1
|
Akbar H, Jarosinski KW. Temporal Dynamics of Purinergic Receptor Expression in the Lungs of Marek's Disease (MD) Virus-Infected Chickens Resistant or Susceptible to MD. Viruses 2024; 16:1130. [PMID: 39066292 PMCID: PMC11281646 DOI: 10.3390/v16071130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Marek's disease virus (MDV) is an economic concern for the poultry industry due to its poorly understood pathophysiology. Purinergic receptors (PRs) are potential therapeutic targets for viral infections, including herpesviruses, prompting our investigation into their role in MDV pathogenesis. The current study is part of an experimental series analyzing the expression of PRs during MDV infection. To address the early or short-acting P2 PR responses during natural MDV infection, we performed an "exposure" experiment where age-matched chickens were exposed to experimentally infected shedders to initiate natural infection. In addition, select non-PR regulatory gene responses were measured. Two groups of naïve contact chickens (n = 5/breed/time point) from MD-resistant (White Leghorns: WL) and -susceptible (Pure Columbian) chicken lines were housed separately with experimentally infected PC (×PC) and WL (×WL) chickens for 6 or 24 h. Whole lung lavage cells (WLLC) were collected, RNA was extracted, and RT-qPCR assays were used to measure specific PR responses. In addition, other potentially important markers in pathophysiology were measured. Our study revealed that WL chickens exhibited higher P1 PR expression during natural infection. WL chickens also showed higher expression of P1A3 and P2X3 at 6 and 24 h when exposed to PC-infected chickens. P2X5 and P2Y1 showed higher expression at 6 h, while P2Y5 showed higher expression at 6 and 24 h; regardless of the chicken line, PC chickens exhibited higher expression of P2X2, P2Y8, P2Y10, P2Y13, and P2Y14 when exposed to either group of infected chickens. In addition, MDV infection altered the expression of DDX5 in both WL and PC groups exposed to PC-infected birds only. However, irrespective of the source of exposure, BCL2 and ANGPTL4 showed higher expression in both WL and PC. The expression of STAT1A and STAT5A was influenced by time and breed, with major changes observed in STAT5A. CAT and SOD1 expression significantly increased in both WL and PC birds, regardless of the source of infection. GPX1 and GPX2 expression also increased in both WL and PC, although overall lower expression was observed in PC chickens at 24 h compared to 6 h. Our data suggest systemic changes in the host during early infection, indicated by the altered expression of PRs, DDX5, BCL2, ANGPTL4, and other regulatory genes during early MDV infection. The relative expression of these responses in PC and WL chickens suggests they may play a key role in their response to natural MDV infection in the lungs and long-term pathogenesis and survival.
Collapse
Affiliation(s)
| | - Keith W. Jarosinski
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA;
| |
Collapse
|
2
|
Zhang W, Zhu F, Zhu J, Liu K. Phospholipase D, a Novel Therapeutic Target Contributes to the Pathogenesis of Neurodegenerative and Neuroimmune Diseases. Anal Cell Pathol (Amst) 2024; 2024:6681911. [PMID: 38487684 PMCID: PMC10940030 DOI: 10.1155/2024/6681911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 01/10/2024] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Phospholipase D (PLD) is an enzyme that consists of six isoforms (PLD1-PLD6) and has been discovered in different organisms including bacteria, viruses, plants, and mammals. PLD is involved in regulating a wide range of nerve cells' physiological processes, such as cytoskeleton modulation, proliferation/growth, vesicle trafficking, morphogenesis, and development. Simultaneously, PLD, which also plays an essential role in the pathogenesis of neurodegenerative and neuroimmune diseases. In this review, family members, characterizations, structure, functions and related signaling pathways, and therapeutic values of PLD was summarized, then five representative diseases including Alzheimer disease (AD), Parkinson's disease (PD), etc. were selected as examples to tell the involvement of PLD in these neurological diseases. Notably, recent advances in the development of tools for studying PLD therapy envisaged novel therapeutic interventions. Furthermore, the limitations of PLD based therapy were also analyzed and discussed. The content of this review provided a thorough and reasonable basis for further studies to exploit the potential of PLD in the treatment of neurodegenerative and neuroimmune diseases.
Collapse
Affiliation(s)
- Weiwei Zhang
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Feiqi Zhu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China
| | - Jie Zhu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Kangding Liu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Abstract
Glycerophospholipids are major components of cellular membranes and provide important signaling molecules. Besides shaping membrane properties, some bind to specific receptors to activate biological pathways. Untangling the roles of individual glycerophospholipids requires clearly defined molecular species, a challenge that can be best addressed through chemical synthesis. However, glycerophospholipid syntheses are often lengthy due to the contrasting polarities found within these lipids. We now report a general strategy to quickly access glycerophospholipids via opening of a phosphate triester epoxide with carboxylic acids catalyzed by Jacobsen's Co(salen) complex. We show that this method can be applied to a variety of commercially available fatty acids, photoswitchable fatty acids, and other carboxylic acids to provide the corresponding glycerophosphate derivatives.
Collapse
Affiliation(s)
- Tufan K Mukhopadhyay
- Department of Chemistry, New York University, Silver Center, 31 Washington Place, New York, New York 10003, United States
| | - Dirk Trauner
- Department of Chemistry, College of Arts and Sciences, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104-6323, United States
| |
Collapse
|
4
|
Akbar H, Fasick JJ, Ponnuraj N, Jarosinski KW. Purinergic signaling during Marek's disease in chickens. Sci Rep 2023; 13:2044. [PMID: 36739336 PMCID: PMC9899245 DOI: 10.1038/s41598-023-29210-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Purinergic receptors (PRs) have been reported as potential therapeutic targets for many viral infections including herpesviruses, which urges the investigation into their role in Marek's disease (MD), a herpesvirus induced cancer in chickens that is an important pathogen for the poultry industry. MD is caused by MD virus (MDV) that has a similar viral life cycle as human varicella zoster virus in that it is shed from infected epithelial skin cells and enters the host through the respiratory route. In this report, PR responses during natural MDV infection and disease progression was examined in MD-resistant white Leghorns (WL) and MD-susceptible Pure Columbian (PC) chickens during natural infection. Whole lung lavage cells (WLLC) and liver tissue samples were collected from chickens infected but showing no clinical signs of MD (Infected) or presenting with clinical disease (Diseased). RNA was extracted followed by RT-qPCR analysis with gene specific primers against members of the P1, P2X, and P2Y PR families. Differential expression (p < 0.05) was observed in breed and disease conditions. Some PRs showed tissue specific expression (P1A1, P2X1, and P2X6 in WLLC) whereas others responded to MDV infection only in MD-susceptible (PC) chickens (P1A2A, P2X1, P2X5, P2X7). P2Y PRs had differential expression in both chicken lines in response to MDV infection and MD progression. This study is the first to our knowledge to examine PR responses during MDV infection and disease progression. These results suggest PR signaling may an important area of research for MDV replication and MD.
