou M, Kurabayashi M, Iso T. FABP5 Is a Sensitive Marker for Lipid-Rich Macrophages in the Luminal Side of Atherosclerotic Lesions. Int Heart J 2021; 62:666-676. [PMID: 33994513 DOI: 10.1536/ihj.20-676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Lipid-rich macrophages in atherosclerotic lesions are thought to be derived from myeloid and vascular smooth muscle cells. A series of studies with genetic and pharmacological inhibition of fatty acid binding protein 4 (FABP4) and FABP5 and bone marrow transplant experiments with FABP4/5 deficient cells in mice have demonstrated that these play an important role in the development of atherosclerosis. However, it is still uncertain about the differential cell-type specificity and distribution between FABP4- and FABP5-expressing cells in early- and late-stage atherosclerotic lesions. In this study, we first explored spatial distribution of FABP4/5 in atherosclerotic lesions in apolipoprotein E deficient (ApoE-/-) mice. FABP4 was only marginally detected in early and advanced lesions, whereas FABP5 was abundantly expressed in these lesions. In advanced lesions, the FABP5-positive area was mostly restricted to the foam cell layer adjacent to the lumen above collagen and elastic fibers with a high signal/noise ratio. Oil red O (ORO) staining revealed that FABP5-positive cells were lipid-rich in early and advanced lesions. Together, most of lipid-rich FABP5-positive cells reside adjacent to the lumen above collagen and elastic fibers. We next studied involvement of FABP5 in lesion formation of atherosclerosis using ApoE-/- FABP5-/- mice. However, deletion of FABP5 did not affect the development of atherosclerosis. These findings, along with previous reports, suggest a novel notion that FABP5 is a sensitive marker for bone marrow-derived lipid-rich macrophages in the luminal side of atherosclerotic lesions, although its functional significance remains elusive.
Collapse
Affiliation(s)
- Yogi Umbarawan
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine
- Department of Internal Medicine, Faculty of Medicine Universitas Indonesia
| | - Aiko Enoura
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences
| | - Harumi Ogura
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences
| | - Tomohito Sato
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine
- International Mass Imaging Center, Hamamatsu University School of Medicine
| | - Makoto Horikawa
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine
- International Mass Imaging Center, Hamamatsu University School of Medicine
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University
| | - Tomoaki Ishii
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences
| | - Hiroaki Sunaga
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine
- Center for Liberal Arts and Sciences, Ashikaga University
| | - Hiroki Matsui
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences
| | - Tomoyuki Yokoyama
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences
| | - Ryo Kawakami
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine
| | - Toshitaka Maeno
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine
- International Mass Imaging Center, Hamamatsu University School of Medicine
- Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center
| | - Masahiko Kurabayashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine
| | - Tatsuya Iso
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine
| |
Collapse
|
26
|
Mineo C. Lipoprotein receptor signalling in atherosclerosis. Cardiovasc Res 2021; 116:1254-1274. [PMID: 31834409 DOI: 10.1093/cvr/cvz338] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/01/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
The founding member of the lipoprotein receptor family, low-density lipoprotein receptor (LDLR) plays a major role in the atherogenesis through the receptor-mediated endocytosis of LDL particles and regulation of cholesterol homeostasis. Since the discovery of the LDLR, many other structurally and functionally related receptors have been identified, which include low-density lipoprotein receptor-related protein (LRP)1, LRP5, LRP6, very low-density lipoprotein receptor, and apolipoprotein E receptor 2. The scavenger receptor family members, on the other hand, constitute a family of pattern recognition proteins that are structurally diverse and recognize a wide array of ligands, including oxidized LDL. Among these are cluster of differentiation 36, scavenger receptor class B type I and lectin-like oxidized low-density lipoprotein receptor-1. In addition to the initially assigned role as a mediator of the uptake of macromolecules into the cell, a large number of studies in cultured cells and in in vivo animal models have revealed that these lipoprotein receptors participate in signal transduction to modulate cellular functions. This review highlights the signalling pathways by which these receptors influence the process of atherosclerosis development, focusing on their roles in the vascular cells, such as macrophages, endothelial cells, smooth muscle cells, and platelets. Human genetics of the receptors is also discussed to further provide the relevance to cardiovascular disease risks in humans. Further knowledge of the vascular biology of the lipoprotein receptors and their ligands will potentially enhance our ability to harness the mechanism to develop novel prophylactic and therapeutic strategies against cardiovascular diseases.
Collapse
Affiliation(s)
- Chieko Mineo
- Department of Pediatrics and Cell Biology, Center for Pulmonary and Vascular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA
| |
Collapse
|
27
|
Kim KW, Ivanov S, Williams JW. Monocyte Recruitment, Specification, and Function in Atherosclerosis. Cells 2020; 10:E15. [PMID: 33374145 PMCID: PMC7823291 DOI: 10.3390/cells10010015] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/21/2022] Open
Abstract
Atherosclerotic lesions progress through the continued recruitment of circulating blood monocytes that differentiate into macrophages within plaque. Lesion-associated macrophages are the primary immune cells present in plaque, where they take up cholesterol and store lipids in the form of small droplets resulting in a unique morphology termed foam cell. Recent scientific advances have used single-cell gene expression profiling, live-cell imaging, and fate mapping approaches to describe macrophage and monocyte contributions to pro- or anti-inflammatory mechanisms, in addition to functions of motility and proliferation within lesions. Yet, many questions regarding tissue-specific regulation of monocyte-to-macrophage differentiation and the contribution of recruited monocytes at stages of atherosclerotic disease progression remain unknown. In this review, we highlight recent advances regarding the role of monocyte and macrophage dynamics in atherosclerotic disease and identify gaps in knowledge that we hope will allow for advancing therapeutic treatment or prevention strategies for cardiovascular disease.
