1
|
Lei C, Zhou Q, Lv L, Liu D, Qian H. Inhibition of GPR4 Attenuates the Formation of Abdominal Aortic Aneurysm Through Inhibiting the SP-1/VEGF-A Signaling. J Biochem Mol Toxicol 2025; 39:e70118. [PMID: 39799555 DOI: 10.1002/jbt.70118] [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: 09/17/2024] [Revised: 11/30/2024] [Accepted: 12/21/2024] [Indexed: 01/15/2025]
Abstract
Abdominal aortic aneurysm (AAA) is a severe cardiovascular disease (CVD) that is partly attributable to endothelial dysfunction, inflammatory response, and angiogenesis. G protein-coupled receptor 4 (GPR4), a proton-sensitive G protein-coupled receptor that is abundantly expressed in vascular endothelial cells, has been associated with numerous physiological functions. Nevertheless, its potential involvement in the development of AAA remains unexplored. In this study, we examined the impact of GPR4 deletion on the development of AAA in ApoE-deficient mice. The mice were categorized into four distinct groups: the ApoE-/- with saline group, the ApoE-/-GPR4-/- with saline group, the ApoE-/- with Ang II group, and the ApoE-/-GPR4-/- with Ang II group. AAA were induced in the ApoE-/- mice through the perfusion of angiotensin II (Ang II). Notably, GPR4 was substantially elevated in the AAA tissues from both human subjects and experimental mice. The deletion of GPR4 substantially decreased the formation of Ang II-induced AAA, damages to elastin, and the expression of aortic inflammatory cytokines interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α), as well as vascular endothelial growth factor A/vascular endothelial growth factor receptor 2 (VEGF-A/VEGF-R2), in ApoE-/- mice. Human aortic endothelial cells (HAECs) were transfected with lenti-viral GPR4 shRNA and subsequently stimulated with Ang II. Our findings indicate that the knockout of GPR4 attenuated Ang II-induced angiogenic tube formation in HAECs by decreasing the expression of VEGF-A and VEGF-R2. Furthermore, GPR4 knockout also hindered the activation of specificity protein-1 (SP-1) by reducing its expression and transcriptional activity. Notably, the overexpression of SP-1 reversed the inhibitory effects of GPR4 knockout on angiogenic tube formation and the expression of VEGF-A/VEGF-R2. This suggests that the protective effects of GPR4 knockout are achieved through the inhibition of SP-1. In summary, the absence of GPR4 impeded AAA formation, indicating that GPR4 could potentially serve as a therapeutic target for AAA.
Collapse
MESH Headings
- Animals
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/prevention & control
- Mice
- Humans
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor A/genetics
- Signal Transduction
- Sp1 Transcription Factor/metabolism
- Sp1 Transcription Factor/genetics
- Male
- Mice, Knockout
- Angiotensin II/metabolism
- Mice, Knockout, ApoE
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Chenggang Lei
- Department of Cardiothoracic Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| | - Qian Zhou
- Department of Cardiothoracic Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| | - Lizhen Lv
- Department of Cardiothoracic Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| | - Di Liu
- Department of Cardiothoracic Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| | - Haiyun Qian
- Department of Cardiothoracic Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| |
Collapse
|
2
|
Prabutzki P, Schiller J, Engel KM. Phospholipid-derived lysophospholipids in (patho)physiology. Atherosclerosis 2024; 398:118569. [PMID: 39227208 DOI: 10.1016/j.atherosclerosis.2024.118569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/17/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024]
Abstract
Phospholipids (PL) are major components of cellular membranes and changes in PL metabolism have been associated with the pathogenesis of numerous diseases. Lysophosphatidylcholine (LPC) in particular, is a comparably abundant component of oxidatively damaged tissues. LPC originates from the cleavage of phosphatidylcholine (PC) by phospholipase A2 or the reaction of lipids with reactive oxygen species (ROS) such as HOCl. Another explanation of increased LPC concentration is the decreased re-acylation of LPC into PC. While there are also several other lysophospholipids, LPC is the most abundant lysophospholipid in mammals and will therefore be the focus of this review. LPC is involved in many physiological processes. It induces the migration of lymphocytes, fostering the production of pro-inflammatory compounds by inducing oxidative stress. LPC also "signals" via G protein-coupled and Toll-like receptors and has been implicated in the development of different diseases. However, LPCs are not purely "bad": this is reflected by the fact that the concentration and fatty acyl composition of LPC varies under different conditions, in plasma of healthy and diseased individuals, in tissues and different tumors. Targeting LPC and lipid metabolism and restoring homeostasis might be a potential therapeutic method for inflammation-related diseases.
Collapse
Affiliation(s)
- Patricia Prabutzki
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Härtelstr. 16-18, D 04107 Leipzig, Germany
| | - Jürgen Schiller
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Härtelstr. 16-18, D 04107 Leipzig, Germany
| | - Kathrin M Engel
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Härtelstr. 16-18, D 04107 Leipzig, Germany.
| |
Collapse
|
3
|
Liang S, Zhou J, Cao C, Liu Y, Ming S, Liu X, Shang Y, Lao J, Peng Q, Yang J, Wu M. GITR exacerbates lysophosphatidylcholine-induced macrophage pyroptosis in sepsis via posttranslational regulation of NLRP3. Cell Mol Immunol 2024; 21:674-688. [PMID: 38740925 PMCID: PMC11214634 DOI: 10.1038/s41423-024-01170-w] [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: 11/03/2023] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
The NLRP3 inflammasome functions as an inflammatory driver, but its relationship with lipid metabolic changes in early sepsis remains unclear. Here, we found that GITR expression in monocytes/macrophages was induced by lysophosphatidylcholine (LPC) and was positively correlated with the severity of sepsis. GITR is a costimulatory molecule that is mainly expressed on T cells, but its function in macrophages is largely unknown. Our in vitro data showed that GITR enhanced LPC uptake by macrophages and specifically enhanced NLRP3 inflammasome-mediated macrophage pyroptosis. Furthermore, in vivo studies using either cecal ligation and puncture (CLP) or LPS-induced sepsis models demonstrated that LPC exacerbated sepsis severity/lethality, while conditional knockout of GITR in myeloid cells or NLRP3/caspase-1/IL-1β deficiency attenuated sepsis severity/lethality. Mechanistically, GITR specifically enhanced inflammasome activation by regulating the posttranslational modification (PTM) of NLRP3. GITR competes with NLRP3 for binding to the E3 ligase MARCH7 and recruits MARCH7 to induce deacetylase SIRT2 degradation, leading to decreasing ubiquitination but increasing acetylation of NLRP3. Overall, these findings revealed a novel role of macrophage-derived GITR in regulating the PTM of NLRP3 and systemic inflammatory injury, suggesting that GITR may be a potential therapeutic target for sepsis and other inflammatory diseases. GITR exacerbates LPC-induced macrophage pyroptosis in sepsis via posttranslational regulation of NLRP3. According to the model, LPC levels increase during the early stage of sepsis, inducing GITR expression on macrophages. GITR not only competes with NLRP3 for binding to the E3 ligase MARCH7 but also recruits MARCH7 to induce the degradation of the deacetylase SIRT2, leading to decreasing ubiquitination but increasing acetylation of NLRP3 and therefore exacerbating LPC-induced NLRP3 inflammasome activation, macrophage pyroptosis and systemic inflammatory injury.