Collapse
Affiliation(s)
- Haji Akbar
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Julia J Fasick
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Nagendraprabhu Ponnuraj
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Keith W Jarosinski
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
5
|
Ikubo M, Uwamizu A, Chen L, Nakamura S, Sayama M, Kawana H, Otani Y, Kano K, Inoue A, Aoki J, Ohwada T. Isosteric Replacement of Ester Linkage of Lysophospholipids with Heteroaromatic Rings Retains Potency and Subtype Selectivity. Chem Pharm Bull (Tokyo) 2023; 71:584-615. [PMID: 37394607 DOI: 10.1248/cpb.c23-00250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Our group has reported various derivatives of lysophosphatidylserine (LysoPS) as potent and subtype-selective agonists for G-protein-coupled receptors (GPCRs). However, the ester linkage between the glycerol moiety and fatty acid or fatty acid surrogate is present in all of them. In order to develop these LysoPS analogs as drug candidates, appropriate pharmacokinetic consideration is essential. Here, we found that the ester bond of LysoPS is highly susceptible to metabolic degradation in mouse blood. Accordingly, we examined isosteric replacement of the ester linkage with heteroaromatic rings. The resulting compounds showed excellent retention of potency and receptor subtype selectivity, as well as increased metabolic stability in vitro.
Collapse
Affiliation(s)
- Masaya Ikubo
- Department of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Akiharu Uwamizu
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Luying Chen
- Department of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Sho Nakamura
- Department of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Misa Sayama
- Department of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Hiroki Kawana
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Yuko Otani
- Department of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Kuniyuki Kano
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Asuka Inoue
- Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University
- AMED-PRIME, Japan Science and Technology Corporation
- AMED-LEAP, Japan Science and Technology Corporation
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
- AMED-LEAP, Japan Science and Technology Corporation
- AMED-CREST, Japan Science and Technology Corporation
| | - Tomohiko Ohwada
- Department of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| |
Collapse
|
6
|
Agarwal P, Gordon S, Martinez FO. Foam Cell Macrophages in Tuberculosis. Front Immunol 2022; 12:775326. [PMID: 34975863 PMCID: PMC8714672 DOI: 10.3389/fimmu.2021.775326] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022] Open
Abstract
Mycobacterium tuberculosis infects primarily macrophages in the lungs. Infected macrophages are surrounded by other immune cells in well organised structures called granulomata. As part of the response to TB, a type of macrophage loaded with lipid droplets arises which we call Foam cell macrophages. They are macrophages filled with lipid laden droplets, which are synthesised in response to increased uptake of extracellular lipids, metabolic changes and infection itself. They share the appearance with atherosclerosis foam cells, but their lipid contents and roles are different. In fact, lipid droplets are immune and metabolic organelles with emerging roles in Tuberculosis. Here we discuss lipid droplet and foam cell formation, evidence regarding the inflammatory and immune properties of foam cells in TB, and address gaps in our knowledge to guide further research.
Collapse
Affiliation(s)
- Pooja Agarwal
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Fernando O Martinez
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
7
|
G-protein-coupled receptor P2Y10 facilitates chemokine-induced CD4 T cell migration through autocrine/paracrine mediators. Nat Commun 2021; 12:6798. [PMID: 34815397 PMCID: PMC8611058 DOI: 10.1038/s41467-021-26882-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
G-protein-coupled receptors (GPCRs), especially chemokine receptors, play a central role in the regulation of T cell migration. Various GPCRs are upregulated in activated CD4 T cells, including P2Y10, a putative lysophospholipid receptor that is officially still considered an orphan GPCR, i.e., a receptor with unknown endogenous ligand. Here we show that in mice lacking P2Y10 in the CD4 T cell compartment, the severity of experimental autoimmune encephalomyelitis and cutaneous contact hypersensitivity is reduced. P2Y10-deficient CD4 T cells show normal activation, proliferation and differentiation, but reduced chemokine-induced migration, polarization, and RhoA activation upon in vitro stimulation. Mechanistically, CD4 T cells release the putative P2Y10 ligands lysophosphatidylserine and ATP upon chemokine exposure, and these mediators induce P2Y10-dependent RhoA activation in an autocrine/paracrine fashion. ATP degradation impairs RhoA activation and migration in control CD4 T cells, but not in P2Y10-deficient CD4 T cells. Importantly, the P2Y10 pathway appears to be conserved in human T cells. Taken together, P2Y10 mediates RhoA activation in CD4 T cells in response to auto-/paracrine-acting mediators such as LysoPS and ATP, thereby facilitating chemokine-induced migration and, consecutively, T cell-mediated diseases.
Collapse
|
8
|
Abstract
Free fatty acids (FFAs) are implicated in the pathogenesis of metabolic diseases that includes obesity, type 2 diabetes mellitus, and cardiovascular disease (CVD). FFAs serve as ligands for free fatty acid receptors (FFARs) that belong to the family of rhodopsin-like G protein-coupled receptors (GPCRs) and are expressed throughout the body to maintain energy homeostasis under changing nutritional conditions. Free fatty acid receptor 4 (FFAR4), also known as G protein-coupled receptor 120, is a long-chain fatty acid receptor highly expressed in adipocytes, endothelial cells, and macrophages. Activation of FFAR4 helps maintain metabolic homeostasis by regulating adipogenesis, insulin sensitivity, and inflammation. Furthermore, dysfunction of FFAR4 is associated with insulin resistance, obesity, and eccentric remodeling in both humans and mice, making FFAR4 an attractive therapeutic target for treating or preventing metabolic diseases. While much of the previous literature on FFAR4 has focused on its role in obesity and diabetes, recent studies have demonstrated that FFAR4 may also play an important role in the development of atherosclerosis and CVD. Most notably, FFAR4 activation reduces monocyte-endothelial cell interaction, enhances cholesterol efflux from macrophages, reduces lesion size in atherogenic mouse models, and stimulates oxylipin production in myocytes that functions in a feed-forward cardioprotective mechanism. This review will focus on the role of FFAR4 in metabolic diseases and highlights an underappreciated role of FFAR4 in the development of atherosclerosis and CVD.
Collapse
Affiliation(s)
- Gage M Stuttgen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daisy Sahoo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Division of Endocrinology & Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
9
|
Park SJ, Im DS. 2-Arachidonyl-lysophosphatidylethanolamine Induces Anti-Inflammatory Effects on Macrophages and in Carrageenan-Induced Paw Edema. Int J Mol Sci 2021; 22:ijms22094865. [PMID: 34064436 PMCID: PMC8125189 DOI: 10.3390/ijms22094865] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 11/16/2022] Open
Abstract
2-Arachidonyl-lysophosphatidylethanolamine, shortly 2-ARA-LPE, is a polyunsaturated lysophosphatidylethanolamine. 2-ARA-LPE has a very long chain arachidonic acid, formed by an ester bond at the sn-2 position. It has been reported that 2-ARA-LPE has anti-inflammatory effects in a zymosan-induced peritonitis model. However, it’s action mechanisms are poorly investigated. Recently, resolution of inflammation is considered to be an active process driven by M2 polarized macrophages. Therefore, we have investigated whether 2-ARA-LPE acts on macrophages for anti-inflammation, whether 2-ARA-LPE modulates macrophage phenotypes to reduce inflammation, and whether 2-ARA-LPE is anti-inflammatory in a carrageenan-induced paw edema model. In mouse peritoneal macrophages, 2-ARA-LPE was found to inhibit lipopolysaccharide (LPS)-induced M1 macrophage polarization, but not induce M2 polarization. 2-ARA-LPE inhibited the inductions of inducible nitric oxide synthase and cyclooxygenase-2 in mouse peritoneal macrophages at the mRNA and protein levels. Furthermore, products of the two genes, nitric oxide and prostaglandin E2, were also inhibited by 2-ARA-LPE. However, 1-oleoyl-LPE did not show any activity on the macrophage polarization and inflammatory responses. The anti-inflammatory activity of 2-ARA-LPE was also verified in vivo in a carrageenan-induced paw edema model. 2-ARA-LPE inhibits LPS-induced M1 polarization, which contributes to anti-inflammation and suppresses the carrageenan-induced paw edema in vivo.
