1
|
Huang Q, Wang J. CBP-mediated FOXO4 acetylation facilitates postmenopausal osteoporosis (PMO) progression through the inhibition of the Wnt/β-catenin signaling pathway. Histol Histopathol 2024; 39:1017-1024. [PMID: 38037460 DOI: 10.14670/hh-18-680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
FOXO4 was previously identified as a potential biomarker and therapeutic target for postmenopausal osteoporosis (PMO) using bioinformatic analysis, but its specific function and molecular mechanism in the progression of osteoporosis was not reported. The current study was designed to investigate the biological function and underlying mechanism of FOXO4 in PMO. Our results showed that FOXO4 expression was significantly upregulated in the serum samples of PMO patients, which was also negatively correlated with the expression of osteogenesis genes (OCN and ALP). In addition, FOXO4 depletion alleviated osteoporosis by facilitating osteogenic differentiation and inhibiting adipogenic differentiation in human bone marrow mesenchymal stem cells (hBMSCs). Overexpression of FOXO4 exerted the opposite effects on the osteogenic/adipogenic differentiation in hBMSCs. Moreover, FOXO4 knockdown activated the Wnt/β-catenin signaling whereas the inhibition of Wnt/β-catenin signaling overturned the effects of FOXO4 deficiency on osteoporosis. Furthermore, FOXO4 upregulation in PMO was caused by CBP-induced acetylation. In summary, our data demonstrated that FOXO4 was a potent biomarker for PMO and mediated the balance between osteogenesis and adipogenesis in hBMSCs by regulating Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Qiubo Huang
- Department of Orthopedics, Liyang People's Hospital, Liyang City, Jiangsu Province, PR China
| | - Jiang Wang
- Department of Orthopedics, Liyang People's Hospital, Liyang City, Jiangsu Province, PR China.
| |
Collapse
|
2
|
Wang Y, Liu H, Zhang Z. Recent Advance in Regulatory Effect of GRP120 on Bone Metabolism. Aging Dis 2023; 14:1714-1727. [PMID: 37196107 PMCID: PMC10529742 DOI: 10.14336/ad.2023.0216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/16/2023] [Indexed: 05/19/2023] Open
Abstract
The link between fatty acids and bone metabolism is complex and can be direct and indirect. This link has been reported in different types of bone cells and various stages of bone metabolism. G-protein coupled receptor 120 (GPR120), also called free fatty acid receptor 4 (FFAR4), is a member of the recently discovered G protein-coupled receptor family that can interact with both long-chain saturated fatty acids (C14-C18) and long-chain unsaturated fatty acids (C16-C22). Research shows that GPR120 regulates processes in different types of bone cells, directly or indirectly affecting bone metabolism. Our research reviewed the literature on the effects of GPR120 on bone marrow mesenchymal stem cells (BMMSCs), osteoblasts, osteoclasts, and chondrocytes, focusing on the research findings regarding the mechanism by which GPR120 alters specific bone metabolic diseases-osteoporosis and osteoarthritis. The data reviewed here provide a basis for clinical and basic research into the role of GPR120 on bone metabolic diseases.
Collapse
Affiliation(s)
| | - Haixia Liu
- Institute of Basic Theory for Chinese Medicine, Chinese Academy of Chinese Medical Sciences, Beijing, China.
| | - Zhiguo Zhang
- Institute of Basic Theory for Chinese Medicine, Chinese Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
3
|
Huang F, Liu X, Fu X, Chen Y, Jiang D, Wang T, Hu R, Zou X, Hu H, Liu C. 3D-Printed Bioactive Scaffold Loaded with GW9508 Promotes Critical-Size Bone Defect Repair by Regulating Intracellular Metabolism. Bioengineering (Basel) 2023; 10:bioengineering10050535. [PMID: 37237605 DOI: 10.3390/bioengineering10050535] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/14/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
The process of bone regeneration is complicated, and it is still a major clinical challenge to regenerate critical-size bone defects caused by severe trauma, infection, and tumor resection. Intracellular metabolism has been found to play an important role in the cell fate decision of skeletal progenitor cells. GW9508, a potent agonist of the free fatty acid receptors GPR40 and GPR120, appears to have a dual effect of inhibiting osteoclastogenesis and promoting osteogenesis by regulating intracellular metabolism. Hence, in this study, GW9508 was loaded on a scaffold based on biomimetic construction principles to facilitate the bone regeneration process. Through 3D printing and ion crosslinking, hybrid inorganic-organic implantation scaffolds were obtained after integrating 3D-printed β-TCP/CaSiO3 scaffolds with a Col/Alg/HA hydrogel. The 3D-printed β-TCP/CaSiO3 scaffolds had an interconnected porous structure that simulated the porous structure and mineral microenvironment of bone, and the hydrogel network shared similar physicochemical properties with the extracellular matrix. The final osteogenic complex was obtained after GW9508 was loaded into the hybrid inorganic-organic scaffold. To investigate the biological effects of the obtained osteogenic complex, in vitro studies and a rat cranial critical-size bone defect model were utilized. Metabolomics analysis was conducted to explore the preliminary mechanism. The results showed that 50 μM GW9508 facilitated osteogenic differentiation by upregulating osteogenic genes, including Alp, Runx2, Osterix, and Spp1 in vitro. The GW9508-loaded osteogenic complex enhanced osteogenic protein secretion and facilitated new bone formation in vivo. Finally, the results from metabolomics analysis suggested that GW9508 promoted stem cell differentiation and bone formation through multiple intracellular metabolism pathways, including purine and pyrimidine metabolism, amino acid metabolism, glutathione metabolism, and taurine and hypotaurine metabolism. This study provides a new approach to address the challenge of critical-size bone defects.
Collapse
Affiliation(s)
- Fangli Huang
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao Liu
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xihong Fu
- Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yan Chen
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Dong Jiang
- Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Tingxuan Wang
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Rongcheng Hu
- Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Hao Hu
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Chun Liu
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
- Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
4
|
Immune regulation of poly unsaturated fatty acids and free fatty acid receptor 4. J Nutr Biochem 2023; 112:109222. [PMID: 36402250 DOI: 10.1016/j.jnutbio.2022.109222] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 09/24/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022]
Abstract
Fatty acid metabolism contributes to energy supply and plays an important role in regulating immunity. Free fatty acids (FFAs) bind to free fatty acid receptors (FFARs) on the cell surface and mediate effects through the intra-cellular FFAR signaling pathways. FFAR4, also known as G-protein coupled receptor 120 (GPR120), has been identified as the primary receptor of omega-3 polyunsaturated fatty acids (ω-3 PUFAs). FFAR4 is a promising target for treating metabolic and inflammatory disorders due to its immune regulatory functions and the discovery of highly selective and efficient agonists. This review summarizes the reported immune regulatory functions of ω-3 PUFAs and FFAR4 in immune cells and immune-related diseases. We also speculate possible involvements of ω-3 PUFAs and FFAR4 in other types of inflammatory disorders.
Collapse
|
5
|
How Arrestins and GRKs Regulate the Function of Long Chain Fatty Acid Receptors. Int J Mol Sci 2022; 23:ijms232012237. [PMID: 36293091 PMCID: PMC9602559 DOI: 10.3390/ijms232012237] [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: 09/08/2022] [Revised: 10/03/2022] [Accepted: 10/08/2022] [Indexed: 11/16/2022] Open
Abstract
FFA1 and FFA4, two G protein-coupled receptors that are activated by long chain fatty acids, play crucial roles in mediating many biological functions in the body. As a result, these fatty acid receptors have gained considerable attention due to their potential to be targeted for the treatment of type-2 diabetes. However, the relative contribution of canonical G protein-mediated signalling versus the effects of agonist-induced phosphorylation and interactions with β-arrestins have yet to be fully defined. Recently, several reports have highlighted the ability of β-arrestins and GRKs to interact with and modulate different functions of both FFA1 and FFA4, suggesting that it is indeed important to consider these interactions when studying the roles of FFA1 and FFA4 in both normal physiology and in different disease settings. Here, we discuss what is currently known and show the importance of understanding fully how β-arrestins and GRKs regulate the function of long chain fatty acid receptors.
