1
|
Liang W, Yang H, Pan L, Wei S, Li Z, Zhang P, Li R, Wu Y, Liu M, Liu X. Ginkgo biloba Extract 50 (GBE50) Exerts Antifibrotic and Antioxidant Effects on Pulmonary Fibrosis in Mice by Regulating Nrf2 and TGF-β1/Smad Pathways. Appl Biochem Biotechnol 2024; 196:4807-4822. [PMID: 37971580 DOI: 10.1007/s12010-023-04755-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a progressive lung disorder with a poor prognosis. GBE50 is a new standardized Ginkgo biloba extract that has been widely used in cardiovascular diseases. However, the protective mechanism of GBE50 against PF remains to be elucidated. METHODS C57BL/6J mice were treated with bleomycin (Bleo) to induce PF in the presence or absence of GBE50. Protein content in bronchoalveolar lavage fluid (BALF) and wet weight/dry weight ratio were examined for analysis of pulmonary edema. Hematoxylin-eosin staining and Masson trichrome staining were used for histopathological observation of murine lung tissues. Ashcroft score was used for semi-quantitation of lung fibrosis degree. RT-qPCR was utilized for assessing mRNA levels of pro-fibrotic mediators in lung tissues. TUNEL staining was implemented for cell apoptosis assessment. The levels of oxidative stress- and inflammation-related markers were evaluated by corresponding commercial assay kits. Western blotting was used to evaluate levels of nuclear factor erythroid 2-related factor 2 (Nrf2) signaling- and transforming growth factor (TGF)-β1/SMAD signaling-related proteins. RESULTS GBE50 alleviated lung injury and severity of fibrosis, reduced collagen deposition and cell apoptosis in lung tissues, and suppressed inflammatory response and oxidative stress injury in Bleo-stimulated PF mice. GBE50 activated Nrf2 signaling pathway and inactivated TGF-β1/SMAD signaling pathway in the lungs of Bleo-induced PF mice. Inhibition of Nrf2 signaling reversed GBE50-mediated inactivation of TGF-β1/SMAD signaling and attenuation of inflammation and oxidative stress in Bleo-induced PF mice. CONCLUSION GBE50 protects against Bleo-induced PF in mice by mitigating fibrosis, inflammation and oxidative stress via Nrf2 and TGF-β1/SMAD signaling pathways.
Collapse
Affiliation(s)
- Wei Liang
- Department of Pulmonary and Critical Care Medicine, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, 10 Huadong Road, Nanning, 530000, Guangxi, China
| | - Hongmei Yang
- Department of Pulmonary and Critical Care Medicine, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, 10 Huadong Road, Nanning, 530000, Guangxi, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Ling Pan
- Department of Pulmonary and Critical Care Medicine, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, 10 Huadong Road, Nanning, 530000, Guangxi, China.
| | - Sizun Wei
- Department of Pulmonary and Critical Care Medicine, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, 10 Huadong Road, Nanning, 530000, Guangxi, China.