Collapse
Affiliation(s)
- Soo-Jin Park
- College of Pharmacy, Pusan National University, Busan 46241, Korea;
| | - Dong-Soon Im
- College of Pharmacy, Pusan National University, Busan 46241, Korea;
- Laboratory of Pharmacology, College of Pharmacy, and Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Correspondence: ; Tel.: 82-2-961-9377
| |
Collapse
|
10
|
Lee JH, Im DS. 4-CMTB Ameliorates Ovalbumin-Induced Allergic Asthma through FFA2 Activation in Mice. Biomol Ther (Seoul) 2021; 29:427-433. [PMID: 33875623 PMCID: PMC8255137 DOI: 10.4062/biomolther.2020.176] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/27/2022] Open
Abstract
Free fatty acid receptor 2 (FFA2, also known as GPR43), a G-protein-coupled receptor, has been known to recognize short-chain fatty acids and regulate inflammatory responses. FFA2 gene deficiency exacerbated disease states in several models of inflammatory conditions including asthma. However, in vivo efficacy of FFA2 agonists has not been tested in allergic asthma. Thus, we investigated effect of 4-chloro-α-(1-methylethyl)-N-2-thiazoylylbenzeneacetanilide (4-CMTB), a FFA2 agonist, on antigen-induced degranulation in RBL-2H3 cells and ovalbumin-induced allergic asthma in BALB/c mice. Treatment of 4-CMTB inhibited the antigen-induced degranulation concentration-dependently. Administration of 4-CMTB decreased the immune cell numbers in the bronchoalveolar lavage fluid and suppressed the expression of inflammatory Th2 cytokines (IL-4, IL-5, and IL-13) in the lung tissues. Histological studies revealed that 4-CMTB suppressed mucin production and inflammation in the lungs. Thus, results proved that FFA2 functions to suppress allergic asthma, suggesting 4-CMTB activation of FFA2 as a therapeutic tool for allergic asthma.
Collapse
Affiliation(s)
- Ju-Hyun Lee
- Laboratory of Pharmacology, College of Pharmacy, and Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dong-Soon Im
- Laboratory of Pharmacology, College of Pharmacy, and Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
11
|
Son SE, Kim NJ, Im DS. Development of Free Fatty Acid Receptor 4 (FFA4/GPR120) Agonists in Health Science. Biomol Ther (Seoul) 2021; 29:22-30. [PMID: 33372166 PMCID: PMC7771848 DOI: 10.4062/biomolther.2020.213] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Till the 21st century, fatty acids were considered as merely building blocks for triglycerides, phospholipids, or cholesteryl esters. However, the discovery of G protein-coupled receptors (GPCRs) for free fatty acids at the beginning of the 21st century challenged that idea and paved way for a new field of research, merged into the field of receptor pharmacology for intercellular lipid mediators. Among the GPCRs for free fatty acids, free fatty acid receptor 4 (FFA4, also known as GPR120) recognizes long-chain polyunsaturated fatty acids such as DHA and EPA. It is significant in drug discovery because it regulates obesity-induced metaflammation and GLP-1 secretion. Our study reviews information on newly developed FFA4 agonists and their application in pathophysiologic studies and drug discovery. It also offers a potency comparison of the FFA4 agonists in an AP-TGF-α shedding assay.
Collapse
Affiliation(s)
- So-Eun Son
- Department of Pharmacy, College of Pharmacy, and Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Nam-Jung Kim
- Department of Pharmacy, College of Pharmacy, and Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dong-Soon Im
- Department of Pharmacy, College of Pharmacy, and Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
12
|
Sênos Demarco R, Clémot M, Jones DL. The impact of ageing on lipid-mediated regulation of adult stem cell behavior and tissue homeostasis. Mech Ageing Dev 2020; 189:111278. [PMID: 32522455 DOI: 10.1016/j.mad.2020.111278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/05/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023]
Abstract
Adult stem cells sustain tissue homeostasis throughout life and provide an important reservoir of cells capable of tissue repair in response to stress and tissue damage. Age-related changes to stem cells and/or the specialized niches that house them have been shown to negatively impact stem cell maintenance and activity. In addition, metabolic inputs have surfaced as another crucial layer in the control of stem cell behavior (Chandel et al., 2016; Folmes and Terzic, 2016; Ito and Suda, 2014; Mana et al., 2017; Shyh-Chang and Ng, 2017). Here, we will present a brief review of how lipid metabolism influences adult stem cell behavior under homeostatic conditions and speculate on how changes in lipid metabolism may impact stem cell ageing. This review considers the future of lipid metabolism research in stem cells, with the long-term goal of identifying mechanisms that could be targeted to counter or slow the age-related decline in stem cell function.
Collapse
Affiliation(s)
- Rafael Sênos Demarco
- Department of Molecular, Cell and Developmental Biology, Los Angeles, CA, 90095, USA
| | - Marie Clémot
- Department of Molecular, Cell and Developmental Biology, Los Angeles, CA, 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - D Leanne Jones
- Department of Molecular, Cell and Developmental Biology, Los Angeles, CA, 90095, USA; Molecular Biology Institute, Los Angeles, CA, 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
13
|
Im DS. Maresin-1 resolution with RORα and LGR6. Prog Lipid Res 2020; 78:101034. [DOI: 10.1016/j.plipres.2020.101034] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/17/2020] [Accepted: 04/24/2020] [Indexed: 12/14/2022]
|
14
|
Inhibition of store-operated calcium channels by N-arachidonoyl glycine (NAGly): no evidence for the involvement of lipid-sensing G protein coupled receptors. Sci Rep 2020; 10:2649. [PMID: 32060392 PMCID: PMC7021695 DOI: 10.1038/s41598-020-59565-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 01/28/2020] [Indexed: 01/08/2023] Open
Abstract
N-arachidonoyl glycine (NAGly) is an endogenous lipid deriving from the endocannabinoid anandamide (AEA). Identified as a ligand of several G-protein coupled receptors (GPCRs), it can however exert biological responses independently of GPCRs. NAGly was recently shown to depress store-operated Ca2+ entry (SOCE) but its mechanism of action remains elusive. The major aim of this study was to gain a better knowledge on the NAGly-dependent impairment of SOCE in neurons of the central nervous system (CNS) from mice. First, we examined the expression of genes encoding for putative lipid sensing GPCRs using transcriptomic data publicly available. This analysis showed that the most abundant GPCRs transcripts present in the cerebral cortices of embryonic brains were coding for lysophosphatidic acid (LPA) and sphingosine-1 phosphate (S1P) receptors. Next, the presence of functional receptors was assessed with live-cell calcium imaging experiments. In primary cortical cells S1P and LPA mobilize Ca2+ from internal stores via a mechanism sensitive to the S1P and LPA receptor antagonists Ex26, H2L5186303, or Ki16425. However, none of these compounds prevented or attenuated the NAGly-dependent impairment of SOCE. We found no evidence for the requirement of lipid sensing GPCRs in this inhibitory process, indicating that NAGly is an endogenous modulator interfering with the core machinery of SOCE. Moreover, these data also raise the intriguing possibility that the depression of SOCE could play a role in the central effects of NAGly.