Collapse
Affiliation(s)
- Siping Liang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Jinyu Zhou
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Can Cao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Yiting Liu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Siqi Ming
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Xi Liu
- Department of Infectious Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yuqi Shang
- Department of Infectious Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Juanfeng Lao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Qin Peng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Jiahui Yang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Minhao Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| |
Collapse
|
4
|
Janovicz A, Majer A, Kosztelnik M, Geiszt M, Chun J, Ishii S, Tigyi GJ, Benyó Z, Ruisanchez É. Autotaxin-lysophosphatidic acid receptor 5 axis evokes endothelial dysfunction via reactive oxygen species signaling. Exp Biol Med (Maywood) 2023; 248:1887-1894. [PMID: 37837357 PMCID: PMC10792427 DOI: 10.1177/15353702231199081] [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: 12/28/2022] [Accepted: 07/29/2023] [Indexed: 10/16/2023] Open
Abstract
Lysophosphatidylcholine (LPC) is a bioactive lipid that has been shown to attenuate endothelium-dependent vasorelaxation contributing to endothelial dysfunction; however, the underlying mechanisms are not well understood. In this study, we investigated the molecular mechanisms involved in the development of LPC-evoked impairment of endothelium-dependent vasorelaxation. In aortic rings isolated from wild-type (WT) mice, a 20-min exposure to LPC significantly reduced the acetylcholine chloride (ACh)-induced vasorelaxation indicating the impairment of normal endothelial function. Interestingly, pharmacological inhibition of autotaxin (ATX) by GLPG1690 partially reversed the endothelial dysfunction, suggesting that lysophosphatidic acid (LPA) derived from LPC may be involved in the effect. Therefore, the effect of LPC was also tested in aortic rings isolated from different LPA receptor knock-out (KO) mice. LPC evoked a marked reduction in ACh-dependent vasorelaxation in Lpar1, Lpar2, and Lpar4 KO, but its effect was significantly attenuated in Lpar5 KO vessels. Furthermore, addition of superoxide dismutase reduced the LPC-induced endothelial dysfunction in WT but not in the Lpar5 KO mice. In addition, LPC increased H2O2 release from WT vessels, which was significantly reduced in Lpar5 KO vessels. Our findings indicate that the ATX-LPA-LPA5 receptor axis is involved in the development of LPC-induced impairment of endothelium-dependent vasorelaxation via LPA5 receptor-mediated reactive oxygen species production. Taken together, in this study, we identified a new pathway contributing to the development of LPC-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Anna Janovicz
- Institute of Translational Medicine, Semmelweis University, H-1094 Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Cerebrovascular and Neurocognitive Disorders Research Group, H-1052 Budapest, Hungary
| | - Aliz Majer
- Institute of Translational Medicine, Semmelweis University, H-1094 Budapest, Hungary
| | - Mónika Kosztelnik
- Institute of Translational Medicine, Semmelweis University, H-1094 Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Cerebrovascular and Neurocognitive Disorders Research Group, H-1052 Budapest, Hungary
| | - Miklós Geiszt
- Department of Physiology, Faculty of Medicine, Semmelweis University, H-1094 Budapest, Hungary
| | - Jerold Chun
- Translational Neuroscience at Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Satoshi Ishii
- Department of Immunology, Graduate School of Medicine, Akita University, Akita 010-8543, Japan
| | - Gábor József Tigyi
- Institute of Translational Medicine, Semmelweis University, H-1094 Budapest, Hungary
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, H-1094 Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Cerebrovascular and Neurocognitive Disorders Research Group, H-1052 Budapest, Hungary
| | - Éva Ruisanchez
- Institute of Translational Medicine, Semmelweis University, H-1094 Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Cerebrovascular and Neurocognitive Disorders Research Group, H-1052 Budapest, Hungary
| |
Collapse
|
5
|
Liu X, Liu H, Deng Y. Efferocytosis: An Emerging Therapeutic Strategy for Type 2 Diabetes Mellitus and Diabetes Complications. J Inflamm Res 2023; 16:2801-2815. [PMID: 37440994 PMCID: PMC10335275 DOI: 10.2147/jir.s418334] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Increasing evidence indicates that chronic, low-grade inflammation is a significant contributor to the fundamental pathogenesis of type 2 diabetes mellitus (T2DM). Efferocytosis, an effective way to eliminate apoptotic cells (ACs), plays a critical role in inflammation resolution. Massive accumulation of ACs and the proliferation of persistent inflammation caused by defective efferocytosis have been proven to be closely associated with pancreatic islet β cell destruction, adipose tissue inflammation, skeletal muscle dysfunction, and liver metabolism abnormalities, which together are considered the most fundamental pathological mechanism underlying T2DM. Therefore, here we outline the association between the molecular mechanisms of efferocytosis in glucose homeostasis, T2DM, and its complications, and we analyzed the present constraints and potential future prospects for therapeutic targets in T2DM and its complications.
Collapse
Affiliation(s)
- Xun Liu
- Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Hua Liu
- Southern Theater General Hospital of the Chinese People’s Liberation Army, Guangzhou, Guangdong, 510010, People’s Republic of China
| | - Yihui Deng
- Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| |
Collapse
|
6
|
Hung CH, Chin Y, Fong YO, Lee CH, Han DS, Lin JH, Sun WH, Chen CC. Acidosis-related pain and its receptors as targets for chronic pain. Pharmacol Ther 2023; 247:108444. [PMID: 37210007 DOI: 10.1016/j.pharmthera.2023.108444] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/24/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Sensing acidosis is an important somatosensory function in responses to ischemia, inflammation, and metabolic alteration. Accumulating evidence has shown that acidosis is an effective factor for pain induction and that many intractable chronic pain diseases are associated with acidosis signaling. Various receptors have been known to detect extracellular acidosis and all express in the somatosensory neurons, such as acid sensing ion channels (ASIC), transient receptor potential (TRP) channels and proton-sensing G-protein coupled receptors. In addition to sense noxious acidic stimulation, these proton-sensing receptors also play a vital role in pain processing. For example, ASICs and TRPs are involved in not only nociceptive activation but also anti-nociceptive effects as well as some other non-nociceptive pathways. Herein, we review recent progress in probing the roles of proton-sensing receptors in preclinical pain research and their clinical relevance. We also propose a new concept of sngception to address the specific somatosensory function of acid sensation. This review aims to connect these acid-sensing receptors with basic pain research and clinical pain diseases, thus helping with better understanding the acid-related pain pathogenesis and their potential therapeutic roles via the mechanism of acid-mediated antinociception.