Collapse
|
6
|
Molecular Targets of Pinocembrin Underlying Its Regenerative Activities in Human Keratinocytes. Pharmaceuticals (Basel) 2022; 15:ph15080954. [PMID: 36015102 PMCID: PMC9415973 DOI: 10.3390/ph15080954] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/24/2022] [Accepted: 07/27/2022] [Indexed: 02/05/2023] Open
Abstract
Pinocembrin is one of the well-known compounds in the group of flavonoids. The pharmacological activities of pinocembrin in association with wound-healing activities have been reported. However, its effects on the aspect of cellular interaction underlying growth and survival are still unidentified in human keratinocytes. Our previous study reported that Boesenbergia rotunda potently stimulated survival and proliferation of a human keratinocyte cell line (HaCaT). On the basis that pinocembrin is revealed to be one of the major constituents of this plant, we aimed to define the survival- and proliferation-enhancing effects of this compound at the cellular level. Results from the current study confirmed that pinocembrin induced an increase in HaCaT cell number. At the signaling perspective, we identified that pinocembrin significantly triggered ERK1/2 and Akt activation. The stimulating effects of pinocembrin were clearly inhibited by MEK and PI3K inhibitors authenticating that proliferation- and survival-promoting activities of pinocembrin were mainly acted on these two signaling cascades. Altogether, we successfully identified that pinocembrin functions to induce keratinocyte proliferation and survival, at least by provoking MAPK and PI3K pathways. Our study encourages the fact that pinocembrin is one of the interesting natural flavonoid compounds to be developed as a wound closure-promoting agent.
Collapse
|
7
|
Zhu M, Fan Z. The role of the Wnt signalling pathway in the energy metabolism of bone remodelling. Cell Prolif 2022; 55:e13309. [PMID: 35811348 DOI: 10.1111/cpr.13309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/07/2022] [Accepted: 06/24/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVES Bone remodelling is necessary to repair old and impaired bone caused by aging and its effects. Injury in the process of bone remodelling generally leads to the development of various bone diseases. Energy metabolism plays crucial roles in bone cell formation and function, the disorder of which will disrupt the balance between bone formation and bone resorption. MATERIALS AND METHODS Here, we review the intrinsic interactions between bone remodelling and energy metabolism and the role of the Wnt signalling pathway. RESULTS We found a close interplay between metabolic pathways and bone homeostasis, demonstrating that bone plays an important role in the regulation of energy balance. We also discovered that Wnt signalling is associated with multiple biological processes regulating energy metabolism in bone cells. CONCLUSIONS Thus, targeted regulation of Wnt signalling and the recovery of the energy metabolism function of bone cells are key means for the treatment of metabolic bone diseases.
Collapse
Affiliation(s)
- Mengyuan Zhu
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Ning K, Liu S, Yang B, Wang R, Man G, Wang DE, Xu H. Update on the Effects of Energy Metabolism in Bone Marrow Mesenchymal Stem Cells Differentiation. Mol Metab 2022; 58:101450. [PMID: 35121170 PMCID: PMC8888956 DOI: 10.1016/j.molmet.2022.101450] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/16/2022] [Accepted: 01/27/2022] [Indexed: 11/29/2022] Open
Abstract
Background As common progenitor cells of osteoblasts and adipocytes, bone marrow mesenchymal (stromal) stem cells (BMSCs) play key roles in bone homeostasis, tissue regeneration, and global energy homeostasis; however, the intrinsic mechanism of BMSC differentiation is not well understood. Plasticity in energy metabolism allows BMSCs to match the divergent demands of osteo-adipogenic differentiation. Targeting BMSC metabolic pathways may provide a novel therapeutic perspective for BMSC differentiation unbalance related diseases. Scope of review This review covers the recent studies of glucose, fatty acids, and amino acids metabolism fuel the BMSC differentiation. We also discuss recent findings about energy metabolism in BMSC differentiation. Major conclusions Glucose, fatty acids, and amino acids metabolism provide energy to fuel BMSC differentiation. Moreover, some well-known regulators including environmental stress, hormone drugs, and biological and pathological factors may also influence BMSC differentiation by altering metabolism. This offers insight to the significance of metabolism on BMSC fate determination and provides the possibility of treating diseases related to BMSC differentiation, such as obesity and osteoporosis, from a metabolic perspective.
Collapse
|
9
|
Palmitic Acid Methyl Ester Enhances Adipogenic Differentiation in Rat Adipose Tissue-Derived Mesenchymal Stem Cells through a G Protein-Coupled Receptor-Mediated Pathway. Stem Cells Int 2021; 2021:9938649. [PMID: 34650609 PMCID: PMC8510814 DOI: 10.1155/2021/9938649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/29/2021] [Accepted: 09/11/2021] [Indexed: 11/29/2022] Open
Abstract
Adipogenic differentiation from stem cells has become a research target due to the increasing interest in obesity. It has been indicated that adipocytes can secrete palmitic acid methyl ester (PAME), which is able to regulate stem cell proliferation. However, the effects of PAME on adipogenic differentiation in stem cell remain unclear. Here, we present that the adipogenic differentiation medium supplemented with PAME induced the differentiation of rat adipose tissue-derived mesenchymal stem cells (rAD-MSCs) into adipocyte. rAD-MSCs were treated with PAME for 12 days and then subjected to various analyses. The results from the present study show that PAME significantly increased the levels of adipogenic differentiation markers, PPARγ and Gpd1, and enhanced adipogenic differentiation in rAD-MSCs. Furthermore, the level of GPR40/120 protein increased during induction of adipocyte differentiation in rAD-MSCs. Cotreatment with PAME and a GPR40/120 antagonist together inhibited the PAME-enhanced adipogenic differentiation. Moreover, PAME significantly increased phosphorylation of extracellular signal-regulated kinases (ERK), but not AKT and mTOR. Cotreatment with PAME and a GPR40/120 antagonist together inhibited the PAME-enhanced ERK phosphorylation and adipogenic differentiation. PAME also increased the intracellular Ca2+ levels. Cotreatment with PAME and a Ca2+ chelator or a phospholipase C (PLC) inhibitor prevented the PAME-enhanced ERK phosphorylation and adipogenic differentiation. Our data suggest that PAME activated the GPR40/120/PLC-mediated pathway, which in turn increased the intracellular Ca2+ levels, thereby activating the ERK, and eventually enhanced adipogenic differentiation in rAD-MSCs. The findings from the present study might help get insight into the physiological roles and molecular mechanism of PAME in regulating stem cell differentiation.
Collapse
|
10
|
Sithole C, Pieterse C, Howard K, Kasonga A. GPR120 Inhibits RANKL-Induced Osteoclast Formation and Resorption by Attenuating Reactive Oxygen Species Production in RAW264.7 Murine Macrophages. Int J Mol Sci 2021; 22:ijms221910544. [PMID: 34638884 PMCID: PMC8508775 DOI: 10.3390/ijms221910544] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 02/06/2023] Open
Abstract
Osteoclasts are large, multinucleated cells that are responsible for the resorption of bone. Bone degenerative diseases, such as osteoporosis, are characterized by overactive osteoclasts. Receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL) binding to its receptor on osteoclast precursors will trigger osteoclast formation and resorption. The production of reactive oxygen species (ROS) is known to play a crucial role in RANKL-induced osteoclast formation and resorption. G-protein coupled receptor 120 (GPR120) signalling has been shown to affect osteoclast formation, but the exact mechanisms of action require further investigation. RAW264.7 murine macrophages were seeded into culture plates and exposed to the GPR120 agonist, TUG-891, at varying concentrations (20–100 µM) and RANKL to induce osteoclast formation. TUG-891 was shown to inhibit osteoclast formation and resorption without affecting cell viability in RAW264.7 macrophages. TUG-891 further decreased ROS production when compared to RANKL only cells. Antioxidant proteins, Nrf2, HO-1 and NQO1 were shown to be upregulated while the ROS inducing protein, Nox1, was downregulated by TUG-891. Gene silencing revealed that TUG-891 exerted its effects specifically through GPR120. This study reveals that GPR120 signalling may inhibit osteoclast formation and resorption through inhibition on ROS production.