Collapse
|
15
|
Spatiotemporal dynamic monitoring of fatty acid-receptor interaction on single living cells by multiplexed Raman imaging. Proc Natl Acad Sci U S A 2020; 117:3518-3527. [PMID: 32015136 DOI: 10.1073/pnas.1916238117] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Numerous fatty acid receptors have proven to play critical roles in normal physiology. Interactions among these receptor types and their subsequent membrane trafficking has not been fully elucidated, due in part to the lack of efficient tools to track these cellular events. In this study, we fabricated the surface-enhanced Raman scattering (SERS)-based molecular sensors for detection of two putative fatty acid receptors, G protein-coupled receptor 120 (GPR120) and cluster of differentiation 36 (CD36), in a spatiotemporal manner in single cells. These SERS probes allowed multiplex detection of GPR120 and CD36, as well as a peak that represented the cell. This multiplexed sensing system enabled the real-time monitoring of fatty acid-induced receptor activation and dynamic distributions on the cell surface, as well as tracking of the receptors' internalization processes on the addition of fatty acid. Increased SERS signals were seen in engineered HEK293 cells with higher fatty acid concentrations, while decreased responses were found in cell line TBDc1, suggesting that the endocytic process requires innate cellular components. SERS mapping results confirm that GPR120 is the primary receptor and may work synergistically with CD36 in sensing polyunsaturated fatty acids and promoting Ca2+ mobilization, further activating the process of fatty acid uptake. The ability to detect receptors' locations and monitor fatty acid-induced receptor redistribution demonstrates the specificity and potential of our multiplexed SERS imaging platform in the study of fatty acid-receptor interactions and might provide functional information for better understanding their roles in fat intake and development of fat-induced obesity.
Collapse
|
16
|
Bartoszek A, Moo EV, Binienda A, Fabisiak A, Krajewska JB, Mosińska P, Niewinna K, Tarasiuk A, Martemyanov K, Salaga M, Fichna J. Free Fatty Acid Receptors as new potential therapeutic target in inflammatory bowel diseases. Pharmacol Res 2019; 152:104604. [PMID: 31846762 DOI: 10.1016/j.phrs.2019.104604] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/19/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023]
Abstract
Family of Free Fatty Acid Receptors (FFARs), specific G protein-coupled receptors comprises of four members: FFAR1-4, where each responds to different chain length of fatty acids (FAs). Over the years, FFARs have become attractive pharmacological targets in the treatment of type 2 diabetes, metabolic syndrome, cardiovascular diseases and asthma; recent studies also point to their role in inflammation. It is now well-established that activation of FFAR1 and FFAR4 by long and medium chain FAs may lead to reduction of inflammatory state; FFAR2 and FFAR3 are activated by short chain FAs, but only FFAR2 was shown to alleviate inflammation, mostly by neutrophil inhibition. All FFARs have thus been proposed as targets in inflammatory bowel diseases (IBD). Here we discuss current knowledge and future directions in FFAR research related to IBD.
Collapse
Affiliation(s)
- Adrian Bartoszek
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Ee Von Moo
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Agata Binienda
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Adam Fabisiak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland; Department of Digestive Tract Diseases, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Julia B Krajewska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Paula Mosińska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Karolina Niewinna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Aleksandra Tarasiuk
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Kirill Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Maciej Salaga
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
17
|
Bae IS, Kim SH. Expression and Secretion of an Atrial Natriuretic Peptide in Beige-Like 3T3-L1 Adipocytes. Int J Mol Sci 2019; 20:ijms20246128. [PMID: 31817347 PMCID: PMC6940835 DOI: 10.3390/ijms20246128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 01/08/2023] Open
Abstract
The browning of white adipose tissue (beige adipocytes) stimulates energy expenditure. Omega-3 fatty acids have been shown to induce thermogenic action in adipocytes via G-protein coupled receptor 120 (GPR120). Atrial natriuretic peptide (ANP) is a peptide hormone that plays the role of maintaining normal blood pressure in kidneys to inhibit Na+ reuptake. Recently, ANP was found to induce adipocyte browning by binding to NPR1, an ANP receptor. However, the expression of ANP in adipocytes has not yet been studied. Therefore, in this study, we investigate the expression of ANP in beige-like adipocytes induced by docosahexaenoic acids (DHA), T3, or a PPAR agonist, rosiglitazone. First, we found that brown adipocyte-specific genes were upregulated in beige-like adipocytes. DHA promoted ANP expression in beige-like cells, whereas DHA-induced ANP expression was abolished by GPR120 knockout. ANP secretion of beige-like adipocytes was increased via PKC/ERK1/2 signaling in the GPR120 pathway. Furthermore, ANP secreted from beige-like adipocytes acted on HEK-293 cells, the recipient cells, leading to increased cGMP activity. After the NPR1 knockdown of HEK-293 cells, cGMP activity was not changed. Taken together, our findings indicate that beige-like adipocytes induce ANP secretion, which may contribute to improving obesity-associated metabolic disease.
Collapse
|
18
|
Gut microbiota confers host resistance to obesity by metabolizing dietary polyunsaturated fatty acids. Nat Commun 2019; 10:4007. [PMID: 31488836 PMCID: PMC6728375 DOI: 10.1038/s41467-019-11978-0] [Citation(s) in RCA: 223] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/15/2019] [Indexed: 12/15/2022] Open
Abstract
Gut microbiota mediates the effects of diet, thereby modifying host metabolism and the incidence of metabolic disorders. Increased consumption of omega-6 polyunsaturated fatty acid (PUFA) that is abundant in Western diet contributes to obesity and related diseases. Although gut-microbiota-related metabolic pathways of dietary PUFAs were recently elucidated, the effects on host physiological function remain unclear. Here, we demonstrate that gut microbiota confers host resistance to high-fat diet (HFD)-induced obesity by modulating dietary PUFAs metabolism. Supplementation of 10-hydroxy-cis-12-octadecenoic acid (HYA), an initial linoleic acid-related gut-microbial metabolite, attenuates HFD-induced obesity in mice without eliciting arachidonic acid-mediated adipose inflammation and by improving metabolic condition via free fatty acid receptors. Moreover, Lactobacillus-colonized mice show similar effects with elevated HYA levels. Our findings illustrate the interplay between gut microbiota and host energy metabolism via the metabolites of dietary omega-6-FAs thereby shedding light on the prevention and treatment of metabolic disorders by targeting gut microbial metabolites. The gut microbiome is an important regulator of metabolic health. Here the authors show that intestinal bacteria metabolize dietary linoleic acid to 10-hydroxy-cis-12-octadecenoic acid (HYA) which confers host resistance to high fat diet-induced obesity in mice.