Collapse
Affiliation(s)
- Ki-Wook Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| | - Stoyan Ivanov
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire C3M, Université Côte Azur, 06204 Nice, France;
| | - Jesse W. Williams
- Center for Immunology, Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
28
|
Gallerand A, Stunault MI, Merlin J, Guinamard RR, Yvan-Charvet L, Ivanov S. Myeloid Cell Diversity and Impact of Metabolic Cues during Atherosclerosis. IMMUNOMETABOLISM 2020; 2:immunometab20200028. [PMID: 39649554 PMCID: PMC7617020 DOI: 10.20900/immunometab20200028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Myeloid cells are key contributors to tissue, immune and metabolic homeostasis and their alteration fuels inflammation and associated disorders such as atherosclerosis. Conversely, in a classical chicken-and-egg situation, systemic and local metabolism, together with receptor-mediated activation, regulate intracellular metabolism and reprogram myeloid cell functions. Those regulatory loops are notable during the development of atherosclerotic lesions. Therefore, understanding the intricate metabolic mechanisms regulating myeloid cell biology could lead to innovative approaches to prevent and treat cardiovascular diseases. In this review, we will attempt to summarize the different metabolic factors regulating myeloid cell homeostasis and contribution to atherosclerosis, the most frequent cardiovascular disease.
Collapse
Affiliation(s)
- Alexandre Gallerand
- Mediterranean center of molecular medicine (C3M)-Université Côte d’Azur–INSERM U1065, Team 13, Nice, 06200, France
| | - Marion I. Stunault
- Mediterranean center of molecular medicine (C3M)-Université Côte d’Azur–INSERM U1065, Team 13, Nice, 06200, France
| | - Johanna Merlin
- Mediterranean center of molecular medicine (C3M)-Université Côte d’Azur–INSERM U1065, Team 13, Nice, 06200, France
| | - Rodolphe R. Guinamard
- Mediterranean center of molecular medicine (C3M)-Université Côte d’Azur–INSERM U1065, Team 13, Nice, 06200, France
| | - Laurent Yvan-Charvet
- Mediterranean center of molecular medicine (C3M)-Université Côte d’Azur–INSERM U1065, Team 13, Nice, 06200, France
| | - Stoyan Ivanov
- Mediterranean center of molecular medicine (C3M)-Université Côte d’Azur–INSERM U1065, Team 13, Nice, 06200, France
| |
Collapse
|
29
|
Stapleton K, Das S, Reddy MA, Leung A, Amaram V, Lanting L, Chen Z, Zhang L, Palanivel R, Deiuliis JA, Natarajan R. Novel Long Noncoding RNA, Macrophage Inflammation-Suppressing Transcript ( MIST), Regulates Macrophage Activation During Obesity. Arterioscler Thromb Vasc Biol 2020; 40:914-928. [PMID: 32078363 PMCID: PMC7098442 DOI: 10.1161/atvbaha.119.313359] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Supplemental Digital Content is available in the text. Objective: Systemic low-grade inflammation associated with obesity and metabolic syndrome is a strong risk factor for the development of diabetes mellitus and associated cardiovascular complications. This inflammatory state is caused by release of proinflammatory cytokines by macrophages, especially in adipose tissue. Long noncoding RNAs regulate macrophage activation and inflammatory gene networks, but their role in macrophage dysfunction during diet-induced obesity has been largely unexplored. Approach and Results: We sequenced total RNA from peritoneal macrophages isolated from mice fed either high-fat diet or standard diet and performed de novo transcriptome assembly to identify novel differentially expressed mRNAs and long noncoding RNAs. A top candidate long noncoding RNA, macrophage inflammation-suppressing transcript (Mist), was downregulated in both peritoneal macrophages and adipose tissue macrophages from high-fat diet–fed mice. GapmeR-mediated Mist knockdown in vitro and in vivo upregulated expression of genes associated with immune response and inflammation and increased modified LDL (low-density lipoprotein) uptake in macrophages. Conversely, Mist overexpression decreased basal and LPS (lipopolysaccharide)-induced expression of inflammatory response genes and decreased modified LDL uptake. RNA-pull down coupled with mass spectrometry showed that Mist interacts with PARP1 (poly [ADP]-ribose polymerase-1). Disruption of this RNA-protein interaction increased PARP1 recruitment and chromatin PARylation at promoters of inflammatory genes, resulting in increased gene expression. Furthermore, human orthologous MIST was also downregulated by proinflammatory stimuli, and its expression in human adipose tissue macrophages inversely correlated with obesity and insulin resistance. Conclusions: Mist is a novel protective long noncoding RNA, and its loss during obesity contributes to metabolic dysfunction and proinflammatory phenotype of macrophages via epigenetic mechanisms.
Collapse
Affiliation(s)
- Kenneth Stapleton
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA.,Irell and Manella Graduate School of Biological Sciences (K.S., V.A., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Sadhan Das
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Marpadga A Reddy
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Amy Leung
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Vishnu Amaram
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA.,Irell and Manella Graduate School of Biological Sciences (K.S., V.A., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Linda Lanting
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Zhuo Chen
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Lingxiao Zhang
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Rengasamy Palanivel
- Cardiovascular Research Institute of the Case Western Reserve University, Cleveland, OH (R.P., J.A.D.)
| | - Jeffrey A Deiuliis
- Cardiovascular Research Institute of the Case Western Reserve University, Cleveland, OH (R.P., J.A.D.)
| | - Rama Natarajan
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA.,Irell and Manella Graduate School of Biological Sciences (K.S., V.A., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| |
Collapse
|
30
|
Furuhashi M, Sakuma I, Morimoto T, Higashiura Y, Sakai A, Matsumoto M, Sakuma M, Shimabukuro M, Nomiyama T, Arasaki O, Node K, Ueda S. Independent and Distinct Associations of FABP4 and FABP5 With Metabolic Parameters in Type 2 Diabetes Mellitus. Front Endocrinol (Lausanne) 2020; 11:575557. [PMID: 33071982 PMCID: PMC7538548 DOI: 10.3389/fendo.2020.575557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/21/2020] [Indexed: 01/22/2023] Open
Abstract
Objective: Among fatty acid-binding proteins (FABPs), secreted forms of FABP4 and FABP5, which are expressed in adipocytes and macrophages, act as bioactive molecules. We investigated concentrations of FABP4 and FABP5 in patients with type 2 diabetes mellitus. Methods: As a sub-analysis study of the Randomized Evaluation of Anagliptin vs. Sitagliptin On low-density lipoproteiN cholesterol in diabetes (REASON) trial, 256 patients (male/female: 146/110, age: 68 ± 10 years) with type 2 diabetes mellitus and dyslipidemia who were receiving statin therapy were recruited. Patients who had been treated with a thiazolidinedione were excluded. Results: Several drugs which may modulate FABP4 levels including statins, dipeptidyl peptidase-4 inhibitors and angiotensin II receptor blockers had been administered in 100, 81, and 51% of the recruited patients, respectively. The level of FABP4, but not that of FABP5, was significantly higher in females than in males. Multivariable linear regression analysis demonstrated that waist circumference (β = 0.21), estimated glomerular filtration rate (β = -0.31), triglycerides (β = 0.16), and FABP5 (β = 0.39) were independent predictors of FABP4 level after adjusting age and sex. On the other hand, FABP5 level was independently associated with levels of FABP4 (β = 0.57) and high-density lipoprotein (HDL) cholesterol (β = -0.12). Conclusions: Concentrations of FABP4 and FABP5 are independent predictors of each other in patients with type 2 diabetes mellitus. There are distinct independent associations of FABP4 with renal dysfunction, adiposity and hypertriglyceridemia and there is a distinct independent association of FABP5 with a low HDL cholesterol level in type 2 diabetic patients with dyslipidemia at high risks for cardiovascular disease who are receiving statin therapy.