Collapse
Affiliation(s)
- Cynthia Sithole
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (C.S.); (C.P.)
| | - Carla Pieterse
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (C.S.); (C.P.)
| | - Kayla Howard
- Division of Clinical Pharmacology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 8000, South Africa;
| | - Abe Kasonga
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (C.S.); (C.P.)
- Correspondence: ; Tel.: +27-12-319-2433
| |
Collapse
|
11
|
Guo D, Zhao M, Xu W, He H, Li B, Hou T. Dietary interventions for better management of osteoporosis: An overview. Crit Rev Food Sci Nutr 2021; 63:125-144. [PMID: 34251926 DOI: 10.1080/10408398.2021.1944975] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Osteoporosis is a public health concern and a cause of bone loss, increased risk of skeletal fracture, and a heavy economic burden. It is common in postmenopausal women and the elderly and is impacted by dietary factors, lifestyle and some secondary factors. Although many drugs are available for the treatment of osteoporosis, these therapies are accompanied by subsequent side effects. Hence, dietary interventions are highly important to prevent osteoporosis. This review was aimed to provide a comprehensive understanding of the roles of dietary nutrients derived from natural foods and of common dietary patterns in the regulation of osteoporosis. Nutrients from daily diets, such as unsaturated fatty acids, proteins, minerals, peptides, phytoestrogens, and prebiotics, can regulate bone metabolism and reverse bone loss. Meanwhile, these nutrients generally existed in food groups and certain dietary patterns also play critical roles in skeletal health. Appropriate dietary interventions (nutrients and dietary patterns) could be primary and effective strategies to prevent and treat osteoporosis across the lifespan for the consumers and food enterprises.
Collapse
Affiliation(s)
- Danjun Guo
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China.,College of Food Science & Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Mengge Zhao
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wei Xu
- College of Food Science & Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Hui He
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Tao Hou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
12
|
Cugno C, Kizhakayil D, Calzone R, Rahman SM, Halade GV, Rahman MM. Omega-3 fatty acid-rich fish oil supplementation prevents rosiglitazone-induced osteopenia in aging C57BL/6 mice and in vitro studies. Sci Rep 2021; 11:10364. [PMID: 33990655 PMCID: PMC8121944 DOI: 10.1038/s41598-021-89827-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Rosiglitazone is an effective insulin-sensitizer, however associated with bone loss mainly due to increased bone resorption and bone marrow adiposity. We investigated the effect of the co-administration of fish oil rich in omega-3 fatty acids (FAs) on rosiglitazone-induced bone loss in C57BL/6 mice and the mechanisms underlying potential preventive effect. Mice fed the iso-caloric diet supplemented with fish oil exhibited significantly higher levels of bone density in different regions compared to the other groups. In the same cohort of mice, reduced activity of COX-2, enhanced activity of alkaline phosphatase, lower levels of cathepsin k, PPAR-γ, and pro-inflammatory cytokines, and a higher level of anti-inflammatory cytokines were observed. Moreover, fish oil restored rosiglitazone-induced down-regulation of osteoblast differentiation and up-regulation of adipocyte differentiation in C3H10T1/2 cells and inhibited the up-regulation of osteoclast differentiation of RANKL-treated RAW264.7 cells. We finally tested our hypothesis on human Mesenchymal Stromal Cells differentiated to osteocytes and adipocytes confirming the beneficial effect of docosahexaenoic acid (DHA) omega-3 FA during treatment with rosiglitazone, through the down-regulation of adipogenic genes, such as adipsin and FABP4 along the PPARγ/FABP4 axis, and reducing the capability of osteocytes to switch toward adipogenesis. Fish oil may prevent rosiglitazone-induced bone loss by inhibiting inflammation, osteoclastogenesis, and adipogenesis and by enhancing osteogenesis in the bone microenvironment.
Collapse
Affiliation(s)
- Chiara Cugno
- Advanced Cell Therapy Core, Sidra Medicine, Doha, Qatar
| | | | - Rita Calzone
- Advanced Cell Therapy Core, Sidra Medicine, Doha, Qatar
| | - Shaikh Mizanoor Rahman
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, The University of South Florida Health, Tampa, FL, USA
| | - Md M Rahman
- Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar.
| |
Collapse
|
13
|
Grundmann M, Bender E, Schamberger J, Eitner F. Pharmacology of Free Fatty Acid Receptors and Their Allosteric Modulators. Int J Mol Sci 2021; 22:ijms22041763. [PMID: 33578942 PMCID: PMC7916689 DOI: 10.3390/ijms22041763] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 12/19/2022] Open
Abstract
The physiological function of free fatty acids (FFAs) has long been regarded as indirect in terms of their activities as educts and products in metabolic pathways. The observation that FFAs can also act as signaling molecules at FFA receptors (FFARs), a family of G protein-coupled receptors (GPCRs), has changed the understanding of the interplay of metabolites and host responses. Free fatty acids of different chain lengths and saturation statuses activate FFARs as endogenous agonists via binding at the orthosteric receptor site. After FFAR deorphanization, researchers from the pharmaceutical industry as well as academia have identified several ligands targeting allosteric sites of FFARs with the aim of developing drugs to treat various diseases such as metabolic, (auto)inflammatory, infectious, endocrinological, cardiovascular, and renal disorders. GPCRs are the largest group of transmembrane proteins and constitute the most successful drug targets in medical history. To leverage the rich biology of this target class, the drug industry seeks alternative approaches to address GPCR signaling. Allosteric GPCR ligands are recognized as attractive modalities because of their auspicious pharmacological profiles compared to orthosteric ligands. While the majority of marketed GPCR drugs interact exclusively with the orthosteric binding site, allosteric mechanisms in GPCR biology stay medically underexploited, with only several allosteric ligands currently approved. This review summarizes the current knowledge on the biology of FFAR1 (GPR40), FFAR2 (GPR43), FFAR3 (GPR41), FFAR4 (GPR120), and GPR84, including structural aspects of FFAR1, and discusses the molecular pharmacology of FFAR allosteric ligands as well as the opportunities and challenges in research from the perspective of drug discovery.
Collapse
Affiliation(s)
- Manuel Grundmann
- Research and Early Development, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany;
- Correspondence:
| | - Eckhard Bender
- Drug Discovery Sciences, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany; (E.B.); (J.S.)
| | - Jens Schamberger
- Drug Discovery Sciences, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany; (E.B.); (J.S.)
| | - Frank Eitner
- Research and Early Development, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany;
| |
Collapse
|
14
|
Merlotti D, Cosso R, Eller-Vainicher C, Vescini F, Chiodini I, Gennari L, Falchetti A. Energy Metabolism and Ketogenic Diets: What about the Skeletal Health? A Narrative Review and a Prospective Vision for Planning Clinical Trials on this Issue. Int J Mol Sci 2021; 22:ijms22010435. [PMID: 33406758 PMCID: PMC7796307 DOI: 10.3390/ijms22010435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/27/2020] [Accepted: 12/30/2020] [Indexed: 12/15/2022] Open
Abstract
The existence of a common mesenchymal cell progenitor shared by bone, skeletal muscle, and adipocytes cell progenitors, makes the role of the skeleton in energy metabolism no longer surprising. Thus, bone fragility could also be seen as a consequence of a “poor” quality in nutrition. Ketogenic diet was originally proven to be effective in epilepsy, and long-term follow-up studies on epileptic children undergoing a ketogenic diet reported an increased incidence of bone fractures and decreased bone mineral density. However, the causes of such negative impacts on bone health have to be better defined. In these subjects, the concomitant use of antiepileptic drugs and the reduced mobilization may partly explain the negative effects on bone health, but little is known about the effects of diet itself, and/or generic alterations in vitamin D and/or impaired growth factor production. Despite these remarks, clinical studies were adequately designed to investigate bone health are scarce and bone health related aspects are not included among the various metabolic pathologies positively influenced by ketogenic diets. Here, we provide not only a narrative review on this issue, but also practical advice to design and implement clinical studies on ketogenic nutritional regimens and bone health outcomes. Perspectives on ketogenic regimens, microbiota, microRNAs, and bone health are also included.