| | - Zhanhua Li
- Department of Pulmonary and Critical Care Medicine, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, 10 Huadong Road, Nanning, 530000, Guangxi, China
| | - Pengfei Zhang
- Department of Pulmonary and Critical Care Medicine, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, 545001, Guangxi, China
| | - Ruixiang Li
- Intensive Care Unit, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning530000, Guangxi, China
| | - Yangcong Wu
- Guangxi Traditional Chinese Medicine University, Nanning530000, Guangxi, China
| | - Maohua Liu
- Guangxi Traditional Chinese Medicine University, Nanning530000, Guangxi, China
| | - Xiaohong Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| |
Collapse
|
2
|
Vén K, Besztercei B, Janovicz A, Karsai N, Chun J, Tigyi G, Benyó Z, Ruisanchez É. LPA-Induced Thromboxane A2-Mediated Vasoconstriction Is Limited to Poly-Unsaturated Molecular Species in Mouse Aortas. Int J Mol Sci 2024; 25:6872. [PMID: 38999980 PMCID: PMC11241118 DOI: 10.3390/ijms25136872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
We have previously reported that, in aortic rings, 18:1 lysophosphatidic acid (LPA) can induce both vasodilation and vasoconstriction depending on the integrity of the endothelium. The predominant molecular species generated in blood serum are poly-unsaturated LPA species, yet the vascular effects of these species are largely unexplored. We aimed to compare the vasoactive effects of seven naturally occurring LPA species in order to elucidate their potential pathophysiological role in vasculopathies. Vascular tone was measured using myography, and thromboxane A2 (TXA2) release was detected by ELISA in C57Bl/6 mouse aortas. The Ca2+-responses to LPA-stimulated primary isolated endothelial cells were measured by Fluo-4 AM imaging. Our results indicate that saturated molecular species of LPA elicit no significant effect on the vascular tone of the aorta. In contrast, all 18 unsaturated carbon-containing (C18) LPAs (18:1, 18:2, 18:3) were effective, with 18:1 LPA being the most potent. However, following inhibition of cyclooxygenase (COX), these LPAs induced similar vasorelaxation, primarily indicating that the vasoconstrictor potency differed among these species. Indeed, C18 LPA evoked a similar Ca2+-signal in endothelial cells, whereas in endothelium-denuded aortas, the constrictor activity increased with the level of unsaturation, correlating with TXA2 release in intact aortas. COX inhibition abolished TXA2 release, and the C18 LPA induced vasoconstriction. In conclusion, polyunsaturated LPA have markedly increased TXA2-releasing and vasoconstrictor capacity, implying potential pathophysiological consequences in vasculopathies.
Collapse
Affiliation(s)
- Krisztina Vén
- Institute of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (K.V.); (B.B.); (A.J.); (N.K.); (G.T.); (Z.B.)
- Department of Neurology, Semmelweis University, 1085 Budapest, Hungary
| | - Balázs Besztercei
- Institute of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (K.V.); (B.B.); (A.J.); (N.K.); (G.T.); (Z.B.)
| | - Anna Janovicz
- Institute of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (K.V.); (B.B.); (A.J.); (N.K.); (G.T.); (Z.B.)
- HUN-REN-SU Cerebrovascular and Neurocognitive Disorders Research Group, 1094 Budapest, Hungary
| | - Noémi Karsai
- Institute of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (K.V.); (B.B.); (A.J.); (N.K.); (G.T.); (Z.B.)
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Gábor Tigyi
- Institute of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (K.V.); (B.B.); (A.J.); (N.K.); (G.T.); (Z.B.)
- Department of Physiology, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (K.V.); (B.B.); (A.J.); (N.K.); (G.T.); (Z.B.)
- HUN-REN-SU Cerebrovascular and Neurocognitive Disorders Research Group, 1094 Budapest, Hungary
| | - Éva Ruisanchez
- Institute of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (K.V.); (B.B.); (A.J.); (N.K.); (G.T.); (Z.B.)