Collapse
|
19
|
Gerstmeier J, Seegers J, Witt F, Waltenberger B, Temml V, Rollinger JM, Stuppner H, Koeberle A, Schuster D, Werz O. Ginkgolic Acid is a Multi-Target Inhibitor of Key Enzymes in Pro-Inflammatory Lipid Mediator Biosynthesis. Front Pharmacol 2019; 10:797. [PMID: 31379572 PMCID: PMC6650749 DOI: 10.3389/fphar.2019.00797] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/20/2019] [Indexed: 12/19/2022] Open
Abstract
Introduction: Lipid mediators (LMs) comprise bioactive metabolites of polyunsaturated fatty acids, including pro-inflammatory prostaglandins (PGs), thromboxanes (TXs), and leukotrienes (LTs), as well as specialized pro-resolving mediators (SPMs). They are essentially biosynthesized via cyclooxygenase (COX) and lipoxygenase (LO) pathways in complex networks and regulate the progression as well as the resolution of inflammatory disorders including inflammation-triggered cancer. Ginkgolic acid (GA) is a phenolic acid contained in Ginkgo biloba L. with neuroprotective, antimicrobial, and antitumoral properties. Although LMs regulate microbial infections and tumor progression, whether GA affects LM biosynthesis is unknown and was investigated here in detail. Methods: Pharmacophore-based virtual screening was performed along with docking simulations. Activity assays were conducted for isolated human recombinant 5-LO, cytosolic phospholipase (PLA)2α, COX-2, and ovine COX-1. The activity of human mPGES-1 and thromboxane A2 synthase (TXAS) was determined in crude cellular fractions. Cellular LM formation was studied using human monocytes, neutrophils, platelets, and M1- and M2-like macrophages. LMs were identified after (ultra)high-performance liquid chromatography by UV detection or ESI-tandem mass spectrometry. Results: GA was identified as virtual hit in an mPGES-1 pharmacophore-based virtual screening. Cell-free assays revealed potent suppression of mPGES-1 activity (IC50 = 0.7 µM) that is fully reversible and essentially independent of the substrate concentration. Moreover, cell-free assays revealed COX-1 and TXAS as additional targets of GA with lower affinity (IC50 = 8.1 and 5.2 µM). Notably, 5-LO, the key enzyme in LT biosynthesis, was potently inhibited by GA (IC50 = 0.2 µM) in a reversible and substrate-independent manner. Docking simulations support the molecular interaction of GA with mPGES-1 and 5-LO and suggest concrete binding sites. Interestingly, interference of GA with mPGES-1, COX-1, TXAS, and 5-LO was evident also in intact cells with IC50 values of 2.1-3.8 µM; no radical scavenging or cytotoxic properties were obvious. Analysis of LM profiles from bacteria-stimulated human M1- and M2-like macrophages confirmed the multi-target features of GA and revealed LM redirection towards the formation of 12-/15-LO products including SPM. Conclusions: We reveal GA as potent multi-target inhibitor of key enzymes in the biosynthesis of pro-inflammatory LMs that contribute to the complex pharmacological and toxicological properties of GA.
Collapse
Affiliation(s)
- Jana Gerstmeier
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Julia Seegers
- Department of Pharmaceutical Analytics, Pharmaceutical Institute, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | - Finja Witt
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Veronika Temml
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Judith M. Rollinger
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Hermann Stuppner
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Andreas Koeberle
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Daniela Schuster
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Oliver Werz
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
20
|
Takemasu S, Ito M, Morioka S, Nigorikawa K, Kofuji S, Takasuga S, Eguchi S, Nakanishi H, Matsuoka I, Sasaki J, Sasaki T, Hazeki K. Lysophosphatidylinositol-acyltransferase-1 is involved in cytosolic Ca 2+ oscillations in macrophages. Genes Cells 2019; 24:366-376. [PMID: 30851234 DOI: 10.1111/gtc.12681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 12/18/2022]
Abstract
Lysophosphatidylinositol-acyltransferase-1 (LPIAT1) specifically catalyzes the transfer of arachidonoyl-CoA to lysophosphoinositides. LPIAT-/- mice have been shown to have severe defects in the brain and liver; however, the exact molecular mechanisms behind these conditions are not well understood. As immune cells have been implicated in liver inflammation based on disfunction of LPIAT1, we generated Raw264.7 macrophages deficient in LPIAT1, using shRNA and CRISPR/Cas9. The amount of C38:4 species in phosphoinositides, especially in PtdInsP2 , was remarkably decreased in these cells. Unlike in wild-type cells, LPIAT1-deficient cells showed prolonged oscillations of intracellular Ca2+ upon UDP stimulation, which is known to activate phospholipase Cβ through the Gq-coupled P2Y6 receptor, even in the absence of extracellular Ca2+ . It is speculated that the prolonged Ca2+ response may be relevant to the increased risk of liver inflammation induced by LPIAT1 disfunction.
Collapse
Affiliation(s)
- Shinya Takemasu
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masaaki Ito
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Japan
| | - Shin Morioka
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kiyomi Nigorikawa
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Satoshi Kofuji
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shunsuke Takasuga
- Department of Pathology and Immunology, Akita University School of Medicine, Akita, Japan
| | - Satoshi Eguchi
- Department of Pathology and Immunology, Akita University School of Medicine, Akita, Japan
| | - Hiroki Nakanishi
- Department of Pathology and Immunology, Akita University School of Medicine, Akita, Japan
| | - Isao Matsuoka
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Japan
| | - Junko Sasaki
- Department of Pathology and Immunology, Akita University School of Medicine, Akita, Japan.,Department of Biochemical Pathophysiology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Japan
| | - Takehiko Sasaki
- Department of Pathology and Immunology, Akita University School of Medicine, Akita, Japan.,Department of Biochemical Pathophysiology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Japan
| | - Kaoru Hazeki
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
21
|
Papsdorf K, Brunet A. Linking Lipid Metabolism to Chromatin Regulation in Aging. Trends Cell Biol 2019; 29:97-116. [PMID: 30316636 PMCID: PMC6340780 DOI: 10.1016/j.tcb.2018.09.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022]
Abstract
The lifespan of an organism is strongly influenced by environmental factors (including diet) and by internal factors (notably reproductive status). Lipid metabolism is critical for adaptation to external conditions or reproduction. Interestingly, specific lipid profiles are associated with longevity, and increased uptake of certain lipids extends longevity in Caenorhabditis elegans and ameliorates disease phenotypes in humans. How lipids impact longevity, and how lipid metabolism is regulated during aging, is just beginning to be unraveled. This review describes recent advances in the regulation and role of lipids in longevity, focusing on the interaction between lipid metabolism and chromatin states in aging and age-related diseases.