Collapse
Affiliation(s)
- Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- *Correspondence: Masato Furuhashi
| | - Ichiro Sakuma
- Caress Sapporo Hokko Memorial Clinic, Sapporo, Japan
| | - Takeshi Morimoto
- Department of Clinical Epidemiology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Yukimura Higashiura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Akiko Sakai
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Megumi Matsumoto
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mio Sakuma
- Department of Clinical Epidemiology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Michio Shimabukuro
- Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Takashi Nomiyama
- Department of Diabetes, Metabolism and Endocrinology, International University of Health and Welfare Ichikawa Hospital, Ichikawa, Japan
| | - Osamu Arasaki
- Department of Cardiology, Tomishiro Central Hospital, Tomigusuku, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
| | - Shinichiro Ueda
- Department of Pharmacology and Therapeutics, University of the Ryukyus, Okinawa, Japan
| |
Collapse
|
31
|
Ménégaut L, Jalil A, Thomas C, Masson D. Macrophage fatty acid metabolism and atherosclerosis: The rise of PUFAs. Atherosclerosis 2019; 291:52-61. [PMID: 31693943 DOI: 10.1016/j.atherosclerosis.2019.10.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/26/2019] [Accepted: 10/09/2019] [Indexed: 01/24/2023]
Abstract
Among the pathways involved in the regulation of macrophage functions, the metabolism of unsaturated fatty acids is central. Indeed, unsaturated fatty acids act as precursors of bioactive molecules such as prostaglandins, leukotrienes, resolvins and related compounds. As components of phospholipids, they have a pivotal role in cell biology by regulating membrane fluidity and membrane-associated cellular processes. Finally, polyunsaturated fatty acids (PUFAs) are also endowed with ligand properties for numerous membrane or nuclear receptors. Although myeloid cells are dependent on the metabolic context for the uptake of essential FAs, recent studies showed that these cells autonomously handle the synthesis of n-3 and n-6 long chain PUFAs such as arachidonic acid and eicosapentaenoic acid. Moreover, targeting PUFA metabolism in macrophages influences pathological processes, including atherosclerosis, by modulating macrophage functions. Omics evidence also supports a role for macrophage PUFA metabolism in the development of cardiometabolic diseases in humans. Currently, there is a renewed interest in the role of n-3/n-6 PUFAs and their oxygenated derivatives in the onset of atherosclerosis and plaque rupture. Purified n-3 FA supplementation appears as a potential strategy in the treatment and prevention of cardiovascular diseases. In this context, the ability of immune cells to handle and to synthesize very long chain PUFA must absolutely be integrated and better understood.
Collapse
Affiliation(s)
- Louise Ménégaut
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Antoine Jalil
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Charles Thomas
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - David Masson
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France.
| |
Collapse
|
32
|
Identification of new dual FABP4/5 inhibitors based on a naphthalene-1-sulfonamide FABP4 inhibitor. Bioorg Med Chem 2019; 27:115015. [PMID: 31420256 DOI: 10.1016/j.bmc.2019.07.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 12/30/2022]
Abstract
Fatty acid binding protein 4 (FABP4) and fatty acid binding protein 5 (FABP5) are mainly expressed in adipocytes and/or macrophages and play essential roles in energy metabolism and inflammation. When FABP4 function is diminished, FABP5 expression is highly increased possibly as a functional compensation. Dual FABP4/5 inhibitors are expected to provide beneficial synergistic effect on treating diabetes, atherosclerosis, and inflammation-related diseases. Starting from our previously reported selective FABP4 inhibitor 8, structural biology information was used to modulate the selectivity profile and to design potent dual FABP4/5 inhibitors with good selectivity against FABP3. Two compounds A16 and B8 were identified to show inhibitory activities against both FABP4/5 and good selectivity over FABP3, which could also reduce the level of forskolin-stimulated lipolysis in mature 3T3-L1 adipocytes. Compared with compound 8, these two compounds exhibited better anti-inflammatory effects in lipopolysaccharide-stimulated RAW264.7 murine macrophages, with decreased levels of pro-inflammatory cytokines TNFα and MCP-1 and apparently inhibited IKK/NF-κB pathway.
Collapse
|
33
|
LI TT, WANG ZB, LI Y, CAO F, YANG BY, KUANG HX. The mechanisms of traditional Chinese medicine underlying the prevention and treatment of atherosclerosis. Chin J Nat Med 2019; 17:401-412. [DOI: 10.1016/s1875-5364(19)30048-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Indexed: 02/07/2023]
|
34
|
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
|
35
|
Melville D, Gorur A, Schekman R. Fatty-acid binding protein 5 modulates the SAR1 GTPase cycle and enhances budding of large COPII cargoes. Mol Biol Cell 2018; 30:387-399. [PMID: 30485159 PMCID: PMC6589570 DOI: 10.1091/mbc.e18-09-0548] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
COPII-coated vesicles are the primary mediators of ER-to-Golgi trafficking. Sar1, one of the five core COPII components, is a highly conserved small GTPase, which, upon GTP binding, recruits the other COPII proteins to the ER membrane. It has been hypothesized that the changes in the kinetics of SAR1 GTPase may allow for the secretion of large cargoes. Here we developed a cell-free assay to recapitulate COPII-dependent budding of large lipoprotein cargoes from the ER. We identified fatty-acid binding protein 5 (FABP5) as an enhancer of this budding process. We found that FABP5 promotes the budding of particles ∼150 nm in diameter and modulates the kinetics of the SAR1 GTPase cycle. We further found that FABP5 enhances the trafficking of lipoproteins and of other cargoes, including collagen. These data identify a novel regulator of SAR1 GTPase activity and highlight the importance of this activity for trafficking of large cargoes.