- HUN-REN-SU Cerebrovascular and Neurocognitive Disorders Research Group, 1094 Budapest, Hungary
| |
Collapse
|
3
|
Ruzza C, Argentieri M, Ferrari F, Armani E, Trevisani M, Marchini G, Calo’ G. In vitro pharmacological characterization of standard and new lysophosphatidic acid receptor antagonists using dynamic mass redistribution assay. Front Pharmacol 2023; 14:1267414. [PMID: 38035009 PMCID: PMC10682101 DOI: 10.3389/fphar.2023.1267414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Lysophosphatidic acid (LPA) is a bioactive phospholipid that acts as an agonist of six G protein-coupled receptors named LPA receptors (LPA1-6). LPA elicits diverse intracellular events and modulates several biological functions, including cell proliferation, migration, and invasion. Overactivation of the LPA-LPA receptor system is reported to be involved in several pathologies, including cancer, neuropathic pain, fibrotic diseases, atherosclerosis, and type 2 diabetes. Thus, LPA receptor modulators may be clinically relevant in numerous diseases, making the identification and pharmacodynamic characterization of new LPA receptor ligands of strong interest. In the present work, label-free dynamic mass redistribution (DMR) assay has been used to evaluate the pharmacological activity of some LPA1 and LPA2 standard antagonists at the recombinant human LPA1 and LPA2 receptors. These results are compared to those obtained in parallel experiments with the calcium mobilization assay. Additionally, the same experimental protocol has been used for the pharmacological characterization of the new compound CHI. KI 16425, RO 6842262, and BMS-986020 behaved as LPA1 inverse agonists in DMR experiments and as LPA1 antagonists in calcium mobilization assays. Amgen compound 35 behaved as an LPA2 antagonist, while Merck compound 20 from WO2012028243 was detected as an LPA2 inverse agonist using the DMR test. Of note, for all the compounds, similar potency values were estimated by DMR and calcium assay. The new compound CHI was found to be an LPA1 inverse agonist, but with potency lower than that of the standard compounds. In conclusion, we have demonstrated that DMR assay can be successfully used to characterize LPA1 and LPA2 ligands. Compared to the classical calcium mobilization assay, DMR offers some advantages, in particular allowing the identification of inverse agonists. Finally, in the frame of this study, a new LPA1 inverse agonist has been identified.
Collapse
Affiliation(s)
- C. Ruzza
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
- LTTA Laboratory for Advanced Therapies, Technopole of Ferrara, Ferrara, Italy
| | - M. Argentieri
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - F. Ferrari
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - E. Armani
- Chiesi Farmaceutici SpA, Parma, Italy
| | | | | | - G. Calo’
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| |
Collapse
|
4
|
Ismail A, Abulsoud AI, Fathi D, Elshafei A, El-Mahdy HA, Elsakka EG, Aglan A, Elkhawaga SY, Doghish AS. The role of miRNAs in Ovarian Cancer Pathogenesis and Therapeutic Resistance - A Focus on Signaling Pathways Interplay. Pathol Res Pract 2022; 240:154222. [DOI: 10.1016/j.prp.2022.154222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/09/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022]
|
5
|
Wang X, Zhao S, Lai J, Guan W, Gao Y. Anti-Inflammatory, Antioxidant, and Antifibrotic Effects of Gingival-Derived MSCs on Bleomycin-Induced Pulmonary Fibrosis in Mice. Int J Mol Sci 2021; 23:ijms23010099. [PMID: 35008524 PMCID: PMC8745316 DOI: 10.3390/ijms23010099] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) intervention has been associated with lung protection. We attempted to determine whether mouse gingival-derived mesenchymal stem cells (GMSCs) could protect against bleomycin-induced pulmonary fibrosis. METHODS Mice were divided into three groups: control (Con), bleomycin (Bl), and bleomycin + MSCs (Bl + MSCs). Mice were treated with 5 mg/kg bleomycin via transtracheal instillation to induce pulmonary fibrosis. We assessed the following parameters: histopathological severity of injury in the lung, liver, kidney, and aortic tissues; the degree of pulmonary fibrosis; pulmonary inflammation; pulmonary oedema; profibrotic factor levels in bronchoalveolar lavage fluid (BALF) and lung tissue; oxidative stress-related indicators and apoptotic index in lung tissue; and gene expression levels of IL-1β, IL-8, TNF-α, lysophosphatidic acid (LPA), lysophosphatidic acid receptor 1 (LPA1), TGF-β, matrix metalloproteinase 9 (MMP-9), neutrophil elastase (NE), MPO, and IL-10 in lung tissue. RESULTS GMSC intervention attenuated bleomycin-induced pulmonary fibrosis, pulmonary inflammation, pulmonary oedema, and apoptosis. Bleomycin instillation notably increased expression levels of the IL-1β, IL-8, TNF-α, LPA, LPA1, TGF-β, MMP-9, NE, and MPO genes and attenuated expression levels of the IL-10 gene in lung tissue, and these effects were reversed by GMSC intervention. Bleomycin instillation notably upregulated MDA and MPO levels and downregulated GSH and SOD levels in lung tissue, and these effects were reversed by GMSC intervention. GMSC intervention prevented upregulation of neutrophil content in the lung, liver, and kidney tissues and the apoptotic index in lung tissue. CONCLUSIONS GMSC intervention exhibits anti-inflammatory and antioxidant capacities. Deleterious accumulation of neutrophils, which is reduced by GMSC intervention, is a key component of bleomycin-induced pulmonary fibrosis. GMSC intervention impairs bleomycin-induced NE, MMP-9, LPA, APL1, and TGF-β release.