Collapse
Affiliation(s)
- Daniela Merlotti
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (D.M.); (L.G.)
| | - Roberta Cosso
- Istituto Auxologico Italiano “Scientific Institute for Hospitalisation and Care”, 20100 Milano, Italy; (R.C.); (I.C.)
| | - Cristina Eller-Vainicher
- Unit of Endocrinology, Fondazione IRCCS Cà Granda-Ospedale Maggiore Policlinico Milano, 20122 Milano, Italy;
| | - Fabio Vescini
- Endocrinology and Metabolism Unit, University-Hospital S. Maria della Misericordia of Udine, 33100 Udine, Italy;
| | - Iacopo Chiodini
- Istituto Auxologico Italiano “Scientific Institute for Hospitalisation and Care”, 20100 Milano, Italy; (R.C.); (I.C.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milano, Italy
| | - Luigi Gennari
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (D.M.); (L.G.)
| | - Alberto Falchetti
- Istituto Auxologico Italiano “Scientific Institute for Hospitalisation and Care”, 20100 Milano, Italy; (R.C.); (I.C.)
- Correspondence:
| |
Collapse
|
15
|
Role of Metabolism in Bone Development and Homeostasis. Int J Mol Sci 2020; 21:ijms21238992. [PMID: 33256181 PMCID: PMC7729585 DOI: 10.3390/ijms21238992] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/22/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
Carbohydrates, fats, and proteins are the underlying energy sources for animals and are catabolized through specific biochemical cascades involving numerous enzymes. The catabolites and metabolites in these metabolic pathways are crucial for many cellular functions; therefore, an imbalance and/or dysregulation of these pathways causes cellular dysfunction, resulting in various metabolic diseases. Bone, a highly mineralized organ that serves as a skeleton of the body, undergoes continuous active turnover, which is required for the maintenance of healthy bony components through the deposition and resorption of bone matrix and minerals. This highly coordinated event is regulated throughout life by bone cells such as osteoblasts, osteoclasts, and osteocytes, and requires synchronized activities from different metabolic pathways. Here, we aim to provide a comprehensive review of the cellular metabolism involved in bone development and homeostasis, as revealed by mouse genetic studies.
Collapse
|
16
|
Yang J, Ueharu H, Mishina Y. Energy metabolism: A newly emerging target of BMP signaling in bone homeostasis. Bone 2020; 138:115467. [PMID: 32512164 PMCID: PMC7423769 DOI: 10.1016/j.bone.2020.115467] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022]
Abstract
Energy metabolism is the process of generating energy (i.e. ATP) from nutrients. This process is indispensable for cell homeostasis maintenance and responses to varying conditions. Cells require energy for growth and maintenance and have evolved to have multiple pathways to produce energy. Both genetic and functional studies have demonstrated that energy metabolism, such as glucose, fatty acid, and amino acid metabolism, plays important roles in the formation and function of bone cells including osteoblasts, osteocytes, and osteoclasts. Dysregulation of energy metabolism in bone cells consequently disturbs the balance between bone formation and bone resorption. Metabolic diseases have also been reported to affect bone homeostasis. Bone morphogenic protein (BMP) signaling plays critical roles in regulating the formation and function of bone cells, thus affecting bone development and homeostasis. Mutations of BMP signaling-related genes in mice have been reported to show abnormalities in energy metabolism in many tissues, including bone. In addition, BMP signaling correlates with critical signaling pathways such as mTOR, HIF, Wnt, and self-degradative process autophagy to coordinate energy metabolism and bone homeostasis. These findings will provide a newly emerging target of BMP signaling and potential therapeutic strategies and the improved management of bone diseases. This review summarizes the recent advances in our understanding of (1) energy metabolism in regulating the formation and function of bone cells, (2) function of BMP signaling in whole body energy metabolism, and (3) mechanistic interaction of BMP signaling with other signaling pathways and biological processes critical for energy metabolism and bone homeostasis.
Collapse
Affiliation(s)
- Jingwen Yang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA; The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China.
| | - Hiroki Ueharu
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
17
|
Xu Z, Ke T, Zhang Y, Fu C, He W. Agonism of GPR120 prevented IL-1β-induced reduction of extracellular matrix through SOX-9. Aging (Albany NY) 2020; 12:12074-12085. [PMID: 32580167 PMCID: PMC7343462 DOI: 10.18632/aging.103375] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 04/07/2020] [Indexed: 12/20/2022]
Abstract
Osteoarthritis (OA) is a whole-joint disease with extremely high prevalence. In all treatment approaches of OA, blocking the degradation of the cartilage extracellular matrix is an important treatment. In OA, overexpression of derivative enzymes leads to excessive catabolism and reduced synthesis of cartilage including type II collagen and aggrecan, which results in irreversible destruction of the joint. SOX9 is a transcription factor that regulates the synthesis of type II collagen and aggrecan and is significantly downregulated in OA. GPR120 has been reported to affect the pathophysiology of OA. In this study, we used the GPR120 agonist GW9508 and TUG891 in ATDC5 chondrocytes exposed to interleukin (IL)-1β to investigate the involvement of GPR120 in SOX9-mediated expression of type II collagen and aggrecan. Our findings show that agonism of GPR120 can reduce inflammation by inhibiting the expression of IL-6 and IL-8 induced by IL-1β. We also show that GW9508 and TUG891 rescue the expression of type II collagen and aggrecan by preventing the reduction of SOX9 expression. Additionally, we demonstrate that the effects of GW9508 on SOX9 expression are mediated through CREB and that GPR120 is indeed required for this effect. Thus, agonism of GPR120 by GW9508 might be a potential therapeutic strategy to halt or prevent cartilage degradation.
Collapse
Affiliation(s)
- Zhixian Xu
- Department of Emergency Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, Fujian, P.R. China
| | - Tie Ke
- Department of Emergency Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, Fujian, P.R. China
| | - Yongfa Zhang
- Department of Emergency Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, Fujian, P.R. China
| | - Chaofeng Fu
- Department of Emergency Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, Fujian, P.R. China
| | - Wubing He
- Department of Emergency Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, Fujian, P.R. China
| |
Collapse
|
18
|
Huang Z, Guo F, Xia Z, Liang Y, Lei S, Tan Z, Ma L, Fu P. Activation of GPR120 by TUG891 ameliorated cisplatin-induced acute kidney injury via repressing ER stress and apoptosis. Biomed Pharmacother 2020; 126:110056. [PMID: 32145589 DOI: 10.1016/j.biopha.2020.110056] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 02/08/2023] Open
Abstract
Activation of G protein-coupled receptor 120 (GPR120) could inhibit apoptosis and inflammation in cerebral ischemic injury and liver ischemia-reperfusion injury. However, whether GPR120 agonism exerted potential for cisplatin-induced acute kidney injury and the involved mechanisms remained unknown. In our study, pharmacological activation of GPR120 by TUG891 treatment remarkably reduced the elevated serum creatinine level and attenuated tubular injury. Cisplatin triggered ATF6, PERK and IRE1 pathways of unfolded protein response (UPR) of ER stress in the injured kidney tissue, as well as the downstream molecules eIF2α, ATF4 and XBP1. Protein of ER stress-mediated apoptosis, CHOP, was overexpressed in the cisplatin group. Oral application of TUG891 displayed effective inhibition of ER stress and apoptosis. TUG891 treatment significantly decreased the TUNEL positive cells and the flow cytometry of HK-2 cells delineated the similar results that the apoptosis rates were considerably reduced in the TUG891 group compared to cisplatin group. Collectively, activation of GPR120 by TUG891 exhibited renal protection against cisplatin-induced AKI via suppressing ER-associated apoptosis in tubular epithelial cells.