Collapse
Affiliation(s)
- David Melville
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720
| | - Amita Gorur
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720
| | - Randy Schekman
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
36
|
Wall VZ, Barnhart S, Kramer F, Kanter JE, Vivekanandan-Giri A, Pennathur S, Bolego C, Ellis JM, Gijón MA, Wolfgang MJ, Bornfeldt KE. Inflammatory stimuli induce acyl-CoA thioesterase 7 and remodeling of phospholipids containing unsaturated long (≥C20)-acyl chains in macrophages. J Lipid Res 2017; 58:1174-1185. [PMID: 28416579 DOI: 10.1194/jlr.m076489] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Indexed: 01/01/2023] Open
Abstract
Acyl-CoA thioesterase 7 (ACOT7) is an intracellular enzyme that converts acyl-CoAs to FFAs. ACOT7 is induced by lipopolysaccharide (LPS); thus, we investigated downstream effects of LPS-induced induction of ACOT7 and its role in inflammatory settings in myeloid cells. Enzymatic thioesterase activity assays in WT and ACOT7-deficient macrophage lysates indicated that endogenous ACOT7 contributes a significant fraction of total acyl-CoA thioesterase activity toward C20:4-, C20:5-, and C22:6-CoA, but contributes little activity toward shorter acyl-CoA species. Lipidomic analyses revealed that LPS causes a dramatic increase, primarily in bis(monoacylglycero)phosphate species containing long (≥C20) polyunsaturated acyl-chains in macrophages, and that the limited effect observed by ACOT7 deficiency is restricted to glycerophospholipids containing 20-carbon unsaturated acyl-chains. Furthermore, ACOT7 deficiency did not detectably alter the ability of LPS to induce cytokines or prostaglandin E2 production in macrophages. Consistently, although ACOT7 was induced in macrophages from diabetic mice, hematopoietic ACOT7 deficiency did not alter the stimulatory effect of diabetes on systemic inflammation or atherosclerosis in LDL receptor-deficient mice. Thus, inflammatory stimuli induce ACOT7 and remodeling of phospholipids containing unsaturated long (≥C20)-acyl chains in macrophages, and, although ACOT7 has preferential thioesterase activity toward these lipid species, loss of ACOT7 has no major detrimental effect on macrophage inflammatory phenotypes.≥.
Collapse
Affiliation(s)
- Valerie Z Wall
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA.,Division of Metabolism, Endocrinology and Nutrition, and Department of Pathology, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Shelley Barnhart
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Farah Kramer
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Jenny E Kanter
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | | | | | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Jessica M Ellis
- Department of Nutrition Science, Purdue University, West Lafayette, IN.,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Miguel A Gijón
- Department of Pharmacology, University of Colorado Denver, Aurora, CO
| | - Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Karin E Bornfeldt
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA .,Division of Metabolism, Endocrinology and Nutrition, and Department of Pathology, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| |
Collapse
|
37
|
Furuhashi M, Ogura M, Matsumoto M, Yuda S, Muranaka A, Kawamukai M, Omori A, Tanaka M, Moniwa N, Ohnishi H, Saitoh S, Harada-Shiba M, Shimamoto K, Miura T. Serum FABP5 concentration is a potential biomarker for residual risk of atherosclerosis in relation to cholesterol efflux from macrophages. Sci Rep 2017; 7:217. [PMID: 28303004 PMCID: PMC5427929 DOI: 10.1038/s41598-017-00177-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/13/2017] [Indexed: 12/16/2022] Open
Abstract
Cholesterol efflux capacity (CEC) from macrophages, the first step in the reverse cholesterol transport pathway, is inversely associated with residual risk for atherosclerotic cardiovascular disease. Fatty acid-binding protein 4 (FABP4) and FABP5 are expressed in both adipocytes and macrophages and play significant roles in the development of insulin resistance and atherosclerosis. Both FABP4 and FABP5 are secreted from cells, and their circulating levels are associated with insulin resistance and atherosclerosis. We investigated the association between CEC and levels of FABP4 and FABP5 in 250 subjects without any medications. CEC was positively correlated with HDL cholesterol level and negatively correlated with concentrations of high-sensitivity C-reactive protein (hsCRP) and FABP5, but not FABP4. Multiple regression analysis demonstrated that FABP5 concentration was an independent predictor of CEC after adjustment of age, gender and levels of HDL cholesterol and hsCRP. In 129 of the 250 subjects who underwent carotid ultrasonography, mean intima-media thickness was negatively correlated with CEC and was positively correlated with concentrations of FABP4 and FABP5. In conclusion, in contrast to FABP4, circulating FABP5 is associated with decreased CEC and carotid atherosclerosis, suggesting that FABP5 level is a regulatory factor of CEC and a potential biomarker for residual risk of atherosclerosis.