Collapse
Affiliation(s)
- Xishuai Wang
- Department of Animal Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (X.W.); (S.Z.); (J.L.)
- College of P.E and Sport, Beijing Normal University, Beijing 100193, China
| | - Shiyu Zhao
- Department of Animal Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (X.W.); (S.Z.); (J.L.)
| | - Junhui Lai
- Department of Animal Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (X.W.); (S.Z.); (J.L.)
| | - Weijun Guan
- Department of Animal Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (X.W.); (S.Z.); (J.L.)
- Correspondence: (W.G.); (Y.G.)
| | - Yang Gao
- Institute of Physical Educational Training, Capital University of Physical Education and Sports, Beijing 100193, China
- Correspondence: (W.G.); (Y.G.)
| |
Collapse
|
6
|
Li Y, Sun Y, Wu N, Ma H. GRK2 promotes activation of lung fibroblast cells and contributes to pathogenesis of pulmonary fibrosis through increasing Smad3 expression. Am J Physiol Cell Physiol 2021; 322:C63-C72. [PMID: 34852209 DOI: 10.1152/ajpcell.00347.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pulmonary fibrosis is a chronic, progressive, and irreversible interstitial lung disease. Transforming growth factor beta1 (TGF-β1) plays a major role in lung fibroblast cell differentiation to myofibroblast cells and production of extracellular matrix, which are hallmarks of pulmonary fibrosis. G protein-coupled receptor kinase-2 (GRK2) has been shown to play controversial roles in TGF-β1-induced signal transduction in different cell types; however, the roles of GRK2 in TGF-β1-induced activation of lung fibroblast cells and development of pulmonary fibrosis have not been revealed. In this study, we found that GRK2 levels were induced in lungs and isolated fibroblast cells in a murine model of pulmonary fibrosis, as well as TGF-β1-treated lung fibroblasts. GRK2 levels were not changed in lungs in the injury phase of pulmonary fibrosis. Post-treatment with GRK2 inhibitor reduced ECM accumulation in lungs in bleomycin-challenged mice, suggesting that GRK2 activation contributes to the progressive phase of pulmonary fibrosis. Inhibition or downregulation of GRK2 attenuates fibronectin, collagen, and α-smooth muscle actin expression in TGF-β1-induced lung fibroblast cells or myofibroblast cells isolated from pulmonary fibrosis patients. Further, we showed that GRK2 regulates Smad3 expression, indicating that inhibition of GRK2 attenuates ECM accumulation through downregulation of Smad3 expression. This study reveals that GRK2 is a therapeutic target in treating pulmonary fibrosis and inhibition of GRK2 dampens pulmonary fibrosis by suppression of Smad3 expression, eventually attenuating TGF-β1 signal pathway and ECM accumulation.