Collapse
Affiliation(s)
- Zhuo Huang
- Division of Nephrology and National Clinical Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China; Division of Pathology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Fan Guo
- Division of Nephrology and National Clinical Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Zijing Xia
- Division of Nephrology and National Clinical Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yan Liang
- Division of Pathology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Song Lei
- Division of Pathology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Zhouke Tan
- Division of Nephrology and National Clinical Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Liang Ma
- Division of Nephrology and National Clinical Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Ping Fu
- Division of Nephrology and National Clinical Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
19
|
The GPR120 Agonist TUG-891 Inhibits the Motility and Phagocytosis of Mouse Alveolar Macrophages. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1706168. [PMID: 32149083 PMCID: PMC7056993 DOI: 10.1155/2020/1706168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/30/2019] [Accepted: 02/05/2020] [Indexed: 01/08/2023]
Abstract
Movement and phagocytosis characterize the fundamental actions of macrophages. Although it is known that the free fatty acid receptor GPR120 is expressed in macrophages and regulates cytokine expression to exert anti-inflammatory activities, the effects of GPR120 activation on the motility and phagocytosis of macrophages are not clear. In this study, mouse alveolar macrophages (AM) were stimulated with the GPR120 agonist TUG-891, and the changes in cell motility, intracellular Ca2+ concentration ([Ca2+]i), and the ability of phagocytosis were measured. Mouse AM in controls exhibited active movement in vitro, and TUG-891 significantly restrained AM movement. Meanwhile, TUG-891 stimulated a quick increase in [Ca2+]i in AM, which was blocked separately by the Gq protein inhibitor YM-254890, the phospholipase C (PLC) inhibitor U73122, or depletion of endoplasmic reticulum (ER) Ca2+ store by thapsigargin. The inhibition of AM movement by TUG-891 was eliminated by YM-254890, U73122, thapsigargin, and chelation of cytosolic Ca2+ by BAPTA. Moreover, TUG-891 inhibited AM phagocytosis of fluorescent microspheres, which was also blocked by YM-254890, U73122, thapsigargin, and BAPTA. In conclusion, GPR120 activation in mouse AM increases [Ca2+]i but inhibits the motility and phagocytosis via Gq protein/PLC-mediated Ca2+ release from ER Ca2+ store.
Collapse
|
20
|
Bao M, Zhang K, Wei Y, Hua W, Gao Y, Li X, Ye L. Therapeutic potentials and modulatory mechanisms of fatty acids in bone. Cell Prolif 2020; 53:e12735. [PMID: 31797479 PMCID: PMC7046483 DOI: 10.1111/cpr.12735] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 02/05/2023] Open
Abstract
Bone metabolism is a lifelong process that includes bone formation and resorption. Osteoblasts and osteoclasts are the predominant cell types associated with bone metabolism, which is facilitated by other cells such as bone marrow mesenchymal stem cells (BMMSCs), osteocytes and chondrocytes. As an important component in our daily diet, fatty acids are mainly categorized as long-chain fatty acids including polyunsaturated fatty acids (LCPUFAs), monounsaturated fatty acids (LCMUFAs), saturated fatty acids (LCSFAs), medium-/short-chain fatty acids (MCFAs/SCFAs) as well as their metabolites. Fatty acids are closely associated with bone metabolism and associated bone disorders. In this review, we summarized the important roles and potential therapeutic implications of fatty acids in multiple bone disorders, reviewed the diverse range of critical effects displayed by fatty acids on bone metabolism, and elucidated their modulatory roles and mechanisms on specific bone cell types. The evidence supporting close implications of fatty acids in bone metabolism and disorders suggests fatty acids as potential therapeutic and nutritional agents for the treatment and prevention of metabolic bone diseases.
Collapse
Affiliation(s)
- Minyue Bao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Kaiwen Zhang
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yangyini Wei
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Weihan Hua
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yanzi Gao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xin Li
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ling Ye
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesDepartment of Cariology and EndodonticsWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
21
|
Wang C, Liu Y, Pan Y, Jin H. Effect of GSK-137647A, the first non-carboxylic FFA4 agonist, on the osteogenic and adipogenic differentiation of bone mesenchymal stem cells in db/db mice. ACTA ACUST UNITED AC 2019; 72:461-469. [PMID: 31858612 DOI: 10.1111/jphp.13217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 11/29/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To investigate the effect of GSK-137647A, the first non-carboxylic FFA4 agonist, on osteogenic and adipogenic differentiation of bone mesenchymal stem cells (BMSCs) of db/db mice. METHODS Bone mesenchymal stem cells were extracted from 8-week-old db/db mice. Cell Counting Kit-8 was used to evaluate the toxicity of GSK-137647A on BMSCs, and the optimal concentration of GSK-137647A was selected to investigate the osteogenic and adipogenic differentiation of BMSCs, and relevant indicators of osteoblasts and adipocytes were detected. KEY FINDINGS GSK-137647A had no significant toxicity on cell growth and proliferation. Moreover, GSK-137647A showed a significant increase in mineralization of BMSCs differentiated osteoblasts compared to the control group and elevated the alkaline phosphatase (ALP) activity in a time-dependent manner. Meanwhile, the treatment of GSK-137647A decreased the adipogenic differentiation of BMSCs. The expression levels of ALP, runt-related transcription factor 2, bone morphogenetic protein 4, osterix and β-catenin were significantly increased in GSK-137647A-treated group, while the gene and protein levels of peroxisome proliferator-activated receptor γ and CCAAT/enhancer binding protein α were significantly reduced. CONCLUSIONS All of these results demonstrated that GSK-137647A suppressed the adipogenic differentiation and promoted osteogenic differentiation of BMSCs, which is partly attributed to the increased expression of β-catenin in wingless/integrated signalling pathway.
Collapse
Affiliation(s)
- Chunlei Wang
- Department of Endocrinology, Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu, China
| | - Yanmei Liu
- Department of Endocrinology, Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu, China
| | - Yuan Pan
- Department of Science and Education, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Hui Jin
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, Medical School, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
22
|
Ren Z, Chen L, Wang Y, Wei X, Zeng S, Zheng Y, Gao C, Liu H. Activation of the Omega-3 Fatty Acid Receptor GPR120 Protects against Focal Cerebral Ischemic Injury by Preventing Inflammation and Apoptosis in Mice. THE JOURNAL OF IMMUNOLOGY 2018; 202:747-759. [PMID: 30598514 DOI: 10.4049/jimmunol.1800637] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 11/22/2018] [Indexed: 12/28/2022]
Abstract
G protein-coupled receptor 120 (GPR120) has been shown to negatively regulate inflammation and apoptosis, but its role in cerebral ischemic injury remains unclear. Using an in vivo model of middle cerebral artery occlusion (MCAO) and an in vitro model of oxygen-glucose deprivation (OGD), we investigated the potential role and molecular mechanisms of GPR120 in focal cerebral ischemic injury. Increased GPR120 expression was observed in microglia and neurons following MCAO-induced ischemia in wild type C57BL/6 mice. Treatment with docosahexaenoic acid (DHA) inhibited OGD-induced inflammatory response in primary microglia and murine microglial BV2 cells, whereas silencing of GPR120 strongly exacerbated the inflammation induced by OGD and abolished the anti-inflammatory effects of DHA. Mechanistically, DHA inhibited OGD-induced inflammation through GPR120 interacting with β-arrestin2. In addition to its anti-inflammatory function, GPR120 also played a role in apoptosis as its knockdown impaired the antiapoptotic effect of DHA in OGD-induced rat pheochromocytoma (PC12) cells. Finally, using MCAO mouse model, we demonstrated that GPR120 activation protected against focal cerebral ischemic injury by preventing inflammation and apoptosis. Our study indicated that pharmacological targeting of GPR120 may provide a novel approach for the treatment of patients with ischemic stroke.