Collapse
Affiliation(s)
- Katharina Papsdorf
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA; Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
22
|
Srikanth M, Chew WS, Hind T, Lim SM, Hay NWJ, Lee JHM, Rivera R, Chun J, Ong WY, Herr DR. Lysophosphatidic acid and its receptor LPA1 mediate carrageenan induced inflammatory pain in mice. Eur J Pharmacol 2018; 841:49-56. [DOI: 10.1016/j.ejphar.2018.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/28/2018] [Accepted: 10/09/2018] [Indexed: 10/28/2022]
|
23
|
Fumagalli M, Lecca D, Coppolino GT, Parravicini C, Abbracchio MP. Pharmacological Properties and Biological Functions of the GPR17 Receptor, a Potential Target for Neuro-Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1051:169-192. [PMID: 28828731 DOI: 10.1007/5584_2017_92] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In 2006, cells heterologously expressing the "orphan" receptor GPR17 were shown to acquire responses to both uracil nucleotides and cysteinyl-leukotrienes, two families of signaling molecules accumulating in brain or heart as a result of hypoxic/traumatic injuries. In subsequent years, evidence of GPR17 key role in oligodendrogenesis and myelination has highlighted it as a "model receptor" for new therapies in demyelinating and neurodegenerative diseases. The apparently contrasting evidence in the literature about the role of GPR17 in promoting or inhibiting myelination can be due to its transient expression in the intermediate stages of differentiation, exerting a pro-differentiating function in early oligodendrocyte precursor cells (OPCs), and an inhibitory role in late stage maturing cells. Meanwhile, several papers extended the initial data on GPR17 pharmacology, highlighting a "promiscuous" behavior of this receptor; indeed, GPR17 is able to respond to other emergency signals like oxysterols or the pro-inflammatory cytokine SDF-1, underlying GPR17 ability to adapt its responses to changes of the surrounding extracellular milieu, including damage conditions. Here, we analyze the available literature on GPR17, in an attempt to summarize its emerging biological roles and pharmacological properties.
Collapse
Affiliation(s)
- Marta Fumagalli
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Giusy T Coppolino
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Chiara Parravicini
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
24
|
Xie J, Gurler Main H, Sacks JD, Muralidhar GG, Barbolina MV. Regulation of DNA damage repair and lipid uptake by CX 3CR1 in epithelial ovarian carcinoma. Oncogenesis 2018; 7:37. [PMID: 29712888 PMCID: PMC5928120 DOI: 10.1038/s41389-018-0046-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/17/2018] [Accepted: 03/26/2018] [Indexed: 12/25/2022] Open
Abstract
Failure of currently used cytotoxic chemotherapy is one of the main reasons behind high mortality from metastatic high grade serous ovarian carcinoma. We found that high expression of a receptor for fractalkine (CX3CR1) significantly correlated with shorter survival of patients with serous ovarian carcinoma treated with cytotoxic DNA damage chemotherapies, and reduction of CX3CR1 expression resulted in sensitization to several DNA damaging modalities, including x-ray radiation and cisplatin. Here, we show that CX3CR1 plays a role in double-strand DNA break response and repair by regulating expression of RAD50 by a MYC-dependent mechanism. We demonstrate that downregulation of CX3CR1 alone and in a combination with irradiation affects peritoneal metastasis in an organ-specific manner; we show that CX3CR1 regulates lipid uptake which could control omental metastasis. This study identifies CX3CR1 as a novel potential target for sensitization of ovarian carcinoma to DNA damage therapies and reduction of peritoneal carcinomatosis.
Collapse
Affiliation(s)
- Jia Xie
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Hilal Gurler Main
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Joelle D Sacks
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Goda G Muralidhar
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Maria V Barbolina
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
25
|
Im DS. FFA4 (GPR120) as a fatty acid sensor involved in appetite control, insulin sensitivity and inflammation regulation. Mol Aspects Med 2017; 64:92-108. [PMID: 28887275 DOI: 10.1016/j.mam.2017.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/03/2017] [Accepted: 09/03/2017] [Indexed: 12/19/2022]
Abstract
Unsaturated long-chain fatty acids have been suggested to be beneficial in the context of cardiovascular disorders based in epidemiologic studies conducted in Greenland and Mediterranean. DHA and EPA are omega-3 polyunsaturated fatty acids that are plentiful in fish oil, and oleic acid is an omega-9 monounsaturated fatty acid, rich in olive oil. Dietary intake of these unsaturated long-chain fatty acids have been associated with insulin sensitivity and weight loss, which contrasts with the impairment of insulin sensitivity and weight gain associated with high intakes of saturated long-chain fatty acids. The recent discovery that free fatty acid receptor 4 (FFA4, also known as GPR120) acts as a sensor for unsaturated long-chain fatty acids started to unveil the molecular mechanisms underlying the beneficial functions played by these unsaturated long-chain fatty acids in various physiological processes, which include the secretions of gastrointestinal peptide hormones and glucose homeostasis. In this review, the physiological roles and therapeutic significance of FFA4 in appetite control, insulin sensitization, and inflammation reduction are discussed in relation to obesity and type 2 diabetes from pharmacological viewpoints.
Collapse
Affiliation(s)
- Dong-Soon Im
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
26
|
Lu J, Fang B, Zheng Y, Yu X, Huang G, Wang Z, Deng X, Guan S. 1,3-dichloro-2-propanol induced lipid accumulation in HepG2 cells through cAMP/protein kinase A and AMP-activated protein kinase pathways via Gi/o-coupled receptors. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 55:118-126. [PMID: 28843851 DOI: 10.1016/j.etap.2017.07.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/26/2017] [Accepted: 07/25/2017] [Indexed: 06/07/2023]
Abstract
1,3-dichloro-2-propanol (1,3-DCP) is a food born hepatoxic chloropropanol contaminant that has been detected in a wide range of foods. In the present study, we investigated the effects and mechanisms of 1,3-DCP on lipid accumulation in HepG2 cells. The data showed 1,3-DCP significantly increased intracellular content of triglyceride (TG) and total cholesterol (TC) at 0.5-2μg/mL. Further results showed that 1,3-DCP greatly decreased cyclic AMP (cAMP) level. In addition, 1,3-DCP inhibited PKA and AMPK signaling pathway, but had no influence on intracellular calcium and regulated proteins. Moreover, Gi/o protein inhibitor PTX significantly inhibited 1,3-DCP induced decrease of cAMP, p-PKA and p-AMPK expression. Furthermore, 1,3-DCP significantly decreased GPR41 and GPR43 expression, but had no effect on GPR109B.Thus, we concluded that 1,3-DCP induced lipid accumulation in HepG2 cells through cAMP/PKA and AMPK signaling pathways via Gi/o-coupled receptor.