Collapse
Affiliation(s)
- Yanhui Li
- Department of Anesthesia, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Sun
- Department of Respiratory and Critical Care Medicine, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Nan Wu
- Department of Anesthesia, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Haichun Ma
- Department of Anesthesia, the First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
7
|
Zheng Q, Dai X, Fang W, Zheng Y, Zhang J, Liu Y, Gu D. Overexpression of microRNA-367 inhibits angiogenesis in ovarian cancer by downregulating the expression of LPA1. Cancer Cell Int 2020; 20:476. [PMID: 33024414 PMCID: PMC7531134 DOI: 10.1186/s12935-020-01551-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023] Open
Abstract
Background Compelling evidences reported the role of microRNAs (miRNAs) in ovarian cancer. However, little was known regarding the molecular mechanism of miR-367 in ovarian cancer. This study intended to investigate the role and regulatory mechanism of miR-367 in ovarian cancer involving lysophosphatidic acid receptor-1 (LPA1). Methods Potentially regulatory miRNAs in ovarian cancer were obtained from bioinformatics analysis. RT-qPCR was used to detect miR-367 expression in both ovarian cancer tissues and relevant adjacent normal tissues. Relationship between miR-367 and LPA1 was predicted by miRNA database and further verified using dual luciferase reporter gene assay and RIP. EdU and Transwell assay were used to measure the proliferation and invasion ability of cells. Moreover, tube formation and chick chorioallantois membrane (CAM) assay were performed to determine angiogenesis of human umbilical vein endothelial cells (HUVECs). Finally, the roles of LPA1 in tumor growth was also studied using nude mice xenograft assay. Results High expression of LPA1 and low expression of miR-367 were observed in ovarian cancer tissues and cells. Overexpressed miR-367 downregulated LPA1 expression to inhibit proliferation, invasion, and angiogenesis of cancer cells. Low expression of LPA1 suppressed tumor formation and repressed angiogenesis in ovarian in vivo. Conclusion All in all, overexpression of miR-367 downregulated LPA1 expression to inhibit ovarian cancer progression, which provided a target for the cancer treatment.
Collapse
Affiliation(s)
- Qingling Zheng
- Department of Obstetrics and Gynecology, School of Medicine and Nursing Sciences, Huzhou University, Huzhou Central Hospital, Huzhou, 313000 People's Republic of China
| | - Xin Dai
- Department of Pathology, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, No. 1, Lijiang Road, Huqiu District, Suzhou, 215153 Jiangsu People's Republic of China
| | - Wei Fang
- Department of Pathology, Huzhou Central Hospital, Huzhou, 313000 People's Republic of China
| | - Yan Zheng
- Department of Pathology, Huzhou Central Hospital, Huzhou, 313000 People's Republic of China
| | - Jin Zhang
- Department of Pathology, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, No. 1, Lijiang Road, Huqiu District, Suzhou, 215153 Jiangsu People's Republic of China
| | - Yanxiang Liu
- Department of Pathology, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, No. 1, Lijiang Road, Huqiu District, Suzhou, 215153 Jiangsu People's Republic of China
| | - Donghua Gu
- Department of Pathology, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, No. 1, Lijiang Road, Huqiu District, Suzhou, 215153 Jiangsu People's Republic of China
| |
Collapse
|
8
|
Antidepressants induce profibrotic responses via the lysophosphatidic acid receptor LPA 1. Eur J Pharmacol 2020; 873:172963. [PMID: 32007501 DOI: 10.1016/j.ejphar.2020.172963] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/16/2019] [Accepted: 01/27/2020] [Indexed: 12/29/2022]
Abstract
Preclinical and clinical studies have indicated that antidepressants can promote inflammation and fibrogenesis, particularly in the lung, by mechanisms not fully elucidated. We have previously shown that different classes of antidepressants can activate the lysophosphatidic acid (LPA) receptor LPA1, a major pathogenetic mediator of tissue fibrosis. The aim of the present study was to investigate whether in cultured human dermal and lung fibroblasts antidepressants could trigger LPA1-mediated profibrotic responses. In both cell types amitriptyline, clomipramine and mianserin mimicked the ability of LPA to induce the phosphorylation/activation of extracellular signal -regulated kinases 1 and 2 (ERK1/2), which was blocked by the selective LPA1 receptor antagonist AM966 and the LPA1/3 antagonist Ki16425. Antidepressant-induced ERK1/2 stimulation was absent in fibroblasts stably depleted of LPA1 by short hairpin RNA transfection and was prevented by pertussis toxin, an uncoupler of receptors from Gi/o proteins. Like LPA, antidepressants stimulated fibroblasts proliferation and this effect was blocked by either AM966 or the MEK1/2 inhibitor PD98059. Moreover, by acting through LPA1 antidepressants induced the expression of α-smooth muscle actin (α-SMA), a marker of myofibroblast differentiation, and caused an ERK1/2-dependent increase in the cellular levels of transforming growth factor-β (TGF-β)1, a potent fibrogenic cytokine. Pharmacological blockade of TGF-β receptor type 1 prevented antidepressant- and LPA-induced α-SMA expression. These data indicate that in human dermal and lung fibroblasts different antidepressants can induce proliferative and differentiating responses by activating the LPA1 receptor coupled to ERK1/2 signalling and suggest that this property may contribute to the promotion of tissue fibrosis by these drugs.