Collapse
Affiliation(s)
- Zhiping Ren
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Lin Chen
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yimeng Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xinbing Wei
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Shenglan Zeng
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yi Zheng
- State Key Laboratory of Microbial Technology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; and.,Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Chengjiang Gao
- State Key Laboratory of Microbial Technology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; .,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; and.,Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Huiqing Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China;
| |
Collapse
|
23
|
Chen Y, Zhang D, Ho KW, Lin S, Suen WCW, Zhang H, Zha Z, Li G, Leung PS. GPR120 is an important inflammatory regulator in the development of osteoarthritis. Arthritis Res Ther 2018; 20:163. [PMID: 30075737 PMCID: PMC6091098 DOI: 10.1186/s13075-018-1660-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 07/04/2018] [Indexed: 12/28/2022] Open
Abstract
Background The aim of this study was to investigate the regulatory role of G-protein coupled receptor 120 (GPR120) in the development and progression of osteoarthritis (OA). Methods GPR120 knockout (KO) and wild-type (WT) mice were used to create an animal model of OA by means of anterior cruciate ligament transection (ACLT) surgery. The severity of OA was staged and evaluated by histological examination, microcomputed tomography scan and enzyme-linked immunosorbent assay (ELISA). The anti-inflammatory effects of the GPR120 agonist docosahexaenoic acid (DHA) on human chondrocytes were further evaluated by specific inflammatory markers. In addition, the healing progression of a skin defect model was determined with histological assays. Results The GPR120-KO mice displayed an accelerated development of OA after ACLT. The secondary inflammation, cartilage degeneration, and subchondral bone aberrant changes were significantly elevated in the early phase of OA in KO mice relative to those in WT mice. In addition, we found that GPR120 levels were downregulated in OA patients compared with control subjects, whereas GPR120 activation with DHA exhibited anti-inflammatory effects in primary human chondrocytes in vitro. Moreover, results from the skin defect model showed that GPR120 agonism with DHA enhanced wound repair in mice, as shown by the downregulation of the number of CD68+ cells. Conclusions Our study suggests that GPR120 is an important inflammatory mediator during the development of OA, and that it is a potential marker for the diagnosis of high-risk patients with OA. Electronic supplementary material The online version of this article (10.1186/s13075-018-1660-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuanfeng Chen
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, People's Republic of China.,Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China
| | - Dan Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, People's Republic of China
| | - Ki Wai Ho
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China
| | - Wade Chun-Wai Suen
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China.,Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, UK
| | - Huantian Zhang
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, People's Republic of China
| | - Zhengang Zha
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, People's Republic of China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China. .,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China.
| | - Po Sing Leung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, People's Republic of China.
| |
Collapse
|
24
|
Maillard V, Desmarchais A, Durcin M, Uzbekova S, Elis S. Docosahexaenoic acid (DHA) effects on proliferation and steroidogenesis of bovine granulosa cells. Reprod Biol Endocrinol 2018; 16:40. [PMID: 29699561 PMCID: PMC5918968 DOI: 10.1186/s12958-018-0357-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/18/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Docosahexaenoic acid (DHA) is a n-3 polyunsaturated fatty acid (PUFA) belonging to a family of biologically active fatty acids (FA), which are known to have numerous health benefits. N-3 PUFAs affect reproduction in cattle, and notably directly affect follicular cells. In terms of reproduction in cattle, n-3 PUFA-enriched diets lead to increased follicle size or numbers. METHODS The objective of the present study was to analyze the effects of DHA (1, 10, 20 and 50 μM) on proliferation and steroidogenesis (parametric and/or non parametric (permutational) ANOVA) of bovine granulosa cells in vitro and mechanisms of action through protein expression (Kruskal-Wallis) and signaling pathways (non parametric ANOVA) and to investigate whether DHA could exert part of its action through the free fatty acid receptor 4 (FFAR4). RESULTS DHA (10 and 50 μM) increased granulosa cell proliferation and DHA 10 μM led to a corresponding increase in proliferating cell nuclear antigen (PCNA) expression level. DHA also increased progesterone secretion at 1, 20 and 50 μM, and estradiol secretion at 1, 10 and 20 μM. Consistent increases in protein levels were also reported for the steroidogenic enzymes, cytochrome P450 family 11 subfamily A member 1 (CYP11A1) and hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 1 (HSD3B1), and of the cholesterol transporter steroidogenic acute regulatory protein (StAR), which are necessary for production of progesterone or androstenedione. FFAR4 was expressed in all cellular types of bovine ovarian follicles, and in granulosa cells it was localized close to the cellular membrane. TUG-891 treatment (1 and 50 μM), a FFAR4 agonist, increased granulosa cell proliferation and MAPK14 phosphorylation in a similar way to that observed with DHA treatment. However, TUG-891 treatment (1, 10 and 50 μM) showed no effect on progesterone or estradiol secretion. CONCLUSIONS These data show that DHA stimulated proliferation and steroidogenesis of bovine granulosa cells and led to MAPK14 phosphorylation. FFAR4 involvement in DHA effects requires further investigation, even if our data might suggest FFAR4 role in DHA effects on granulosa cell proliferation. Other mechanisms of DHA action should be investigated as the steroidogenic effects seemed to be independent of FFAR4 activation.
Collapse
Affiliation(s)
- Virginie Maillard
- 0000 0001 2182 6141grid.12366.30UMR PRC, CNRS, IFCE, INRA, Université de Tours, 37380 Nouzilly, France
- 0000 0004 0385 4036grid.464126.3INRA Centre Val de Loire, Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
| | - Alice Desmarchais
- 0000 0001 2182 6141grid.12366.30UMR PRC, CNRS, IFCE, INRA, Université de Tours, 37380 Nouzilly, France
| | - Maeva Durcin
- 0000 0001 2182 6141grid.12366.30UMR PRC, CNRS, IFCE, INRA, Université de Tours, 37380 Nouzilly, France
| | - Svetlana Uzbekova
- 0000 0001 2182 6141grid.12366.30UMR PRC, CNRS, IFCE, INRA, Université de Tours, 37380 Nouzilly, France
| | - Sebastien Elis
- 0000 0001 2182 6141grid.12366.30UMR PRC, CNRS, IFCE, INRA, Université de Tours, 37380 Nouzilly, France
| |
Collapse
|
25
|
Song T, Yang Y, Zhou Y, Wei H, Peng J. GPR120: a critical role in adipogenesis, inflammation, and energy metabolism in adipose tissue. Cell Mol Life Sci 2017; 74:2723-2733. [PMID: 28285320 PMCID: PMC11107682 DOI: 10.1007/s00018-017-2492-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 02/17/2017] [Accepted: 02/21/2017] [Indexed: 01/12/2023]
Abstract
It is well known that adipose tissue has a critical role in the development of obesity and metabolic diseases and that adipose tissue acts as an endocrine organ to regulate lipid and glucose metabolism. Accumulating in the adipose tissue, fatty acids serve as a primary source of essential nutrients and act on intracellular and cell surface receptors to regulate biological events. G protein-coupled receptor 120 (GPR120) represents a promising target for the treatment of obesity-related metabolic disorders for its involvement in the regulation of adipogenesis, inflammation, glucose uptake, and insulin resistance. In this review, we summarize recent studies and advances regarding the systemic role of GPR120 in adipose tissue, including both white and brown adipocytes. We offer a new perspective by comparing the different roles in a variety of homeostatic processes from adipogenic development to adipocyte metabolism, and we also discuss the effects of natural and synthetic agonists that may be potential agents for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Tongxing Song
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Yang Yang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Yuanfei Zhou
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| |
Collapse
|
26
|
Watterson KR, Hansen SVF, Hudson BD, Alvarez-Curto E, Raihan SZ, Azevedo CMG, Martin G, Dunlop J, Yarwood SJ, Ulven T, Milligan G. Probe-Dependent Negative Allosteric Modulators of the Long-Chain Free Fatty Acid Receptor FFA4. Mol Pharmacol 2017; 91:630-641. [PMID: 28385906 PMCID: PMC5438128 DOI: 10.1124/mol.116.107821] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/29/2017] [Indexed: 12/16/2022] Open
Abstract
High-affinity and selective antagonists that are able to block the actions of both endogenous and synthetic agonists of G protein–coupled receptors are integral to analysis of receptor function and to support suggestions of therapeutic potential. Although there is great interest in the potential of free fatty acid receptor 4 (FFA4) as a novel therapeutic target for the treatment of type II diabetes, the broad distribution pattern of this receptor suggests it may play a range of roles beyond glucose homeostasis in different cells and tissues. To date, a single molecule, 4-methyl-N-9H-xanthen-9-yl-benzenesulfonamide (AH-7614), has been described as an FFA4 antagonist; however, its mechanism of antagonism remains unknown. We synthesized AH-7614 and a chemical derivative and demonstrated these to be negative allosteric modulators (NAMs) of FFA4. Although these NAMs did inhibit FFA4 signaling induced by a range of endogenous and synthetic agonists, clear agonist probe dependence in the nature of allosteric modulation was apparent. Although AH-7614 did not antagonize the second long-chain free fatty acid receptor, free fatty acid receptor 1, the simple chemical structure of AH-7614 containing features found in many anticancer drugs suggests that a novel close chemical analog of AH-7614 devoid of FFA4 activity, 4-methyl-N-(9H-xanthen-9-yl)benzamide (TUG-1387), will also provide a useful control compound for future studies assessing FFA4 function. Using TUG-1387 alongside AH-7614, we show that endogenous activation of FFA4 expressed by murine C3H10T1/2 mesenchymal stem cells is required for induced differentiation of these cells toward a more mature, adipocyte-like phenotype.