Collapse
Affiliation(s)
- Jing Lu
- Department of food quality and safety, Jilin University, Changchun, Jilin 130062, People's Republic of China; College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Baochen Fang
- Department of food quality and safety, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Yiying Zheng
- Department of food quality and safety, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Xin Yu
- Department of food quality and safety, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Guoren Huang
- Department of food quality and safety, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Zhenning Wang
- Department of food quality and safety, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Xuming Deng
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Shuang Guan
- Department of food quality and safety, Jilin University, Changchun, Jilin 130062, People's Republic of China; College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China.
| |
Collapse
|
27
|
Troupiotis-Tsaïlaki A, Zachmann J, González-Gil I, Gonzalez A, Ortega-Gutiérrez S, López-Rodríguez ML, Pardo L, Govaerts C. Ligand chain length drives activation of lipid G protein-coupled receptors. Sci Rep 2017; 7:2020. [PMID: 28515494 PMCID: PMC5435731 DOI: 10.1038/s41598-017-02104-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/07/2017] [Indexed: 01/20/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a lipid mediator that can activate five cell membrane G protein-coupled receptors (GPCRs) which carry a variety of essential functions and are promising drug targets. S1P is composed of a polar zwitterionic head-group and a hydrophobic alkyl chain. This implies an activation mechanism of its cognate receptor that must be significantly different from what is known for prototypical GPCRs (ie receptor to small hydrophilic ligands). Here we aim to identify the structural features responsible for S1P agonism by combining molecular dynamics simulations and functional assays using S1P analogs of different alkyl chain lengths. We propose that high affinity binding involves polar interactions between the lipid head-group and receptor side chains while activation is due to hydrophobic interactions between the lipid tail and residues in a distinct binding site. We observe that ligand efficacy is directly related to alkyl chain length but also varies with receptor subtypes in correlation with the size of this binding pocket. Integrating experimental and computational data, we propose an activation mechanism for the S1P receptors involving agonist-induced conformational events that are conserved throughout class A GPCRs.
Collapse
Affiliation(s)
| | - Julian Zachmann
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Inés González-Gil
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain
| | - Angel Gonzalez
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Silvia Ortega-Gutiérrez
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain
| | - Maria L López-Rodríguez
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain
| | - Leonardo Pardo
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain.
| | - Cedric Govaerts
- Laboratoire de Structure et Fonction des Membranes Biologiques, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
28
|
Pédron N, Artigaud S, Infante JLZ, Le Bayon N, Charrier G, Pichereau V, Laroche J. Proteomic responses of European flounder to temperature and hypoxia as interacting stressors: Differential sensitivities of populations. THE SCIENCE OF THE TOTAL ENVIRONMENT 2017; 586:890-899. [PMID: 28215807 DOI: 10.1016/j.scitotenv.2017.02.068] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 01/30/2017] [Accepted: 02/07/2017] [Indexed: 06/06/2023]
Abstract
In the context of global change, ectotherms are increasingly impacted by abiotic perturbations. Along the distribution area of a species, the populations at low latitudes are particularly exposed to temperature increase and hypoxic events. In this study, we have compared the proteomic responses in the liver of European flounder populations, by using 2-D electrophoresis. One southern peripheral population from Portugal vs two northern core populations from France, were reared in a common garden experiment. Most of the proteomic differences were observed between the two experimental conditions, a cold vs a warm and hypoxic conditions. Consistent differentiations between populations were observed in accumulation of proteins involved in the bioenergetics- and methionine-metabolisms, fatty acids transport, and amino-acid catabolism. The specific regulation of crucial enzymes like ATP-synthase and G6PDH, in the liver of the southern population, could be related to a possible local adaptation. This southern peripheral population is spatially distant from northern core populations and has experienced dissimilar ecological conditions; thus it may contain genotypes that confer resilience to climate changes.
Collapse
Affiliation(s)
- Nicolas Pédron
- Université de Bretagne Occidentale, UMR 6539 CNRS/UBO/IRD/Ifremer, Laboratoire des Sciences de l'Environnement Marin LEMAR, Institut Universitaire Européen de la Mer IUEM, Plouzané, France; Ifremer, Laboratoire Adaptation, Reproduction et Nutrition des Poissons ARN, Unité de Physiologie Fonctionnelle des Organismes Marins PFOM, Plouzané, France
| | - Sébastien Artigaud
- Université de Bretagne Occidentale, UMR 6539 CNRS/UBO/IRD/Ifremer, Laboratoire des Sciences de l'Environnement Marin LEMAR, Institut Universitaire Européen de la Mer IUEM, Plouzané, France
| | - José-Luis Zambonino Infante
- Université de Bretagne Occidentale, UMR 6539 CNRS/UBO/IRD/Ifremer, Laboratoire des Sciences de l'Environnement Marin LEMAR, Institut Universitaire Européen de la Mer IUEM, Plouzané, France; Ifremer, Laboratoire Adaptation, Reproduction et Nutrition des Poissons ARN, Unité de Physiologie Fonctionnelle des Organismes Marins PFOM, Plouzané, France
| | - Nicolas Le Bayon
- Université de Bretagne Occidentale, UMR 6539 CNRS/UBO/IRD/Ifremer, Laboratoire des Sciences de l'Environnement Marin LEMAR, Institut Universitaire Européen de la Mer IUEM, Plouzané, France; Ifremer, Laboratoire Adaptation, Reproduction et Nutrition des Poissons ARN, Unité de Physiologie Fonctionnelle des Organismes Marins PFOM, Plouzané, France
| | - Grégory Charrier
- Université de Bretagne Occidentale, UMR 6539 CNRS/UBO/IRD/Ifremer, Laboratoire des Sciences de l'Environnement Marin LEMAR, Institut Universitaire Européen de la Mer IUEM, Plouzané, France
| | - Vianney Pichereau
- Université de Bretagne Occidentale, UMR 6539 CNRS/UBO/IRD/Ifremer, Laboratoire des Sciences de l'Environnement Marin LEMAR, Institut Universitaire Européen de la Mer IUEM, Plouzané, France
| | - Jean Laroche
- Université de Bretagne Occidentale, UMR 6539 CNRS/UBO/IRD/Ifremer, Laboratoire des Sciences de l'Environnement Marin LEMAR, Institut Universitaire Européen de la Mer IUEM, Plouzané, France.
| |
Collapse
|
29
|
Mackenzie AE, Milligan G. The emerging pharmacology and function of GPR35 in the nervous system. Neuropharmacology 2017; 113:661-671. [DOI: 10.1016/j.neuropharm.2015.07.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/20/2015] [Accepted: 07/27/2015] [Indexed: 02/07/2023]
|
30
|
Park SJ, Im DS. Sphingosine 1-Phosphate Receptor Modulators and Drug Discovery. Biomol Ther (Seoul) 2017; 25:80-90. [PMID: 28035084 PMCID: PMC5207465 DOI: 10.4062/biomolther.2016.160] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/06/2016] [Accepted: 10/27/2016] [Indexed: 01/07/2023] Open
Abstract
Initial discovery on sphingosine 1-phosphate (S1P) as an intracellular second messenger was faced unexpectedly with roles of S1P as a first messenger, which subsequently resulted in cloning of its G protein-coupled receptors, S1P1–5. The molecular identification of S1P receptors opened up a new avenue for pathophysiological research on this lipid mediator. Cellular and molecular in vitro studies and in vivo studies on gene deficient mice have elucidated cellular signaling pathways and the pathophysiological meanings of S1P receptors. Another unexpected finding that fingolimod (FTY720) modulates S1P receptors accelerated drug discovery in this field. Fingolimod was approved as a first-in-class, orally active drug for relapsing multiple sclerosis in 2010, and its applications in other disease conditions are currently under clinical trials. In addition, more selective S1P receptor modulators with better pharmacokinetic profiles and fewer side effects are under development. Some of them are being clinically tested in the contexts of multiple sclerosis and other autoimmune and inflammatory disorders, such as, psoriasis, Crohn’s disease, ulcerative colitis, polymyositis, dermatomyositis, liver failure, renal failure, acute stroke, and transplant rejection. In this review, the authors discuss the state of the art regarding the status of drug discovery efforts targeting S1P receptors and place emphasis on potential clinical applications.