Collapse
Affiliation(s)
- Chih-Hsien Hung
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yin Chin
- Department of Life Science & Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-On Fong
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Han Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Der-Shen Han
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
| | - Jiann-Her Lin
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan; Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Wei-Hsin Sun
- Department of Life Science & Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan; Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
7
|
Patients with Bacterial Sepsis Are Heterogeneous with Regard to Their Systemic Lipidomic Profiles. Metabolites 2022; 13:metabo13010052. [PMID: 36676977 PMCID: PMC9864715 DOI: 10.3390/metabo13010052] [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: 11/14/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. In the present study, we investigated the systemic/serum lipidomic profile at the time of hospital admission for patients with bacterial sepsis. The study included 60 patients; 35 patients fulfilled the most recent 2016 Sepsis-3 criteria (referred to as Sepsis-3) whereas the remaining 25 patients had sepsis only according to the previous Sepsis-2 definition and could be classified as having Systemic Inflammatory Response Syndrome (SIRS). A total of 966 lipid metabolites were identified. Patients fulfilling the Sepsis-3 criteria differed from the Sepsis-2 patients with regard to only 15 lipid metabolites, and especially sphingolipids metabolism differed between these patient subsets. A total of only 43 metabolites differed between patients with and without bacteremia, including 12 lysophosphatidylcholines and 18 triacylglycerols (15 C18/C20 fatty acid metabolites decreased and three C14 myristate acid metabolites that were increased in bacteremia). Unsupervised hierarchical clustering analyses based on the identified sphingolipids, phosphatidylcholine and triacylglycerols showed that (i) the majority of Sepsis-3 patients differed from SIRS patients especially with regard to lysophosphatidylcholine levels; (ii) the minority of Sepsis-3 patients that clustered together with the majority of SIRS patients showed lower Sequential Organ Failure Assessment (SOFA) scores than the other Sepsis-3 patients; and (iii) the variation between the patients in the identified/altered sphingolipid and triacylglycerol metabolites further increased the heterogeneity of Sepsis-3 patients with regard to their systemic lipidomic profile at the time of diagnosis. To conclude, patients fulfilling the Sepsis-3 criteria differ with regard to their metabolic profile, and this variation depends on disease severity.
Collapse
|
8
|
Imenez Silva PH, Câmara NO, Wagner CA. Role of proton-activated G protein-coupled receptors in pathophysiology. Am J Physiol Cell Physiol 2022; 323:C400-C414. [PMID: 35759438 DOI: 10.1152/ajpcell.00114.2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Local acidification is a common feature of many disease processes such as inflammation, infarction, or solid tumor growth. Acidic pH is not merely a sequelae of disease but contributes to recruitment and regulation of immune cells, modifies metabolism of parenchymal, immune and tumor cells, modulates fibrosis, vascular permeability, oxygen availability and consumption, invasiveness of tumor cells, and impacts on cell survival. Thus, multiple pH-sensing mechanisms must exist in cells involved in these processes. These pH-sensors play important roles in normal physiology and pathophysiology, and hence might be attractive targets for pharmacological interventions. Among the pH-sensing mechanisms, OGR1 (GPR68), GPR4 (GPR4), and TDAG8 (GPR65) have emerged as important molecules. These G protein-coupled receptors are widely expressed, are upregulated in inflammation and tumors, sense changes in extracellular pH in the range between pH 8 and 6, and are involved in modulating key processes in inflammation, tumor biology, and fibrosis. This review discusses key features of these receptors and highlights important disease states and pathways affected by their activity.
Collapse
Affiliation(s)
- Pedro H Imenez Silva
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,National Center of Competence in Research NCCR Kidney.CH, Switzerland
| | - Niels Olsen Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,National Center of Competence in Research NCCR Kidney.CH, Switzerland
| |
Collapse
|
9
|
Imenez Silva PH, Wagner CA. Physiological relevance of proton-activated GPCRs. Pflugers Arch 2022; 474:487-504. [PMID: 35247105 PMCID: PMC8993716 DOI: 10.1007/s00424-022-02671-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022]
Abstract
The detection of H+ concentration variations in the extracellular milieu is accomplished by a series of specialized and non-specialized pH-sensing mechanisms. The proton-activated G protein-coupled receptors (GPCRs) GPR4 (Gpr4), TDAG8 (Gpr65), and OGR1 (Gpr68) form a subfamily of proteins capable of triggering intracellular signaling in response to alterations in extracellular pH around physiological values, i.e., in the range between pH 7.5 and 6.5. Expression of these receptors is widespread for GPR4 and OGR1 with particularly high levels in endothelial cells and vascular smooth muscle cells, respectively, while expression of TDAG8 appears to be more restricted to the immune compartment. These receptors have been linked to several well-studied pH-dependent physiological activities including central control of respiration, renal adaption to changes in acid-base status, secretion of insulin and peripheral responsiveness to insulin, mechanosensation, and cellular chemotaxis. Their role in pathological processes such as the genesis and progression of several inflammatory diseases (asthma, inflammatory bowel disease), and tumor cell metabolism and invasiveness, is increasingly receiving more attention and makes these receptors novel and interesting targets for therapy. In this review, we cover the role of these receptors in physiological processes and will briefly discuss some implications for disease processes.
Collapse
Affiliation(s)
- Pedro H Imenez Silva
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
- National Center of Competence in Research NCCR Kidney.CH, Zurich, Switzerland.
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
- National Center of Competence in Research NCCR Kidney.CH, Zurich, Switzerland.
| |
Collapse
|
10
|
Parra-Millán R, Jiménez-Mejías ME, Ayerbe-Algaba R, Domínguez-Herrera J, Díaz C, Pérez Del Palacio J, Pachón J, Smani Y. Impact of the immune response modification by lysophosphatidylcholine in the efficacy of antibiotic therapy of experimental models of peritoneal sepsis and pneumonia by Pseudomonas aeruginosa: LPC therapeutic effect in combined therapy. ENFERMEDADES INFECCIOSAS Y MICROBIOLOGIA CLINICA (ENGLISH ED.) 2022; 40:14-21. [PMID: 34991848 DOI: 10.1016/j.eimce.2020.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/15/2020] [Indexed: 06/14/2023]
Abstract
INTRODUCTION Immune response stimulation may be an adjuvant to antimicrobial treatment. Here, we evaluated the impact of immune response modification by lysophosphatidylcholine (LPC), combined with imipenem or ceftazidime, in murine models of peritoneal sepsis (PS) and pneumonia induced by Pseudomonas aeruginosa. METHODS The imipenem and ceftazidime-susceptible strain (Pa39) and imipenem and ceftazidime-resistant strain (Pa238) were used. Ceftazidime pharmacokinetic and pharmacodynamic parameters were determined. The therapeutic efficacy and TNF-α and IL-10 levels were determined in murine models of PS and pneumonia induced by Pa39 and Pa238 and treated with LPC, imipenem or ceftazidime, alone or in combination. RESULTS In the PS model, LPC+ceftazidime reduced spleen and lung Pa238 concentrations (-3.45 and -3.56log10CFU/g; P<0.05) to a greater extent than ceftazidime monotherapy, while LPC+imipenem maintained the imipenem efficacy (-1.66 and -1.45log10CFU/g; P>0.05). In the pneumonia model, LPC+ceftazidime or LPC+imipenem reduced the lung Pa238 concentrations (-2.37log10CFU/g, P=0.1, or -1.35log10CFU/g, P=0.75). For Pa39, no statistically significant difference was observed in the PS and pneumonia models between combined therapy and monotherapy. Moreover, LPC+imipenem and LPC+ceftazidime significantly decreased and increased the TNF-α and IL-10 levels, respectively, in comparison with the untreated controls and monotherapies. CONCLUSIONS These results demonstrate the impact of immune response modification by LPC plus antibiotics on the prognosis of infections induced by ceftazidime-resistant P. aeruginosa.