Collapse
Affiliation(s)
- Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | - Masatsune Ogura
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Megumi Matsumoto
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Satoshi Yuda
- Devision of Cardiology, Cardiovascular Center, Teine Keijinkai Hospital, Sapporo, Japan
| | - Atsuko Muranaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mina Kawamukai
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Akina Omori
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Norihito Moniwa
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Ohnishi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Public Health, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shigeyuki Saitoh
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Nursing, Division of Medical and Behavioral Subjects, Sapporo Medical University School of Health Sciences, Sapporo, Japan
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | | | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
38
|
Yamamoto T, Furuhashi M, Sugaya T, Oikawa T, Matsumoto M, Funahashi Y, Matsukawa Y, Gotoh M, Miura T. Transcriptome and Metabolome Analyses in Exogenous FABP4- and FABP5-Treated Adipose-Derived Stem Cells. PLoS One 2016; 11:e0167825. [PMID: 27936164 PMCID: PMC5148007 DOI: 10.1371/journal.pone.0167825] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 11/21/2016] [Indexed: 12/30/2022] Open
Abstract
Adipose-derived stem cells (ADSC), which exist near adipocytes in adipose tissue, have been used as a potential tool of regenerative medicine. Lipid chaperones, fatty acid-binding protein 4 (FABP4) and 5 (FABP5), are abundantly expressed in adipocytes. FABP4 has recently been shown to be secreted from adipocytes during lipolysis in a non-classical pathway and may act as an adipokine. Here, we investigated the role of exogenous FABP4 and FABP5 in transcriptional and metabolic regulation in ADSC. FABP4 and FABP5 were little expressed in ADSC. However, both FABP4 and FABP5 were significantly induced after adipocyte differentiation of ADSC and were secreted from the differentiated adipocytes. Analysis of microarray data, including gene ontology enrichment analysis and cascade analysis of the protein-protein interaction network using a transcription factor binding site search, demonstrated that treatment of ADSC with FABP4 or FABP5 affected several kinds of genes related to inflammatory and metabolic responses and the process of cell differentiation. Notably, myogenic factors, including myocyte enhancer factors, myogenic differentiation 1 and myogenin, were modulated by treatment of ADSC with FABP4, indicating that exogenous FABP4 treatment is partially associated with myogenesis in ADSC. Metabolome analysis showed that treatment of ADSC with FABP4 and with FABP5 similarly, but differently in extent, promoted hydrolysis and/or uptake of lipids, consequentially together with enhancement of β oxidation, inhibition of downstream of the glycolysis pathway, accumulation of amino acids, reduction of nucleic acid components and increase in the ratio of reduced and oxidized nicotinamide adenine dinucleotide phosphates (NADPH/NADP+), an indicator of reducing power, and the ratio of adenosine triphosphate and adenosine monophosphate (ATP/AMP), an indicator of the energy state, in ADSC. In conclusion, secreted FABP4 and FABP5 from adipocytes as adipokines differentially affect transcriptional and metabolic regulation in ADSC near adipocytes. The adiposity condition in the host of regenerative medicine may affect characteristics of ADSC by exposure of the balance of FABP4 and FABP5.
Collapse
Affiliation(s)
- Tokunori Yamamoto
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo, Japan
| | - Takeshi Sugaya
- Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan
- CIMIC Co., Ltd; Mass Building Yushima, Bunkyo-ku, Tokyo, Japan
| | - Tsuyoshi Oikawa
- CIMIC Co., Ltd; Mass Building Yushima, Bunkyo-ku, Tokyo, Japan
| | - Megumi Matsumoto
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo, Japan
| | - Yasuhito Funahashi
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihisa Matsukawa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Momokazu Gotoh
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo, Japan
| |
Collapse
|
39
|
Abstract
The remarkable plasticity and plethora of biological functions performed by macrophages have enticed scientists to study these cells in relation to atherosclerosis for >50 years, and major discoveries continue to be made today. It is now understood that macrophages play important roles in all stages of atherosclerosis, from initiation of lesions and lesion expansion, to necrosis leading to rupture and the clinical manifestations of atherosclerosis, to resolution and regression of atherosclerotic lesions. Lesional macrophages are derived primarily from blood monocytes, although recent research has shown that lesional macrophage-like cells can also be derived from smooth muscle cells. Lesional macrophages take on different phenotypes depending on their environment and which intracellular signaling pathways are activated. Rather than a few distinct populations of macrophages, the phenotype of the lesional macrophage is more complex and likely changes during the different phases of atherosclerosis and with the extent of lipid and cholesterol loading, activation by a plethora of receptors, and metabolic state of the cells. These different phenotypes allow the macrophage to engulf lipids, dead cells, and other substances perceived as danger signals; efflux cholesterol to high-density lipoprotein; proliferate and migrate; undergo apoptosis and death; and secrete a large number of inflammatory and proresolving molecules. This review article, part of the Compendium on Atherosclerosis, discusses recent advances in our understanding of lesional macrophage phenotype and function in different stages of atherosclerosis. With the increasing understanding of the roles of lesional macrophages, new research areas and treatment strategies are beginning to emerge.
Collapse
Affiliation(s)
- Ira Tabas
- From the Departments of Medicine (I.T.), Anatomy and Cell Biology (I.T.), and Physiology and Cellular Biophysics (I.T.), Columbia University, New York; and the Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (K.E.B.) and Department of Pathology (K.E.B.), UW Diabetes Institute, University of Washington School of Medicine, Seattle
| | - Karin E Bornfeldt
- From the Departments of Medicine (I.T.), Anatomy and Cell Biology (I.T.), and Physiology and Cellular Biophysics (I.T.), Columbia University, New York; and the Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (K.E.B.) and Department of Pathology (K.E.B.), UW Diabetes Institute, University of Washington School of Medicine, Seattle.