Collapse
Affiliation(s)
- Soo-Jin Park
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Dong-Soon Im
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
31
|
During A, Penel G, Hardouin P. Understanding the local actions of lipids in bone physiology. Prog Lipid Res 2015; 59:126-46. [PMID: 26118851 DOI: 10.1016/j.plipres.2015.06.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 06/12/2015] [Accepted: 06/18/2015] [Indexed: 12/19/2022]
Abstract
The adult skeleton is a metabolically active organ system that undergoes continuous remodeling to remove old and/or stressed bone (resorption) and replace it with new bone (formation) in order to maintain a constant bone mass and preserve bone strength from micro-damage accumulation. In that remodeling process, cellular balances--adipocytogenesis/osteoblastogenesis and osteoblastogenesis/osteoclastogenesis--are critical and tightly controlled by many factors, including lipids as discussed in the present review. Interest in the bone lipid area has increased as a result of in vivo evidences indicating a reciprocal relationship between bone mass and marrow adiposity. Lipids in bones are usually assumed to be present only in the bone marrow. However, the mineralized bone tissue itself also contains small amounts of lipids which might play an important role in bone physiology. Fatty acids, cholesterol, phospholipids and several endogenous metabolites (i.e., prostaglandins, oxysterols) have been purported to act on bone cell survival and functions, the bone mineralization process, and critical signaling pathways. Thus, they can be regarded as regulatory molecules important in bone health. Recently, several specific lipids derived from membrane phospholipids (i.e., sphingosine-1-phosphate, lysophosphatidic acid and different fatty acid amides) have emerged as important mediators in bone physiology and the number of such molecules will probably increase in the near future. The present paper reviews the current knowledge about: (1°) bone lipid composition in both bone marrow and mineralized tissue compartments, and (2°) local actions of lipids on bone physiology in relation to their metabolism. Understanding the roles of lipids in bone is essential to knowing how an imbalance in their signaling pathways might contribute to bone pathologies, such as osteoporosis.
Collapse
Affiliation(s)
- Alexandrine During
- Université Lille 2, Laboratoire de Physiopathologie des maladies osseuses inflammatoires (PMOI), EA4490, Faculté de Chirurgie dentaire, Lille, France.
| | - Guillaume Penel
- Université Lille 2, Laboratoire de Physiopathologie des maladies osseuses inflammatoires (PMOI), EA4490, Faculté de Chirurgie dentaire, Lille, France
| | - Pierre Hardouin
- Université Lille 2, Laboratoire de Physiopathologie des maladies osseuses inflammatoires (PMOI), EA4490, Faculté de Chirurgie dentaire, Lille, France; Université ULCO, Laboratoire de Physiopathologie des maladies osseuses inflammatoires (PMOI), EA4490, Boulogne-sur-Mer, France
| |
Collapse
|
32
|
Im DS. Functions of omega-3 fatty acids and FFA4 (GPR120) in macrophages. Eur J Pharmacol 2015; 785:36-43. [PMID: 25987421 DOI: 10.1016/j.ejphar.2015.03.094] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 02/15/2015] [Accepted: 03/16/2015] [Indexed: 12/21/2022]
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs), which are plentiful in fish oil, have been known for decades to be beneficial functional nutrients in different disease states. GPR120 is a G protein-coupled receptor for long-chain unsaturated fatty acids, including n-3 PUFAs, and was recently renamed free fatty acid receptor 4 (FFA4). Studies on FFA4-deficient mice and the development of specific pharmacological tools have started to unravel the functions of FFA4 associated with the actions of n-3 PUFAs in obesity, type 2 diabetes, and inflammation-related diseases. Here, the state of the art regarding the roles and functions of FFA4 and n-3 PUFA in macrophages are reviewed from the pharmacological perspective. In particular, the functions of n-3 PUFA on the anti-inflammatory M2 phenotypes of macrophages in different organs, such as, adipose tissues and liver, are discussed along with future research directions.
Collapse
Affiliation(s)
- Dong-Soon Im
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea.
| |
Collapse
|
33
|
Ikubo M, Inoue A, Nakamura S, Jung S, Sayama M, Otani Y, Uwamizu A, Suzuki K, Kishi T, Shuto A, Ishiguro J, Okudaira M, Kano K, Makide K, Aoki J, Ohwada T. Structure-activity relationships of lysophosphatidylserine analogs as agonists of G-protein-coupled receptors GPR34, P2Y10, and GPR174. J Med Chem 2015; 58:4204-19. [PMID: 25970039 DOI: 10.1021/jm5020082] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Lysophosphatidylserine (LysoPS) is an endogenous lipid mediator generated by hydrolysis of membrane phospholipid phosphatidylserine. Recent ligand screening of orphan G-protein-coupled receptors (GPCRs) identified two LysoPS-specific human GPCRs, namely, P2Y10 (LPS2) and GPR174 (LPS3), which, together with previously reported GPR34 (LPS1), comprise a LysoPS receptor family. Herein, we examined the structure-activity relationships of a series of synthetic LysoPS analogues toward these recently deorphanized LysoPS receptors, based on the idea that LysoPS can be regarded as consisting of distinct modules (fatty acid, glycerol, and l-serine) connected by phosphodiester and ester linkages. Starting from the endogenous ligand (1-oleoyl-LysoPS, 1), we optimized the structure of each module and the ester linkage. Accordingly, we identified some structural requirements of each module for potency and for receptor subtype selectivity. Further assembly of individually structure-optimized modules yielded a series of potent and LysoPS receptor subtype-selective agonists, particularly for P2Y10 and GPR174.
Collapse
Affiliation(s)
- Masaya Ikubo
- †Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Asuka Inoue
- ‡Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Sho Nakamura
- †Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Sejin Jung
- †Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Misa Sayama
- †Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuko Otani
- †Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Akiharu Uwamizu
- ‡Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Keisuke Suzuki
- ‡Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Takayuki Kishi
- ‡Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Akira Shuto
- ‡Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Jun Ishiguro
- ‡Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Michiyo Okudaira
- ‡Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Kuniyuki Kano
- ‡Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Kumiko Makide
- ‡Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Junken Aoki
- ‡Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Tomohiko Ohwada
- †Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
34
|
The regulatory role of activating transcription factor 2 in inflammation. Mediators Inflamm 2014; 2014:950472. [PMID: 25049453 PMCID: PMC4090481 DOI: 10.1155/2014/950472] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 05/30/2014] [Indexed: 01/06/2023] Open
Abstract
Activating transcription factor 2 (ATF2) is a member of the leucine zipper family of DNA-binding proteins and is widely distributed in tissues including the liver, lung, spleen, and kidney. Like c-Jun and c-Fos, ATF2 responds to stress-related stimuli and may thereby influence cell proliferation, inflammation, apoptosis, oncogenesis, neurological development and function, and skeletal remodeling. Recent studies clarify the regulatory role of ATF2 in inflammation and describe potential inhibitors of this protein. In this paper, we summarize the properties and functions of ATF2 and explore potential applications of ATF2 inhibitors as tools for research and for the development of immunosuppressive and anti-inflammatory drugs.
Collapse
|