Collapse
Affiliation(s)
- Raquel Parra-Millán
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| | - Manuel E Jiménez-Mejías
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain.
| | - Rafael Ayerbe-Algaba
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| | - Juan Domínguez-Herrera
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| | - Caridad Díaz
- Fundación Centro De Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Fundación MEDINA, Granada, Spain
| | - José Pérez Del Palacio
- Fundación Centro De Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Fundación MEDINA, Granada, Spain
| | - Jerónimo Pachón
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| | - Younes Smani
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| |
Collapse
|
11
|
Zhang Q, Zhang W, Liu J, Yang H, Hu Y, Zhang M, Bai T, Chang F. Lysophosphatidylcholine promotes intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 expression in human umbilical vein endothelial cells via an orphan G protein receptor 2-mediated signaling pathway. Bioengineered 2021; 12:4520-4535. [PMID: 34346841 PMCID: PMC8806654 DOI: 10.1080/21655979.2021.1956671] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The oxLDL-based bioactive lipid lysophosphatidylcholine (LPC) is a key regulator of physiological processes including endothelial cell adhesion marker expression. This study explored the relationship between LPC and the human umbilical vein endothelial cell expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) with a particular focus on the regulation of the LPC-G2A-ICAM-1/VCAM-1 pathway in this context. We explored the LPC-inducible role of orphan G protein receptor 2 (G2A) in associated regulatory processes by using human kidney epithelial (HEK293) cells that had been transfected with pET-G2A, human umbilical vein endothelial cells (HUVECs) in which an shRNA was used to knock down G2A, and western blotting and qPCR assays that were used to confirm changes in gene expression. For in vivo studies, a rabbit model of atherosclerosis was established, with serum biochemistry and histological staining approaches being used to assess pathological outcomes in these animals. The treatment of both HEK293 cells and HUVECs with LPC promoted ICAM-1 and VCAM-1 upregulation, while incubation at a pH of 6.8 suppressed such LPC-induced adhesion marker expression. Knocking down G2A by shRNA and inhibiting NF-κB activity yielded opposite outcomes. The application of a Gi protein inhibitor had no impact on LPC-induced ICAM-1/VCAM-1 expression. Atherosclerotic model exhibited high circulating LDL and LPC levels as well as high aortic wall ICAM-1/VCAM-1 expression. Overall, these results suggested that the LPC-G2A-ICAM-1/VCAM-1 pathway may contribute to the atherogenic activity of oxLDL, with NF-κB antagonists representing potentially viable therapeutic tools for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Qian Zhang
- The Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, China.,The Center for New Drug Screening Engineering and Research of Inner Mongolia Autonomous Region, Inner Mongolia Medical University, Hohhot, China.,College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Wei Zhang
- The Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, China.,The Center for New Drug Screening Engineering and Research of Inner Mongolia Autonomous Region, Inner Mongolia Medical University, Hohhot, China.,College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Jing Liu
- The Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, China.,The Center for New Drug Screening Engineering and Research of Inner Mongolia Autonomous Region, Inner Mongolia Medical University, Hohhot, China.,College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Haisen Yang
- First Clinical Medical College, Inner Mongolia Medical University, Hohhot, China
| | - Yuxia Hu
- The Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, China.,The Center for New Drug Screening Engineering and Research of Inner Mongolia Autonomous Region, Inner Mongolia Medical University, Hohhot, China.,College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Mengdi Zhang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Tuya Bai
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Fuhou Chang
- The Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, China.,The Center for New Drug Screening Engineering and Research of Inner Mongolia Autonomous Region, Inner Mongolia Medical University, Hohhot, China.,College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
12
|
Liu T, Wang X, Guo F, Sun X, Yuan K, Wang Q, Lan C. Lysophosphatidylcholine induces apoptosis and inflammatory damage in brain microvascular endothelial cells via GPR4-mediated NLRP3 inflammasome activation. Toxicol In Vitro 2021; 77:105227. [PMID: 34293432 DOI: 10.1016/j.tiv.2021.105227] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/25/2021] [Accepted: 07/15/2021] [Indexed: 01/14/2023]
Abstract
Lysophosphatidylcholine (LPC), as the main active component of oxidized low-density lipoproteins (ox-LDLs), has significant effects in cerebrovascular disease. However, the complex mechanism by which LPC functions in brain microvascular endothelial cells (BMECs) is not clearly understood. In this study, BMECs were transfected with G protein-coupled receptor 4 (GPR4) siRNA or an NLRP3-overexpression plasmid, and GPR4 expression was identified by RT-qPCR and western blotting; IL-1β, IL-18, and IL-33 levels were evaluated by ELISA. Apoptosis was monitored by flow cytometry and Hoechst staining, while Caspase 3, ASC, NLRP3, and GPR4 protein expression were examined by western blotting. Our results showed that LPC significantly increased the levels of inflammatory cytokines (IL-1β, IL-18, and IL-33) and markedly induced apoptosis and NLRP3 inflammasome activation in BMECs. Moreover, LPC notably upregulated GPR4 in BMECs, and knockdown of GPR4 significantly attenuated the effects of LPC in BMECs. Above all, we also proved that LPC induced apoptosis and inflammatory injury in BMECs by causing GPR4 to activate NLRP3 inflammasomes. Therefore, GPR4-mediated activation of NLRP3 inflammasomes might be the underlying mechanism by which LPC promotes the progression of cerebrovascular disease. In summary we found that LPC is an important pathogenic factor in cerebrovascular disease, and can induce GPR4 to active NLRP3 inflammasomes.
Collapse
Affiliation(s)
- Tao Liu
- Department of Neurology, University of Chinese Academy of Sciences Shenzhen Hospital (Guang ming), No. 39 Huaxia Road, Guangming District, Shenzhen 518107, China
| | - Xuegang Wang
- Department of Hepatology, The People's Hospital of Bao an, No. 118, Longjing Second Road, Baoan District, Shenzhen 518107, China
| | - Feng Guo
- Department of Neurology, University of Chinese Academy of Sciences Shenzhen Hospital (Guang ming), No. 39 Huaxia Road, Guangming District, Shenzhen 518107, China
| | - Xiaobo Sun
- Department of Laboratory Diagnostics, Changhai Hospital, No. 168 Changhai Road, Yangpu District, Shanghai 200433, China
| | - Kunxiong Yuan
- Department of Neurology, University of Chinese Academy of Sciences Shenzhen Hospital (Guang ming), No. 39 Huaxia Road, Guangming District, Shenzhen 518107, China
| | - Qingyong Wang
- Department of Neurology, University of Chinese Academy of Sciences Shenzhen Hospital (Guang ming), No. 39 Huaxia Road, Guangming District, Shenzhen 518107, China
| | - Chunwei Lan
- Department of Neurology, University of Chinese Academy of Sciences Shenzhen Hospital (Guang ming), No. 39 Huaxia Road, Guangming District, Shenzhen 518107, China.