Collapse
|
9
|
Audet M, Stevens RC. Emerging structural biology of lipid G protein-coupled receptors. Protein Sci 2019; 28:292-304. [PMID: 30239054 PMCID: PMC6319753 DOI: 10.1002/pro.3509] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 09/07/2018] [Accepted: 09/10/2018] [Indexed: 01/14/2023]
Abstract
The first crystal structure of a G protein-coupled receptor (GPCR) was that of the bovine rhodopsin, solved in 2000, and is a light receptor within retina rode cells that enables vision by transducing a conformational signal from the light-induced isomerization of retinal covalently bound to the receptor. More than 7 years after this initial discovery and following more than 20 years of technological developments in GPCR expression, stabilization, and crystallography, the high-resolution structure of the adrenaline binding β2 -adrenergic receptor, a ligand diffusible receptor, was discovered. Since then, high-resolution structures of more than 53 unique GPCRs have been determined leading to a significant improvement in our understanding of the basic mechanisms of ligand-binding and ligand-mediated receptor activation that revolutionized the field of structural molecular pharmacology of GPCRs. Recently, several structures of eight unique lipid-binding receptors, one of the most difficult GPCR families to study, have been reported. This review presents the outstanding structural and pharmacological features that have emerged from these new lipid receptor structures. The impact of these findings goes beyond mechanistic insights, providing evidence of the fundamental role of GPCRs in the physiological integration of the lipid signaling system, and highlighting the importance of sustained research into the structural biology of GPCRs for the development of new therapeutics targeting lipid receptors.
Collapse
Affiliation(s)
- Martin Audet
- Departments of Biological Sciences and ChemistryBridge Institute, Michelson Center for Convergent Bioscience, University of Southern CaliforniaLos AngelesCalifornia90089
| | - Raymond C. Stevens
- Departments of Biological Sciences and ChemistryBridge Institute, Michelson Center for Convergent Bioscience, University of Southern CaliforniaLos AngelesCalifornia90089
| |
Collapse
|
10
|
Kuwajima K, Sumitani M, Kurano M, Kano K, Nishikawa M, Uranbileg B, Tsuchida R, Ogata T, Aoki J, Yatomi Y, Yamada Y. Lysophosphatidic acid is associated with neuropathic pain intensity in humans: An exploratory study. PLoS One 2018; 13:e0207310. [PMID: 30408112 PMCID: PMC6224112 DOI: 10.1371/journal.pone.0207310] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/29/2018] [Indexed: 12/15/2022] Open
Abstract
The underlying mechanisms of neuropathic pain remain to be elucidated. Basic animal research has suggested that lysophosphatidic acids, which are bioactive lipids produced by autotaxin from lysophosphatidylcholine, may play key roles in the initiation and maintenance of neuropathic pain. Here, we investigated the clinical relevance of lysophosphatidic acids signaling on neuropathic pain in humans. Eighteen patients who had been diagnosed with neuropathic pain with varied etiologies participated in the study. Cerebrospinal fluid samples were obtained by lumbar puncture and the concentrations of 12 species of lysophosphatidic acids and lysophosphatidylcholine, autotaxin, and the phosphorylated neurofilament heavy subunit were measured. Pain symptoms were assessed using an 11-point numeric rating scale and the Neuropathic Pain Symptom Inventory regarding intensity and descriptive dimensions of neuropathic pain. The total lysophosphatidic acids were significantly associated with both pain intensity and symptoms. 18:1 and 20:4 lysophosphatidic acids in particular demonstrated the most correlations with dimensions of pain symptoms. Autotaxin and the phosphorylated neurofilament heavy subunit showed no association with pain symptoms. In conclusions, lysophosphatidic acids were significantly associated with pain symptoms in neuropathic pain patients. These results suggest that lysophosphatidic acids signaling might be a potential therapeutic target for neuropathic pain.