| |
Collapse
|
40
|
Xie LF, Ouyang JZ, Wang AP, Wang WB, Li XT, Wang BJ, Mu YM. Gene Expression Profile of Persistent Postoperative Hypertension Patients with Aldosterone-producing Adenomas. Chin Med J (Engl) 2016; 128:1618-26. [PMID: 26063364 PMCID: PMC4733735 DOI: 10.4103/0366-6999.158318] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Background: Hypertension often persists after adrenalectomy for primary aldosteronism (PA). Many studies have analyzed the outcomes of adrenalectomy for aldosterone-producing adenomas (APA) to identify predictive factors for persistent hypertension. However, differentially expressed genes in persistent postoperative hypertension remain unknown. Our aim was to describe gene expression profile of persistent postoperative hypertension patients with APA. Methods: In this study, we described and compared gene expression profiles in persistent postoperative hypertension and postoperative normotension in Chinese patients with APA using microarray analysis. Confirmation was performed with quantitative real time-polymerase chain reaction analysis. Bioinformatic analysis (gene ontology analysis, pathway analysis and network analysis) was used for further research. Results: Microarray analysis identified a total of 99 differentially expressed genes, including 18 up-regulated and 81 down-regulated genes. Among the dysregulated genes were fat atypical cadherin 1 as well as fatty acid binding protein 4 and other genes that have not been previously studied in persistent postoperative hypertension with APA. Bioinformatics analysis indicated that differentially expressed genes were associated with lipid metabolic process, metal ion binding, and cell differentiation. Pathway analysis determined that five pathways corresponded to the dysregulated transcripts. The mRNAs-ncRNAs co-expression network was composed of 49 network nodes and 72 connections between 18 coding genes and 31 noncoding genes. Conclusions: This study revealed differentially expressed genes in persistent postoperative hypertension with APA and provided a resource of candidate genes for exploration of possible drug targets and prognostic markers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yi-Ming Mu
- Department of Endocrinology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
41
|
Korner G, Scherer T, Adamsen D, Rebuffat A, Crabtree M, Rassi A, Scavelli R, Homma D, Ledermann B, Konrad D, Ichinose H, Wolfrum C, Horsch M, Rathkolb B, Klingenspor M, Beckers J, Wolf E, Gailus-Durner V, Fuchs H, Hrabě de Angelis M, Blau N, Rozman J, Thöny B. Mildly compromised tetrahydrobiopterin cofactor biosynthesis due to Pts variants leads to unusual body fat distribution and abdominal obesity in mice. J Inherit Metab Dis 2016; 39:309-19. [PMID: 26830550 DOI: 10.1007/s10545-015-9909-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 12/31/2022]
Abstract
Tetrahydrobiopterin (BH4) is an essential cofactor for the aromatic amino acid hydroxylases, alkylglycerol monooxygenase, and nitric oxide synthases (NOS). Inborn errors of BH4 metabolism lead to severe insufficiency of brain monoamine neurotransmitters while augmentation of BH4 by supplementation or stimulation of its biosynthesis is thought to ameliorate endothelial NOS (eNOS) dysfunction, to protect from (cardio-) vascular disease and/or prevent obesity and development of the metabolic syndrome. We have previously reported that homozygous knock-out mice for the 6-pyruvolytetrahydropterin synthase (PTPS; Pts-ko/ko) mice with no BH4 biosynthesis die after birth. Here we generated a Pts-knock-in (Pts-ki) allele expressing the murine PTPS-p.Arg15Cys with low residual activity (15% of wild-type in vitro) and investigated homozygous (Pts-ki/ki) and compound heterozygous (Pts-ki/ko) mutants. All mice showed normal viability and depending on the severity of the Pts alleles exhibited up to 90% reduction of PTPS activity concomitant with neopterin elevation and mild reduction of total biopterin while blood L-phenylalanine and brain monoamine neurotransmitters were unaffected. Yet, adult mutant mice with compromised PTPS activity (i.e., Pts-ki/ko, Pts-ki/ki or Pts-ko/wt) had increased body weight and elevated intra-abdominal fat. Comprehensive phenotyping of Pts-ki/ki mice revealed alterations in energy metabolism with proportionally higher fat content but lower lean mass, and increased blood glucose and cholesterol. Transcriptome analysis indicated changes in glucose and lipid metabolism. Furthermore, differentially expressed genes associated with obesity, weight loss, hepatic steatosis, and insulin sensitivity were consistent with the observed phenotypic alterations. We conclude that reduced PTPS activity concomitant with mildly compromised BH4-biosynthesis leads to abnormal body fat distribution and abdominal obesity at least in mice. This study associates a novel single gene mutation with monogenic forms of obesity.
Collapse
Affiliation(s)
- Germaine Korner
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland
- Affiliated with the Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zürich, Switzerland
- Children's Research Center (CRC), Zürich, Switzerland
| | - Tanja Scherer
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland
- Affiliated with the Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zürich, Switzerland
- Children's Research Center (CRC), Zürich, Switzerland
| | - Dea Adamsen
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland
- Affiliated with the Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zürich, Switzerland
- Children's Research Center (CRC), Zürich, Switzerland
| | - Alexander Rebuffat
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland
| | - Mark Crabtree
- BHF Centre of Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DU, Oxford, UK
| | - Anahita Rassi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zürich, Zürich, Switzerland
| | - Rossana Scavelli
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland
| | - Daigo Homma
- Department of Life Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Birgit Ledermann
- Division of Animal Facility, University of Zurich, Zürich, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Hiroshi Ichinose
- Department of Life Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Christian Wolfrum
- Institute of Food Nutrition and Health, Swiss Federal Institute of Technology Zürich, Zürich, Switzerland
| | - Marion Horsch
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 25, 81377, Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Martin Klingenspor
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, Technische Universität München, Am Forum 8, 85354, Freising-Weihenstephan, Germany
- ZIEL - Center for Nutrition and Food Sciences, Technische Universität München, D-85350, Freising, Germany
| | - Johannes Beckers
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, D-85354, Freising-Weihenstephan, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| | - Valérie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, D-85354, Freising-Weihenstephan, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Nenad Blau
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland.
- Dietmar-Hopp Metabolic Center, University Children's Hospital Heidelberg, Im Neuenheimer Feld 669, D-69120, Heidelberg, Germany.
| | - Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany.
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, Technische Universität München, Am Forum 8, 85354, Freising-Weihenstephan, Germany.
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany.
| | - Beat Thöny
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland.
- Affiliated with the Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zürich, Switzerland.
- Children's Research Center (CRC), Zürich, Switzerland.
| |
Collapse
|
42
|
Powell CA, Nasser MW, Zhao H, Wochna JC, Zhang X, Shapiro C, Shilo K, Ganju RK. Fatty acid binding protein 5 promotes metastatic potential of triple negative breast cancer cells through enhancing epidermal growth factor receptor stability. Oncotarget 2016; 6:6373-85. [PMID: 25779666 PMCID: PMC4467443 DOI: 10.18632/oncotarget.3442] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/12/2015] [Indexed: 12/31/2022] Open
Abstract
Fatty acid binding protein 5 (FABP5), an intracellular lipid binding protein, has been shown to play a role in various cancers, including breast cancer. However, FABP5 and its role in triple negative breast cancer (TNBC) have not been studied. We show FABP5 protein expression correlates with TNBC, high grade tumors, and worse disease-free survival in a tissue microarray containing 423 breast cancer patient samples. High FABP5 expression significantly correlates with epidermal growth factor receptor (EGFR) expression in these samples. Decreased tumor growth and lung metastasis were observed in FABP5-/- mice othotopically injected with murine breast cancer cells. FABP5 loss in TNBC tumor cells inhibited motility and invasion. Mechanistic studies revealed that FABP5 knockdown in TNBC cells results in decreased EGFR expression and FABP5 is important for EGF-induced metastatic signaling. Loss of FABP5 leads to proteasomal targeting of EGFR. Our studies show that FABP5 has a role in both host and tumor cell during breast cancer progression. These findings suggest that FABP5 mediates its enhanced effect on TNBC metastasis, in part, through EGFR, by inhibiting EGFR proteasomal degradation. These studies show, for the first time, a correlation between FABP5 and EGFR in enhancing TNBC metastasis through a novel mechanism.