| |
Collapse
|
13
|
Yu C, Zhang Q, Zou Y, Liu R, Zhao J, Bi K, Li D, Li Q. Across-polarity quantification method for broad metabolome coverage based on consecutive nanoconfined liquid phase nanoextraction technology: Application in discovering the plasma potential biomarkers of different types of cancer. Anal Chim Acta 2021; 1167:338577. [DOI: 10.1016/j.aca.2021.338577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/28/2022]
|
14
|
Parra-Millán R, Jiménez-Mejías ME, Ayerbe-Algaba R, Domínguez-Herrera J, Díaz C, Pérez Del Palacio J, Pachón J, Smani Y. Impact of the immune response modification by lysophosphatidylcholine in the efficacy of antibiotic therapy of experimental models of peritoneal sepsis and pneumonia by Pseudomonas aeruginosa: LPC therapeutic effect in combined therapy. Enferm Infecc Microbiol Clin 2020; 40:S0213-005X(20)30233-0. [PMID: 32674904 DOI: 10.1016/j.eimc.2020.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 11/03/2022]
Abstract
INTRODUCTION Immune response stimulation may be an adjuvant to antimicrobial treatment. Here, we evaluated the impact of immune response modification by lysophosphatidylcholine (LPC), combined with imipenem or ceftazidime, in murine models of peritoneal sepsis (PS) and pneumonia induced by Pseudomonas aeruginosa. METHODS The imipenem and ceftazidime-susceptible strain (Pa39) and imipenem and ceftazidime-resistant strain (Pa238) were used. Ceftazidime pharmacokinetic and pharmacodynamic parameters were determined. The therapeutic efficacy and TNF-α and IL-10 levels were determined in murine models of PS and pneumonia induced by Pa39 and Pa238 and treated with LPC, imipenem or ceftazidime, alone or in combination. RESULTS In the PS model, LPC+ceftazidime reduced spleen and lung Pa238 concentrations (-3.45 and -3.56log10CFU/g; P<0.05) to a greater extent than ceftazidime monotherapy, while LPC+imipenem maintained the imipenem efficacy (-1.66 and -1.45log10CFU/g; P>0.05). In the pneumonia model, LPC+ceftazidime or LPC+imipenem reduced the lung Pa238 concentrations (-2.37log10CFU/g, P=0.1, or -1.35log10CFU/g, P=0.75). For Pa39, no statistically significant difference was observed in the PS and pneumonia models between combined therapy and monotherapy. Moreover, LPC+imipenem and LPC+ceftazidime significantly decreased and increased the TNF-α and IL-10 levels, respectively, in comparison with the untreated controls and monotherapies. CONCLUSIONS These results demonstrate the impact of immune response modification by LPC plus antibiotics on the prognosis of infections induced by ceftazidime-resistant P. aeruginosa.
Collapse
Affiliation(s)
- Raquel Parra-Millán
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| | - Manuel E Jiménez-Mejías
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain.
| | - Rafael Ayerbe-Algaba
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| | - Juan Domínguez-Herrera
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| | - Caridad Díaz
- Fundación Centro De Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Fundación MEDINA, Granada, Spain
| | - José Pérez Del Palacio
- Fundación Centro De Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Fundación MEDINA, Granada, Spain
| | - Jerónimo Pachón
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| | - Younes Smani
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| |
Collapse
|
15
|
Knuplez E, Marsche G. An Updated Review of Pro- and Anti-Inflammatory Properties of Plasma Lysophosphatidylcholines in the Vascular System. Int J Mol Sci 2020; 21:E4501. [PMID: 32599910 PMCID: PMC7350010 DOI: 10.3390/ijms21124501] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
Lysophosphatidylcholines are a group of bioactive lipids heavily investigated in the context of inflammation and atherosclerosis development. While present in plasma during physiological conditions, their concentration can drastically increase in certain inflammatory states. Lysophosphatidylcholines are widely regarded as potent pro-inflammatory and deleterious mediators, but an increasing number of more recent studies show multiple beneficial properties under various pathological conditions. Many of the discrepancies in the published studies are due to the investigation of different species or mixtures of lysophatidylcholines and the use of supra-physiological concentrations in the absence of serum or other carrier proteins. Furthermore, interpretation of the results is complicated by the rapid metabolism of lysophosphatidylcholine (LPC) in cells and tissues to pro-inflammatory lysophosphatidic acid. Interestingly, most of the recent studies, in contrast to older studies, found lower LPC plasma levels associated with unfavorable disease outcomes. Being the most abundant lysophospholipid in plasma, it is of utmost importance to understand its physiological functions and shed light on the discordant literature connected to its research. LPCs should be recognized as important homeostatic mediators involved in all stages of vascular inflammation. In this review, we want to point out potential pro- and anti-inflammatory activities of lysophospholipids in the vascular system and highlight recent discoveries about the effect of lysophosphatidylcholines on immune cells at the endothelial vascular interface. We will also look at their potential clinical application as biomarkers.
Collapse
Affiliation(s)
- Eva Knuplez
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
16
|
Zhu C, Wang L, Zhu J, Jiang Y, Du X, Duan Q, Yin H, Huang X, Song Y, Cao B, Li G, An X. OGR1 negatively regulates β-casein and triglyceride synthesis and cell proliferation via the PI3K/AKT/mTOR signaling pathway in goat mammary epithelial cells. Anim Biotechnol 2020; 32:627-636. [PMID: 32167419 DOI: 10.1080/10495398.2020.1737099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Goat milk in some cases is less allergenic than cow milk, therefore, more people drink goat milk in the world, so it is necessary for us to improve the yield and quality of goat milk. Previous studies have shown that some genes are closely related to lactation. Ovarian cancer G protein-coupled 1 (OGR1) is a G protein-coupled receptor discovered recently. OGR1 is widely found in various tissues of organisms and is involved in cell skeleton reorganization, carcinogenesis, cell proliferation, and apoptosis by regulating multiple signaling pathways in cells. However, the modulating effect of OGR1 in lactation is still unknown. Therefore, the objective of this study is to investigate the function of OGR1 in goat mammary epithelial cells (GMECs). Flow cytometry, CCK8, EDU, enzyme-linked immunosorbent assay, and triglyceride test kit assays were performed and we found that OGR1 regulated Bcl-2/Bax ratio, Fas protein expression as well as the phosphorylation of AKT and mammalian target of rapamycin (mTOR). si-OGR1 could enhance the proliferation of GMECs by promoting G1/S phase progression and the synthesis of β-casein and triglyceride. By contrast, OGR1 repressed GMECs proliferation and down-regulated the synthesis of β-casein and triglyceride by blocking the PI3K/AKT/mTOR signaling pathway in GMECs.