Collapse
Affiliation(s)
- Ken Kuwajima
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Masahiko Sumitani
- Department of Pain and Palliative Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Kuniyuki Kano
- Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Masako Nishikawa
- Department of Clinical Laboratory Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Rikuhei Tsuchida
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Toru Ogata
- Department of Rehabilitation for Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Saitama, Japan
| | - Junken Aoki
- Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Yoshitsugu Yamada
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
11
|
Carmona-Rosas G, Alfonzo-Méndez MA, Hernández-Espinosa DA, Romero-Ávila MT, García-Sáinz JA. A549 cells as a model to study endogenous LPA 1 receptor signaling and regulation. Eur J Pharmacol 2017; 815:258-265. [DOI: 10.1016/j.ejphar.2017.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/29/2017] [Accepted: 09/12/2017] [Indexed: 12/12/2022]
|
12
|
Sattikar A, Dowling MR, Rosethorne EM. Endogenous lysophosphatidic acid (LPA 1 ) receptor agonists demonstrate ligand bias between calcium and ERK signalling pathways in human lung fibroblasts. Br J Pharmacol 2017; 174:227-237. [PMID: 27864940 DOI: 10.1111/bph.13671] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/09/2016] [Accepted: 11/10/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Human lung fibroblasts (HLF) express high levels of the LPA1 receptor, a GPCR that responds to the endogenous lipid mediator, lysophosphatidic acid (LPA). Several molecular species or analogues of LPA exist and have been detected in biological fluids such as serum and plasma. The most widely expressed of the LPA receptor family is the LPA1 receptor, which predominantly couples to Gq/11 , Gi/o and G12/13 proteins. This promiscuity of coupling raises the possibility that some of the LPA analogues may bias the LPA1 receptor towards one signalling pathway over another. EXPERIMENTAL APPROACH Here, we have explored the signalling profiles of a range of LPA analogues in HLF that endogenously express the LPA1 receptor. HLF were treated with LPA analogues and receptor activation monitored via calcium mobilization and ERK phosphorylation. KEY RESULTS These analyses demonstrated that the 16:0, 17:0, 18:2 and C18:1 LPA analogues appear to exhibit ligand bias between ERK phosphorylation and calcium mobilization when compared with 18:1 LPA, one of the most abundant forms of LPA that has been found in human plasma. CONCLUSION AND IMPLICATIONS The importance of LPA as a key signalling molecule is shown by its widespread occurrence in biological fluids and its association with disease conditions such as fibrosis and cancer. These findings have important, as yet unexplored, implications for the (patho-) physiological signalling of the LPA1 receptor, as it may be influenced not only by the concentration of endogenous ligand but the isoform as well.
Collapse
Affiliation(s)
- Afrah Sattikar
- Novartis Institutes for Biomedical Research, Horsham, UK
| | - Mark R Dowling
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Elizabeth M Rosethorne
- Novartis Institutes for Biomedical Research, Horsham, UK.,School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|