Collapse
Affiliation(s)
- Kenneth R Watterson
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (K.R.W., B.D.H., E.A.-C., S.Z.R., J.D., S.J.Y., G.M.); Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark (S.V.F.H., C.M.G.A., G.M., T.U.); and Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom (S.J.Y.)
| | - Steffen V F Hansen
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (K.R.W., B.D.H., E.A.-C., S.Z.R., J.D., S.J.Y., G.M.); Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark (S.V.F.H., C.M.G.A., G.M., T.U.); and Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom (S.J.Y.)
| | - Brian D Hudson
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (K.R.W., B.D.H., E.A.-C., S.Z.R., J.D., S.J.Y., G.M.); Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark (S.V.F.H., C.M.G.A., G.M., T.U.); and Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom (S.J.Y.)
| | - Elisa Alvarez-Curto
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (K.R.W., B.D.H., E.A.-C., S.Z.R., J.D., S.J.Y., G.M.); Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark (S.V.F.H., C.M.G.A., G.M., T.U.); and Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom (S.J.Y.)
| | - Sheikh Zahir Raihan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (K.R.W., B.D.H., E.A.-C., S.Z.R., J.D., S.J.Y., G.M.); Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark (S.V.F.H., C.M.G.A., G.M., T.U.); and Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom (S.J.Y.)
| | - Carlos M G Azevedo
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (K.R.W., B.D.H., E.A.-C., S.Z.R., J.D., S.J.Y., G.M.); Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark (S.V.F.H., C.M.G.A., G.M., T.U.); and Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom (S.J.Y.)
| | - Gabriel Martin
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (K.R.W., B.D.H., E.A.-C., S.Z.R., J.D., S.J.Y., G.M.); Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark (S.V.F.H., C.M.G.A., G.M., T.U.); and Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom (S.J.Y.)
| | - Julia Dunlop
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (K.R.W., B.D.H., E.A.-C., S.Z.R., J.D., S.J.Y., G.M.); Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark (S.V.F.H., C.M.G.A., G.M., T.U.); and Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom (S.J.Y.)
| | - Stephen J Yarwood
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (K.R.W., B.D.H., E.A.-C., S.Z.R., J.D., S.J.Y., G.M.); Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark (S.V.F.H., C.M.G.A., G.M., T.U.); and Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom (S.J.Y.)
| | - Trond Ulven
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (K.R.W., B.D.H., E.A.-C., S.Z.R., J.D., S.J.Y., G.M.); Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark (S.V.F.H., C.M.G.A., G.M., T.U.); and Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom (S.J.Y.)
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (K.R.W., B.D.H., E.A.-C., S.Z.R., J.D., S.J.Y., G.M.); Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark (S.V.F.H., C.M.G.A., G.M., T.U.); and Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom (S.J.Y.)
| |
Collapse
|
27
|
Houthuijzen JM, Oosterom I, Hudson BD, Hirasawa A, Daenen LGM, McLean CM, Hansen SVF, van Jaarsveld MTM, Peeper DS, Jafari Sadatmand S, Roodhart JML, van de Lest CHA, Ulven T, Ishihara K, Milligan G, Voest EE. Fatty acid 16:4(n-3) stimulates a GPR120-induced signaling cascade in splenic macrophages to promote chemotherapy resistance. FASEB J 2017; 31:2195-2209. [PMID: 28183801 PMCID: PMC5388545 DOI: 10.1096/fj.201601248r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/23/2017] [Indexed: 12/31/2022]
Abstract
Although chemotherapy is designed to eradicate tumor cells, it also has significant effects on normal tissues. The platinum-induced fatty acid 16:4(n-3) (hexadeca-4,7,10,13-tetraenoic acid) induces systemic resistance to a broad range of DNA-damaging chemotherapeutics. We show that 16:4(n-3) exerts its effect by activating splenic F4/80+/CD11blow macrophages, which results in production of chemoprotective lysophosphatidylcholines (LPCs). Pharmacologic studies, together with analysis of expression patterns, identified GPR120 on F4/80+/CD11blow macrophages as the relevant receptor for 16:4(n-3). Studies that used splenocytes from GPR120-deficient mice have confirmed this conclusion. Activation of the 16:4(n-3)-GPR120 axis led to enhanced cPLA2 activity in these splenic macrophages and secretion of the resistance-inducing lipid mediator, lysophosphatidylcholine(24:1). These studies identify a novel and unexpected function for GPR120 and suggest that antagonists of this receptor might be effective agents to limit development of chemotherapy resistance.—Houthuijzen, J. M., Oosterom, I., Hudson, B. D., Hirasawa, A., Daenen, L. G. M., McLean, C. M., Hansen, S. V. F., van Jaarsveld, M. T. M., Peeper, D. S., Jafari Sadatmand, S., Roodhart, J. M. L., van de Lest, C. H. A., Ulven, T., Ishihara, K., Milligan, G., Voest, E. E. Fatty acid 16:4(n-3) stimulates a GPR120-induced signaling cascade in splenic macrophages to promote chemotherapy resistance.
Collapse
Affiliation(s)
- Julia M Houthuijzen
- Department of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ilse Oosterom
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Brian D Hudson
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, University of Glasgow, Glasgow, United Kingdom
| | - Akira Hirasawa
- Department of Genomic Drug Discovery Science, Kyoto University, Kyoto, Japan
| | - Laura G M Daenen
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Chelsea M McLean
- Department of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Steffen V F Hansen
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark
| | | | - Daniel S Peeper
- Department of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sahar Jafari Sadatmand
- Department of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeanine M L Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Chris H A van de Lest
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Trond Ulven
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark
| | - Kenji Ishihara
- National Research Institute of Fisheries Science, Kanazawaku, Japan
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, University of Glasgow, Glasgow, United Kingdom
| | - Emile E Voest
- Department of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Hansen SVF, Ulven T. Pharmacological Tool Compounds for the Free Fatty Acid Receptor 4 (FFA4/GPR120). Handb Exp Pharmacol 2017; 236:33-56. [PMID: 27807695 DOI: 10.1007/164_2016_60] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The free fatty acid receptor 4 (FFA4), also known as GPR120, is a G protein-coupled receptor that is activated by long-chain fatty acids and that has been associated with regulation of appetite, release of insulin controlling hormones, insulin sensitization, anti-inflammatory and potentially anti-obesity activity, and is progressively appearing as an attractive potential target for the treatment of metabolic dysfunctions such as obesity, type 2 diabetes and inflammatory disorders. Ongoing investigations of the pharmacological functions of FFA4 and validation of its potential as a therapeutic target depend critically on the appropriateness and quality of the available pharmacological probes or tool compounds. After a brief summary of the pharmacological functions of FFA4 and some general considerations on desirable properties for these pharmacological tool compounds, the individual compounds that have been or are currently being used as tools for probing the function of FFA4 in various in vitro and in vivo settings will be discussed and evaluated.