Collapse
Affiliation(s)
- Chao Zhu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Lili Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Junru Zhu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Yue Jiang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Xiaoyan Du
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Quyu Duan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Hao Yin
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Xinrui Huang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Binyun Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Guang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Xiaopeng An
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| |
Collapse
|
17
|
Krewson EA, Sanderlin EJ, Marie MA, Akhtar SN, Velcicky J, Loetscher P, Yang LV. The Proton-Sensing GPR4 Receptor Regulates Paracellular Gap Formation and Permeability of Vascular Endothelial Cells. iScience 2020; 23:100848. [PMID: 32058960 PMCID: PMC6997876 DOI: 10.1016/j.isci.2020.100848] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 11/27/2019] [Accepted: 01/13/2020] [Indexed: 01/31/2023] Open
Abstract
GPR4 is a pH-sensing G protein-coupled receptor highly expressed in vascular endothelial cells and can be activated by protons in the inflamed tissue microenvironment. Herein, we report that acidosis-induced GPR4 activation increases paracellular gap formation and permeability of vascular endothelial cells through the Gα12/13/Rho GTPase signaling pathway. Evaluation of GPR4 in the inflammatory response using the acute hindlimb ischemia-reperfusion mouse model revealed that GPR4 mediates tissue edema, inflammatory exudate formation, endothelial adhesion molecule expression, and leukocyte infiltration in the inflamed tissue. Genetic knockout and pharmacologic inhibition of GPR4 alleviate tissue inflammation. These results suggest GPR4 is a pro-inflammatory receptor and could be targeted for therapeutic intervention. Acidosis/GPR4 regulates endothelial paracellular gap formation and permeability GPR4 exacerbates inflammation by increasing tissue edema and leukocyte infiltration Pharmacological inhibition of GPR4 reduces inflammatory responses
Collapse
Affiliation(s)
- Elizabeth A Krewson
- Department of Anatomy and Cell Biology, East Carolina University, Greenville, NC 27834, USA
| | - Edward J Sanderlin
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Mona A Marie
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Shayan Nik Akhtar
- Department of Anatomy and Cell Biology, East Carolina University, Greenville, NC 27834, USA
| | - Juraj Velcicky
- Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Pius Loetscher
- Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Li V Yang
- Department of Anatomy and Cell Biology, East Carolina University, Greenville, NC 27834, USA; Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
18
|
Zhang Q, Xu H, Liu R, Gao P, Yang X, Jin W, Zhang Y, Bi K, Li Q. A Novel Strategy for Targeted Lipidomics Based on LC-Tandem-MS Parameters Prediction, Quantification, and Multiple Statistical Data Mining: Evaluation of Lysophosphatidylcholines as Potential Cancer Biomarkers. Anal Chem 2019; 91:3389-3396. [DOI: 10.1021/acs.analchem.8b04715] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Qian Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Huarong Xu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Ran Liu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Peng Gao
- Metabolomics Core Facility of RHLCCC, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Xiao Yang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Wei Jin
- Urumqi Traditional Chinese Medicine Hospital, 590 Youhao South Road, Urumqi 830000, China
| | - Yiwen Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Kaishun Bi
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Qing Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| |
Collapse
|
19
|
Bao L, Qi J, Wang YW, Xi Q, Tserennadmid T, Zhao PF, Qi J, Damirin A. The atherogenic actions of LPC on vascular smooth muscle cells and its LPA receptor mediated mechanism. Biochem Biophys Res Commun 2018; 503:1911-1918. [DOI: 10.1016/j.bbrc.2018.07.135] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 07/25/2018] [Indexed: 02/02/2023]
|
20
|
Southan C, Sharman JL, Faccenda E, Pawson AJ, Harding SD, Davies JA. Challenges of Connecting Chemistry to Pharmacology: Perspectives from Curating the IUPHAR/BPS Guide to PHARMACOLOGY. ACS OMEGA 2018; 3:8408-8420. [PMID: 30087946 PMCID: PMC6070956 DOI: 10.1021/acsomega.8b00884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/12/2018] [Indexed: 06/08/2023]
Abstract
Connecting chemistry to pharmacology has been an objective of Guide to PHARMACOLOGY (GtoPdb) and its precursor the International Union of Basic and Clinical Pharmacology Database (IUPHAR-DB) since 2003. This has been achieved by populating our database with expert-curated relationships between documents, assays, quantitative results, chemical structures, their locations within the documents, and the protein targets in the assays (D-A-R-C-P). A wide range of challenges associated with this are described in this perspective, using illustrative examples from GtoPdb entries. Our selection process begins with judgments of pharmacological relevance and scientific quality. Even though we have a stringent focus for our small-data extraction, we note that assessing the quality of papers has become more difficult over the last 15 years. We discuss ambiguity issues with the resolution of authors' descriptions of A-R-C-P entities to standardized identifiers. We also describe developments that have made this somewhat easier over the same period both in the publication ecosystem and recent enhancements of our internal processes. This perspective concludes with a look at challenges for the future, including the wider capture of mechanistic nuances and possible impacts of text mining on automated entity extraction.
Collapse
|
21
|
Weiß KT, Fante M, Köhl G, Schreml J, Haubner F, Kreutz M, Haverkampf S, Berneburg M, Schreml S. Proton-sensing G protein-coupled receptors as regulators of cell proliferation and migration during tumor growth and wound healing. Exp Dermatol 2018; 26:127-132. [PMID: 27623507 DOI: 10.1111/exd.13209] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2016] [Indexed: 12/20/2022]
Abstract
Dysregulation of pH is a feature of both tumor growth and tissue repair. In tumors, microenvironmental changes, like in lactate metabolism, lead to altered intra- and extracellular pH (pHi , pHe ) and vice versa. In wounds, barrier disruption results in extensive variations in pHe on the wound surface. It is known that altered extracellular proton concentrations have a major impact on cell turnover and migration as well as on the metabolic activity of cells involved in tumor spread and wound closure. The proton-sensing G protein-coupled receptors (GPCRs) GPR4, GPR65 (TDAG8), GPR68 (OGR1) and GPR132 (G2A) are activated via a decrease in pHe and transduce this signal to molecular intracellular pathways. Based on the current knowledge, we speculate on the role of proton-sensing GPCRs in wound healing and on their potential as mechanistic linkers of tumor growth and tissue repair.
Collapse
Affiliation(s)
- Katharina T Weiß
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Matthias Fante
- Department of Internal Medicine III, Oncology, University Medical Center Regensburg, Regensburg, Germany
| | - Gudrun Köhl
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Julia Schreml
- Institute of Human Genetics, University of Cologne, Cologne, Germany
| | - Frank Haubner
- Department of Otorhinolaryngology, University Medical Center Regensburg, Regensburg, Germany
| | - Marina Kreutz
- Department of Internal Medicine III, Oncology, University Medical Center Regensburg, Regensburg, Germany
| | - Sonja Haverkampf
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Stephan Schreml
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| |
Collapse
|
22
|
Park S, Lee S, Nam S, Im D. GPR35 mediates lodoxamide-induced migration inhibitory response but not CXCL17-induced migration stimulatory response in THP-1 cells; is GPR35 a receptor for CXCL17? Br J Pharmacol 2018; 175:154-161. [PMID: 29068046 PMCID: PMC5740256 DOI: 10.1111/bph.14082] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 09/29/2017] [Accepted: 10/07/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE GPR35 has long been considered an orphan GPCR, because no endogenous ligand of GPR35 has been discovered. CXCL17 (a chemokine) has been reported to be an endogenous ligand of GPR35, and it has even been suggested that it be called CXCR8. However, at present there is no supporting evidence that CXCL17 does interact with GPR35. EXPERIMENTAL APPROACH We applied two assay systems to explore the relationship between CXCL17 and GPR35. An AP-TGF-α shedding assay in GPR35 over-expressing HEK293 cells was used as a gain-of-function assay. GPR35 knock-down by siRNA transfection was performed in endogenously GPR35-expressing THP-1 cells. KEY RESULTS In the AP-TGF-α shedding assay, lodoxamide, a well-known synthetic GPR35 agonist, was confirmed to be the most potent agonist among other reported agonists. However, neither human nor mouse CXCL17 had an effect on GPR35. Consistent with previous findings, G proteins Gαi/o and Gα12/13 were found to couple with GPR35. Furthermore, lodoxamide-induced activation of GPR35 was concentration-dependently inhibited by CID2745687 (a selective GPR35 antagonist). In endogenously GPR35-expressing THP-1 cells, lodoxamide concentration-dependently inhibited migration and this inhibitory effect was blocked by CID2745687 treatment or GPR35 siRNA transfection. However, even though CXCL17 stimulated the migration of THP-1 cells, which is consistent with a previous report, this stimulatory effect of CXCL17 was not blocked by CID2745687 or GPR35 siRNA. CONCLUSIONS AND IMPLICATIONS The present findings suggest that GPR35 functions as a migration inhibitory receptor, but CXCL17-stimulated migration of THP-1 cells is not dependent on GPR35.