Collapse
Affiliation(s)
| | - Mohd W Nasser
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Helong Zhao
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Jacob C Wochna
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Xiaoli Zhang
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Charles Shapiro
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, USA
| | - Konstantin Shilo
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Ramesh K Ganju
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
43
|
Abstract
Intracellular and extracellular interactions with proteins enables the functional and mechanistic diversity of lipids. Fatty acid-binding proteins (FABPs) were originally described as intracellular proteins that can affect lipid fluxes, metabolism and signalling within cells. As the functions of this protein family have been further elucidated, it has become evident that they are critical mediators of metabolism and inflammatory processes, both locally and systemically, and therefore are potential therapeutic targets for immunometabolic diseases. In particular, genetic deficiency and small molecule-mediated inhibition of FABP4 (also known as aP2) and FABP5 can potently improve glucose homeostasis and reduce atherosclerosis in mouse models. Further research has shown that in addition to their intracellular roles, some FABPs are found outside the cells, and FABP4 undergoes regulated, vesicular secretion. The circulating form of FABP4 has crucial hormonal functions in systemic metabolism. In this Review we discuss the roles and regulation of both intracellular and extracellular FABP actions, highlighting new insights that might direct drug discovery efforts and opportunities for management of chronic metabolic diseases.
Collapse
Affiliation(s)
- Gökhan S Hotamisligil
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, MN 55455, 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
|
Miyazaki H, Sawada T, Kiyohira M, Yu Z, Nakamura K, Yasumoto Y, Kagawa Y, Ebrahimi M, Islam A, Sharifi K, Kawamura S, Kodama T, Yamamoto Y, Adachi Y, Tokuda N, Terai S, Sakaida I, Ishikawa T, Owada Y. Fatty acid binding protein 7 regulates phagocytosis and cytokine production in Kupffer cells during liver injury. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2505-15. [PMID: 25041855 DOI: 10.1016/j.ajpath.2014.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 05/01/2014] [Accepted: 05/21/2014] [Indexed: 02/08/2023]
Abstract
Kupffer cells (KCs) are involved in the progression of liver diseases such as hepatitis and liver cancer. Several members of the fatty acid binding proteins (FABPs) are expressed by tissue macrophages, and FABP7 is localized only in KCs. To clarify the role of FABP7 in the regulation of KC function, we evaluated pathological changes of Fabp7 knockout mice during carbon tetrachloride-induced liver injury. During liver injury in Fabp7 knockout mice, serum liver enzymes were increased, cytokine expression (tumor necrosis factor-α, monocyte chemoattractant protein-1, and transforming growth factor-β) was decreased in the liver, and the number of KCs in the liver necrotic area was significantly decreased. Interestingly, in the FABP7-deficient KCs, phagocytosis of apoptotic cells was impaired, and expression of the scavenger receptor CD36 was markedly decreased. In chronic liver injury, Fabp7 knockout mice showed less fibrogenic response to carbon tetrachloride compared with wild-type mice. Taken together, FABP7 is involved in the liver injury process through its regulation of KC phagocytic activity and cytokine production. Such modulation of KC function by FABP7 may provide a novel therapeutic approach to the treatment of liver diseases.
Collapse
Affiliation(s)
- Hirofumi Miyazaki
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Tomoo Sawada
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Miwa Kiyohira
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Zhiqian Yu
- Department of Molecular Regulation, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Keiji Nakamura
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Yuki Yasumoto
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Yoshiteru Kagawa
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Majid Ebrahimi
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Ariful Islam
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Kazem Sharifi
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Saki Kawamura
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Takanori Kodama
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Yui Yamamoto
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Yasuhiro Adachi
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Nobuko Tokuda
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan; Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shuji Terai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Isao Sakaida
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Toshizo Ishikawa
- Division of Neurosciences, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Graduate School of Medicine, Yamaguchi University, Ube, Japan.
| |
Collapse
|
46
|
Waltmann MD, Basford JE, Konaniah ES, Weintraub NL, Hui DY. Apolipoprotein E receptor-2 deficiency enhances macrophage susceptibility to lipid accumulation and cell death to augment atherosclerotic plaque progression and necrosis. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1395-405. [PMID: 24840660 DOI: 10.1016/j.bbadis.2014.05.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 05/02/2014] [Accepted: 05/07/2014] [Indexed: 01/29/2023]
Abstract
Genome-wide association studies have linked LRP8 polymorphisms to premature coronary artery disease and myocardial infarction in humans. However, the mechanisms by which dysfunctions of apolipoprotein E receptor-2 (apoER2), the protein encoded by LRP8 gene, influence atherosclerosis have not been elucidated completely. The current study focused on the role of apoER2 in macrophages, a cell type that plays an important role in atherosclerosis. Results showed that apoER2-deficient mouse macrophages accumulated more lipids and were more susceptible to oxidized LDL (oxLDL)-induced death compared to control cells. Consistent with these findings, apoER2 deficient macrophages also displayed defective serum-induced Akt activation and higher levels of the pro-apoptotic protein phosphorylated p53. Furthermore, the expression and activation of peroxisome proliferator-activated receptor γ (PPARγ) were increased in apoER2-deficient macrophages. Deficiency of apoER2 in hypercholesterolemic LDL receptor-null mice (Lrp8(-/-)Ldlr(-/-) mice) also resulted in accelerated atherosclerosis with more complex lesions and extensive lesion necrosis compared to Lrp8(+/+)Ldlr(-/-) mice. The atherosclerotic plaques of Lrp8(-/-)Ldlr(-/-) mice displayed significantly higher levels of p53-positive macrophages, indicating that the apoER2-deficient macrophages contribute to the accelerated atherosclerotic lesion necrosis observed in these animals. Taken together, this study indicates that apoER2 in macrophages limits PPARγ expression and protects against oxLDL-induced cell death. Thus, abnormal apoER2 functions in macrophages may at least in part contribute to the premature coronary artery disease and myocardial infarction in humans with LRP8 polymorphisms. Moreover, the elevated PPARγ expression in apoER2-deficient macrophages suggests that LRP8 polymorphism may be a genetic modifier of cardiovascular risk with PPARγ therapy.