Collapse
Affiliation(s)
- Steffen V F Hansen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Trond Ulven
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.
| |
Collapse
|
29
|
Song T, Zhou Y, Peng J, Tao YX, Yang Y, Xu T, Peng J, Ren J, Xiang Q, Wei H. GPR120 promotes adipogenesis through intracellular calcium and extracellular signal-regulated kinase 1/2 signal pathway. Mol Cell Endocrinol 2016; 434:1-13. [PMID: 27302893 DOI: 10.1016/j.mce.2016.06.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 05/24/2016] [Accepted: 06/04/2016] [Indexed: 12/22/2022]
Abstract
Numerous researches have demonstrated that GPR120 (also called FFAR4) exerts novel functions in insulin resistance and adipogenesis. However, the molecular mechanism of GPR120-mediated adipogenic differentiation is still unclear. This study was aimed to interpret the relevant function mechanism of GPR120 in the differentiation of 3T3-L1 adipocytes. The results showed that GPR120 expression was dramatically increased along with the adipogenic differentiation of 3T3-L1 adipocytes and the adipogenic ability was significantly inhibited in shGPR120-transfected cells. TUG-891, a selective agonist of GPR120, promoted the intracellular triglyceride accumulation in a dose-dependent manner and did not enhance adipogenesis in shGPR120-transfected cells. Markedly, TUG-891 increased the activation of PPARγ in a GPR120-dependent pathway as assessed by luciferase reporter assay. Furthermore, in the adipogenic differentiation process of 3T3-L1 adipocytes, TUG-891 increased the [Ca(2+)]i and phosphorylation level of ERK1/2. Pretreatment with inhibitors of either ERK1/2 (U0126) or [Ca(2+)]i (BAPTA-AM) notably attenuated the GPR120-mediated adipogenesis. These results show that GPR120 promotes adipogenesis by increasing PPARγ expression via [Ca(2+)]i and ERK1/2 signal pathway in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Tongxing Song
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Yuanfei Zhou
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, AL, 36849, USA
| | - Yang Yang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Tao Xu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Jie Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Jiao Ren
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Quanhang Xiang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| |
Collapse
|
30
|
Ahn SH, Park SY, Baek JE, Lee SY, Baek WY, Lee SY, Lee YS, Yoo HJ, Kim H, Lee SH, Im DS, Lee SK, Kim BJ, Koh JM. Free Fatty Acid Receptor 4 (GPR120) Stimulates Bone Formation and Suppresses Bone Resorption in the Presence of Elevated n-3 Fatty Acid Levels. Endocrinology 2016; 157:2621-35. [PMID: 27145004 DOI: 10.1210/en.2015-1855] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Free fatty acid receptor 4 (FFA4) has been reported to be a receptor for n-3 fatty acids (FAs). Although n-3 FAs are beneficial for bone health, a role of FFA4 in bone metabolism has been rarely investigated. We noted that FFA4 was more abundantly expressed in both mature osteoclasts and osteoblasts than their respective precursors and that it was activated by docosahexaenoic acid. FFA4 knockout (Ffar4(-/-)) and wild-type mice exhibited similar bone masses when fed a normal diet. Because fat-1 transgenic (fat-1(Tg+)) mice endogenously converting n-6 to n-3 FAs contain high n-3 FA levels, we crossed Ffar4(-/-) and fat-1(Tg+) mice over two generations to generate four genotypes of mice littermates: Ffar4(+/+);fat-1(Tg-), Ffar4(+/+);fat-1(Tg+), Ffar4(-/-);fat-1(Tg-), and Ffar4(-/-);fat-1(Tg+). Female and male littermates were included in ovariectomy- and high-fat diet-induced bone loss models, respectively. Female fat-1(Tg+) mice decreased bone loss after ovariectomy both by promoting osteoblastic bone formation and inhibiting osteoclastic bone resorption than their wild-type littermates, only when they had the Ffar4(+/+) background, but not the Ffar4(-/-) background. In a high-fat diet-fed model, male fat-1(Tg+) mice had higher bone mass resulting from stimulated bone formation and reduced bone resorption than their wild-type littermates, only when they had the Ffar4(+/+) background, but not the Ffar4(-/-) background. In vitro studies supported the role of FFA4 as n-3 FA receptor in bone metabolism. In conclusion, FFA4 is a dual-acting factor that increases osteoblastic bone formation and decreases osteoclastic bone resorption, suggesting that it may be an ideal target for modulating metabolic bone diseases.
Collapse
Affiliation(s)
- Seong Hee Ahn
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Sook-Young Park
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Ji-Eun Baek
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Su-Youn Lee
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Wook-Young Baek
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Sun-Young Lee
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Young-Sun Lee
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Hyun Ju Yoo
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Hyeonmok Kim
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Seung Hun Lee
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Dong-Soon Im
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Sun-Kyeong Lee
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Beom-Jun Kim
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| | - Jung-Min Koh
- Department of Endocrinology and Metabolism (S.H.A.), Inha University Hospital, Inha University School of Medicine, Incheon 402-751, South Korea; Asan Institute for Life Sciences (S.-Y.P., J.-E.B., S.-Youn.L., W.-.Y.B., S.-Young.L., Y.-S.L.) and Biomedical Research Center (H.J.Y.) and Division of Endocrinology and Metabolism (H.K., S.H.L., B.-J.K., J.-M.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Molecular Inflammation Research Center for Aging Intervention and College of Pharmacy (D.-S.I.), Pusan National University, Busan 609-735, South Korea; and UConn Center on Aging (S.-K.L.), University of Connecticut Health Center, Farmington, Connecticut 06030-1601
| |
Collapse
|
31
|
Eicosapentaenoic acid attenuates dexamethasome-induced apoptosis by inducing adaptive autophagy via GPR120 in murine bone marrow-derived mesenchymal stem cells. Cell Death Dis 2016; 7:e2235. [PMID: 27228350 PMCID: PMC4917672 DOI: 10.1038/cddis.2016.144] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 01/04/2023]
Abstract
Long-term use of glucocorticoids is a widespread clinical problem, which currently has no effective solution other than discontinuing the use. Eicosapentaenoic acid (EPA), an omega-3 long chain polyunsaturated fatty acid (n-3 PUFA), which is largely contained in fish or fish oil, has been reported to promote cell viability and improve bone metabolism. However, little is known about the effects of EPA on dexamethasome (Dex)-induced cell apoptosis. In this study, we showed that EPA-induced autophagy of murine bone marrow-derived mesenchymal stem cells (mBMMSCs). Meanwhile, EPA, but not arachidonic acid (AA), markedly inhibited Dex-induced apoptosis and promoted the viability of mBMMSCs. We also observed that EPA-induced autophagy was modulated by GPR120, but not GPR40. Further experiments showed that the mechanism of EPA-induced autophagy associated with GPR120 modulation involved an increase in the active form of AMP-activated protein kinase and a decrease in the activity of mammalian target of RAPA. The protective effect of EPA on Dex-induced apoptosis via GPR120-meditated induction of adaptive autophagy was supported by in vivo experiments. In summary, our findings may have important implications in developing future strategies to use EPA in the prevention and therapy of the side effects induced by long-term Dex-abuse.
Collapse
|
32
|
The Alternative Faces of Macrophage Generate Osteoclasts. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9089610. [PMID: 26977415 PMCID: PMC4761668 DOI: 10.1155/2016/9089610] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/14/2016] [Accepted: 01/18/2016] [Indexed: 01/08/2023]
Abstract
The understanding of how osteoclasts are generated and whether they can be altered by inflammatory stimuli is a topic of particular interest for osteoclastogenesis. It is known that the monocyte/macrophage lineage gives rise to osteoclasts (OCs) by the action of macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor-kB ligand (RANKL), which induce cell differentiation through their receptors, c-fms and RANK, respectively. The multinucleated giant cells (MGCs) generated by the engagement of RANK/RANKL are typical OCs. Nevertheless, very few studies have addressed the question of which subset of macrophages generates OCs. Indeed, two main subsets of macrophages are postulated, the inflammatory or classically activated type (M1) and the anti-inflammatory or alternatively activated type (M2). It has been proposed that macrophages can be polarized in vitro towards a predominantly M1 or M2 phenotype with the addition of granulocyte macrophage- (GM-) CSF or M-CSF, respectively. Various inflammatory stimuli known to induce macrophage polarization, such as LPS or TNF-α, can alter the type of MGC obtained from RANKL-induced differentiation. This review aims to highlight the role of immune-related stimuli and factors in inducing macrophages towards the osteoclastogenesis choice.
Collapse
|