Collapse
Affiliation(s)
- Soo‐Jin Park
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of PharmacyPusan National UniversityBusanKorea
| | - Seung‐Jin Lee
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of PharmacyPusan National UniversityBusanKorea
| | - So‐Yeon Nam
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of PharmacyPusan National UniversityBusanKorea
| | - Dong‐Soon Im
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of PharmacyPusan National UniversityBusanKorea
| |
Collapse
|
23
|
Kang JH, Kim HJ, Park MK, Lee CH. Sphingosylphosphorylcholine Induces Thrombospondin-1 Secretion in MCF10A Cells via ERK2. Biomol Ther (Seoul) 2017; 25:625-633. [PMID: 28274095 PMCID: PMC5685432 DOI: 10.4062/biomolther.2016.228] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/11/2016] [Accepted: 01/09/2017] [Indexed: 01/06/2023] Open
Abstract
Sphingosylphosphorylcholine (SPC) is one of the bioactive phospholipids that has many cellular functions such as cell migration, adhesion, proliferation, angiogenesis, and Ca²⁺ signaling. Recent studies have reported that SPC induces invasion of breast cancer cells via matrix metalloproteinase-3 (MMP-3) secretion leading to WNT activation. Thrombospondin-1 (TSP-1) is a matricellular and calcium-binding protein that binds to a wide variety of integrin and non-integrin cell surface receptors. It regulates cell proliferation, migration, and apoptosis in inflammation, angiogenesis and neoplasia. TSP-1 promotes aggressive phenotype via epithelial mesenchymal transition (EMT). The relationship between SPC and TSP-1 is unclear. We found SPC induced EMT leading to mesenchymal morphology, decrease of E-cadherin expression and increases of N-cadherin and vimentin. SPC induced secretion of thrombospondin-1 (TSP-1) during SPC-induced EMT of various breast cancer cells. Gene silencing of TSP-1 suppressed SPC-induced EMT as well as migration and invasion of MCF10A cells. An extracellular signal-regulated kinase inhibitor, PD98059, significantly suppressed the secretion of TSP-1, expressions of N-cadherin and vimentin, and decrease of E-cadherin in MCF10A cells. ERK2 siRNA suppressed TSP-1 secretion and EMT. From online PROGgene V2, relapse free survival is low in patients having high TSP-1 expressed breast cancer. Taken together, we found that SPC induced EMT and TSP-1 secretion via ERK2 signaling pathway. These results suggests that SPC-induced TSP-1 might be a new target for suppression of metastasis of breast cancer cells.
Collapse
Affiliation(s)
- June Hee Kang
- College of Pharmacy, Dongguk University, Seoul 10326, Republic of Korea
| | - Hyun Ji Kim
- College of Pharmacy, Dongguk University, Seoul 10326, Republic of Korea
| | - Mi Kyung Park
- College of Pharmacy, Dongguk University, Seoul 10326, Republic of Korea.,National Cancer Center, Goyang, 10408, Republic of Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 10326, Republic of Korea
| |
Collapse
|
24
|
Rajkumar P, Pluznick JL. Unsung renal receptors: orphan G-protein-coupled receptors play essential roles in renal development and homeostasis. Acta Physiol (Oxf) 2017; 220:189-200. [PMID: 27699982 DOI: 10.1111/apha.12813] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/23/2016] [Accepted: 09/29/2016] [Indexed: 12/31/2022]
Abstract
Recent studies have shown that orphan GPCRs of the GPR family are utilized as specialized chemosensors in various tissues to detect metabolites, and in turn to activate downstream pathways which regulate systemic homeostasis. These studies often find that such metabolites are generated by well-known metabolic pathways, implying that known metabolites and chemicals may perform novel functions. In this review, we summarize recent findings highlighting the role of deorphanized GPRs in renal development and function. Understanding the role of these receptors is critical in gaining insights into mechanisms that regulate renal function both in health and in disease.
Collapse
Affiliation(s)
- P. Rajkumar
- Department of Physiology; Johns Hopkins School of Medicine; Baltimore; MD USA
| | - J. L. Pluznick
- Department of Physiology; Johns Hopkins School of Medicine; Baltimore; MD USA
| |
Collapse
|
25
|
Ross T, Jakubzig B, Grundmann M, Massing U, Kostenis E, Schlesinger M, Bendas G. The molecular mechanism by which saturated lysophosphatidylcholine attenuates the metastatic capacity of melanoma cells. FEBS Open Bio 2016; 6:1297-1309. [PMID: 28255537 PMCID: PMC5324772 DOI: 10.1002/2211-5463.12152] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/13/2016] [Accepted: 10/25/2016] [Indexed: 12/11/2022] Open
Abstract
Lysophophatidylcholine (LysoPC) is an abundant constituent in human plasma. Patients with malignant cancer diseases have attenuated LysoPC plasma levels, and thus LysoPC has been examined as a metabolic biomarker for cancer prediction. Preclinical studies have shown that solid tumor cells drastically degrade LysoPCs by incorporating their free fatty acids into cell membrane phospholipids. In this way, LysoPC C18:0 reduced the metastatic spread of murine melanoma B16.F10 cells in mice. Although membrane rigidification may have a key role in the attenuation of metastasis, evidence for this has yet to be shown. Therefore, the present study aimed to determine how LysoPC reduces the metastatic capacity of B16.F10 cells. Following cellular preincubation with LysoPC C18:0 at increasing concentrations and lengths of time, cell migration was most significantly attenuated with 450 μm LysoPC C18:0 at 72 h. Biosensor measurements suggest that, despite their abundance in B16.F10 cells, LysoPC‐sensitive G protein‐coupled receptors do not appear to contribute to this effect. Instead, the attenuated migration appears to result from changes in cell membrane properties and their effect on underlying signaling pathways, most likely the formation of focal adhesion complexes. Treatment with 450 μm LysoPC C18:0 activates protein kinase C (PKC)δ to phosphorylate syndecan‐4, accompanied by deactivation of PKCα. Subsequently, focal adhesion complex formation was attenuated, as confirmed by the reduced activity of focal adhesion kinase (FAK). Interestingly, 450 μm LysoPC C18:1 did not affect FAK activity, explaining its lower propensity to affect migration and metastasis. Therefore, membrane rigidification by LysoPC C18:0 appears to prevent the formation of focal adhesion complexes, thus affecting integrin activity as a key for metastatic melanoma spread.
Collapse
Affiliation(s)
- Thomas Ross
- Department of Pharmaceutical Chemistry II University of Bonn Germany
| | - Bastian Jakubzig
- Department of Pharmaceutical Chemistry II University of Bonn Germany
| | | | - Ulrich Massing
- Andreas Hettich GmbH & Co. KGF&E Lifescience Applications Freiburg Germany; Faculty of Chemistry & Pharmacy University of Freiburg Germany
| | - Evi Kostenis
- Department of Pharmaceutical Biology University of Bonn Germany
| | | | - Gerd Bendas
- Department of Pharmaceutical Chemistry II University of Bonn Germany
| |
Collapse
|
26
|
Ren J, Zhang Y, Cai H, Ma H, Zhao D, Zhang X, Li Z, Wang S, Wang J, Liu R, Li Y, Qian J, Wei H, Niu L, Liu Y, Xiao L, Ding M, Jiang S. Human GPR4 and the Notch signaling pathway in endothelial cell tube formation. Mol Med Rep 2016; 14:1235-40. [PMID: 27279286 DOI: 10.3892/mmr.2016.5380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 02/29/2016] [Indexed: 11/06/2022] Open
Abstract
G protein-coupled receptor 4 (GPR4) is hypothesized to function as a pH sensor and is important in the regulation of proliferation, migration and angiogenesis of vascular endothelial cells (ECs). Furthermore, the Notch signaling pathway is significant in the regulation of the angiogenic behavior of ECs. However, whether GPR4 regulates angiogenesis via the Notch signaling pathway remains unclear. The present study evaluated the effect of Notch signaling in human GPR4‑induced angiogenesis in HMEC‑1 cells. The results revealed that GPR4 increased Notch1 expression in a time‑dependent manner. In addition, the inhibition of Notch1 expression using small interfering RNA or the Notch receptor inhibitor, γ-secretase inhibitor I, significantly blocked GPR4‑induced HMEC‑1 tube formation and lymphocyte transendothelial migration. Furthermore, the inhibition of Notch1 blocked GPR4‑induced vascular endothelial growth factor and hypoxia-inducible factor 1α expression. Thus, it was demonstrated that GPR4 affects ECs by regulating Notch1, a function that may be important for physiological and pathological angiogenesis.