Collapse
Affiliation(s)
- Meaghan D Waltmann
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Joshua E Basford
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Eddy S Konaniah
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Neal L Weintraub
- Department of Internal Medicine, Division of Cardiovascular Disease, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA.
| |
Collapse
|
47
|
Kaczocha M, Rebecchi MJ, Ralph BP, Teng YHG, Berger WT, Galbavy W, Elmes MW, Glaser ST, Wang L, Rizzo RC, Deutsch DG, Ojima I. Inhibition of fatty acid binding proteins elevates brain anandamide levels and produces analgesia. PLoS One 2014; 9:e94200. [PMID: 24705380 PMCID: PMC3976407 DOI: 10.1371/journal.pone.0094200] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/12/2014] [Indexed: 11/23/2022] Open
Abstract
The endocannabinoid anandamide (AEA) is an antinociceptive lipid that is inactivated through cellular uptake and subsequent catabolism by fatty acid amide hydrolase (FAAH). Fatty acid binding proteins (FABPs) are intracellular carriers that deliver AEA and related N-acylethanolamines (NAEs) to FAAH for hydrolysis. The mammalian brain expresses three FABP subtypes: FABP3, FABP5, and FABP7. Recent work from our group has revealed that pharmacological inhibition of FABPs reduces inflammatory pain in mice. The goal of the current work was to explore the effects of FABP inhibition upon nociception in diverse models of pain. We developed inhibitors with differential affinities for FABPs to elucidate the subtype(s) that contributes to the antinociceptive effects of FABP inhibitors. Inhibition of FABPs reduced nociception associated with inflammatory, visceral, and neuropathic pain. The antinociceptive effects of FABP inhibitors mirrored their affinities for FABP5, while binding to FABP3 and FABP7 was not a predictor of in vivo efficacy. The antinociceptive effects of FABP inhibitors were mediated by cannabinoid receptor 1 (CB1) and peroxisome proliferator-activated receptor alpha (PPARα) and FABP inhibition elevated brain levels of AEA, providing the first direct evidence that FABPs regulate brain endocannabinoid tone. These results highlight FABPs as novel targets for the development of analgesic and anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Martin Kaczocha
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail:
| | - Mario J. Rebecchi
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York, United States of America
| | - Brian P. Ralph
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Yu-Han Gary Teng
- Department of Chemistry and the Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, United States of America
| | - William T. Berger
- Department of Chemistry and the Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, United States of America
| | - William Galbavy
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York, United States of America
| | - Matthew W. Elmes
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Sherrye T. Glaser
- Department of Biological Sciences, Kingsborough Community College, Brooklyn, New York, United States of America
| | - Liqun Wang
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Robert C. Rizzo
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York, United States of America
| | - Dale G. Deutsch
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Iwao Ojima
- Department of Chemistry and the Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, United States of America
| |
Collapse
|
48
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
49
|
Gally F, Kosmider B, Weaver MR, Pate KM, Hartshorn KL, Oberley-Deegan RE. FABP5 deficiency enhances susceptibility to H1N1 influenza A virus-induced lung inflammation. Am J Physiol Lung Cell Mol Physiol 2013; 305:L64-72. [PMID: 23624787 DOI: 10.1152/ajplung.00276.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The early inflammatory response to influenza A virus infection contributes to severe lung disease and continues to pose a serious threat to human health. The mechanisms by which inflammatory cells invade the respiratory tract remain unclear. Uncontrolled inflammation and oxidative stress cause lung damage in response to influenza A infection. We have previously shown that the fatty acid binding protein 5 (FABP5) has anti-inflammatory properties. We speculate that, as a transporter of fatty acids, FABP5 plays an important protective role against oxidative damage to lipids during infection as well. Using FABP5-/- and wild-type (WT) mice infected with influenza A virus, we showed that FABP5-/- mice had increased cell infiltration of macrophages and neutrophils compared with WT mice. FABP5-/- mice presented lower viral burden but lost as much weight as WT mice. The adaptive immune response was also increased in FABP5-/- mice as illustrated by the accumulation of T and B cells in the lung tissues and increased levels of H1N1-specific IgG antibodies. FABP5 deficiency greatly enhanced oxidative damage and lipid peroxidation following influenza A infection and presented with sustained tissue inflammation. Interestingly, FABP5 expression decreased following influenza A infection in WT lung tissues that corresponded to a decrease in the anti-inflammatory molecule PPAR-γ activity. In conclusion, our results demonstrate a previously unknown contribution of FABP5 to influenza A virus pathogenesis by controlling excessive oxidative damage and inflammation. This property could be exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Fabienne Gally
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | | | | | | | | | | |
Collapse
|
50
|
Nagy ZS, Czimmerer Z, Nagy L. Nuclear receptor mediated mechanisms of macrophage cholesterol metabolism. Mol Cell Endocrinol 2013; 368:85-98. [PMID: 22546548 DOI: 10.1016/j.mce.2012.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 04/05/2012] [Accepted: 04/10/2012] [Indexed: 10/28/2022]
Abstract
Macrophages comprise a family of multi-faceted phagocytic effector cells that differentiate "in situ" from circulating monocytes to exert various functions including clearance of foreign pathogens as well as debris derived from host cells. Macrophages also possess the ability to engulf and metabolize lipids and this way connect lipid metabolism and inflammation. The molecular link between these processes is provided by certain members of the nuclear receptor family. For instance, peroxisome proliferator activated receptors (PPAR) and liver X receptors (LXR) are able to sense the dynamically changing lipid environment and translate it to gene expression changes in order to modulate the cellular phenotype. Atherosclerosis embodies both sides of this coin: it is a disease in which macrophages with altered cholesterol metabolism keep the arteries in a chronically inflamed state. A large body of publications has accumulated during the past few decades describing the role of nuclear receptors in the regulation of macrophage cholesterol homeostasis, their contribution to the formation of atherosclerotic plaques and their crosstalk with inflammatory pathways. This review will summarize the most recent findings from this field narrowly focusing on the contribution of various nuclear receptors to macrophage cholesterol metabolism.
Collapse
Affiliation(s)
- Zsuzsanna S Nagy
- Department of Biochemistry and Molecular Biology, Research Center for Molecular Medicine, University of Debrecen Medical and Health Science Center, H-4032 Debrecen, Nagyerdei krt 98, Hungary.
| | | | | |
Collapse
|