Collapse
Affiliation(s)
- Juan Ren
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yuelang Zhang
- Imaging Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Hui Cai
- Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Hongbing Ma
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Dongli Zhao
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xiaozhi Zhang
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Zongfang Li
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Shufeng Wang
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jiangsheng Wang
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Rui Liu
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yi Li
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jiansheng Qian
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Hongxia Wei
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Liying Niu
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yan Liu
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Lisha Xiao
- Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Muyang Ding
- Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Shiwen Jiang
- Department of Biomedical Science, Mercer University School of Medicine, Macon, GA 31207, USA
| |
Collapse
|
27
|
Kim HJ, Kang GJ, Kim EJ, Park MK, Byun HJ, Nam S, Lee H, Lee CH. Novel effects of sphingosylphosphorylcholine on invasion of breast cancer: Involvement of matrix metalloproteinase-3 secretion leading to WNT activation. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1533-43. [PMID: 27216977 DOI: 10.1016/j.bbadis.2016.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/27/2016] [Accepted: 05/18/2016] [Indexed: 01/17/2023]
Abstract
Sphingosylphosphorylcholine (SPC) participates in several cellular processes including metastasis. SPC induces keratin reorganization and regulates the viscoelasticity of metastatic cancer cells including PANC-1 cancer cells leading to enhanced migration and invasion. The role of SPC and the relevant mechanism in invasion of breast cell are as yet unknown. SPC dose-dependently induces invasion of breast cancer cells or breast immortalized cells. Reverse transcription polymerase chain reaction and Western blot analyses of MCF10A and ZR-75-1 cells indicated that SPC induces expression and secretion of matrix metalloproteinase-3 (MMP3). From online KMPLOT, relapse free survival is high in patients having low MMP3 expressed basal breast cancer (n=581, p=0.032). UK370106 (MMP3 inhibitor) or gene silencing of MMP3 markedly inhibited the SPC-induced invasion of MCF10A cells. An extracellular signal-regulated kinase (ERK) inhibitor, PD98059, significantly suppressed the secretion and the gelatinolytic activity of MMP3, and invasion in MCF10A cells. Over-expression of ERK1 and ERK2 promoted both the expression and secretion of MMP3. In contrast, gene silencing of ERK1 and ERK2 attenuated the secretion of MMP3 in MCF10A cells. The effects of SPC-induced MMP3 secretion on β-catenin and TCF/lymphoid enhancer factor (LEF) promoter activity were examined since MMP3 indirectly activates canonical Wnt signaling. SPC induced translocation of β-catenin to nucleus and increased TCF/LEF promoter activity. These events were suppressed by UK370106 or PD98059. Wnt inhibitor, FH535 inhibited SPC-induced MMP3 secretion and invasion. Taken together, these results suggest that SPC induces MMP3 expression and secretion via ERK leading to Wnt activation.
Collapse
Affiliation(s)
- Hyun Ji Kim
- BK21PLUS R-FIND team, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Gyeoung Jin Kang
- BK21PLUS R-FIND team, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Eun Ji Kim
- BK21PLUS R-FIND team, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Mi Kyung Park
- BK21PLUS R-FIND team, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Hyun Jung Byun
- BK21PLUS R-FIND team, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Seungyoon Nam
- Department of Life Sciences, College of BioNano Technology, Gachon University, Sungnam 13120, Republic of Korea
| | - Ho Lee
- National Cancer Center, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Chang Hoon Lee
- BK21PLUS R-FIND team, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea.
| |
Collapse
|
28
|
Huang YH, Su YS, Chang CJ, Sun WH. Heteromerization of G2A and OGR1 enhances proton sensitivity and proton-induced calcium signals. J Recept Signal Transduct Res 2016; 36:633-644. [PMID: 27049592 DOI: 10.3109/10799893.2016.1155064] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Proton-sensing G-protein-coupled receptors (GPCRs; OGR1, GPR4, G2A, TDAG8), with full activation at pH 6.4 ∼ 6.8, are important to pH homeostasis, immune responses and acid-induced pain. Although G2A mediates the G13-Rho pathway in response to acid, whether G2A activates Gs, Gi or Gq proteins remains debated. In this study, we examined the response of this fluorescence protein-tagged OGR1 family to acid stimulation in HEK293T cells. G2A did not generate detectable intracellular calcium or cAMP signals or show apparent receptor redistribution with moderate acid (pH ≥ 6.0) stimulation but reduced cAMP accumulation under strong acid stimulation (pH ≤ 5.5). Surprisingly, coexpression of OGR1- and G2A-enhanced proton sensitivity and proton-induced calcium signals. This alteration is attributed to oligomerization of OGR1 and G2A. The oligomeric potential locates receptors at a specific site, which leads to enhanced proton-induced calcium signals through channels.
Collapse
Affiliation(s)
- Ya-Han Huang
- a Department of Life Sciences , National Central University , Jhongli , Taiwan and
| | - Yeu-Shiuan Su
- a Department of Life Sciences , National Central University , Jhongli , Taiwan and
| | - Chung-Jen Chang
- a Department of Life Sciences , National Central University , Jhongli , Taiwan and
| | - Wei-Hsin Sun
- a Department of Life Sciences , National Central University , Jhongli , Taiwan and.,b Center for Biotechnology and Biomedical Engineering, National Central University , Jhongli , Taiwan
| |
Collapse
|
29
|
Li L, Matsuoka I, Sakamoto K, Kimura J. Differential effects of lysophosphatidylcholine and ACh on muscarinic K(+),non-selective cation and Ca(2+) currents in guinea-pig atrial cells. Fukushima J Med Sci 2016; 62:27-35. [PMID: 26911304 DOI: 10.5387/fms.2015-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We compared the effects of lysophosphatidylcholine (LPC) and acetylcholine (ACh) on IK(ACh), ICa and a non-selective cation current (INSC) in guinea-pig atrial myocytes to clarify whether LPC and ACh activate similar Gi/o-coupled effector systems. IK(ACh), ICa and INSC were analyzed in single atrial myocytes by the whole cell patch-clamp. LPC induced INSC in a concentration-dependent manner in atrial cells. ACh activated IK(ACh), but failed to evoke INSC. LPC also activated IK(ACh) but with significantly less potency than ACh. The effects of both ligands on IK(ACh) were inhibited by intracellular loading of pre-activated PTX. This treatment also inhibited LPC-induced INSC, indicating that IK(ACh) and INSC induced by LPC are both mediated by Gi/o. LPC and ACh had similar potencies in inhibiting ICa, which was pre-augmented by forskolin, indicating that LPC and ACh activate similar amounts of α-subunits of Gi/o. The different effects of LPC and ACh on IK(ACh) and INSC may suggest that LPC and ACh activate Gi/o having different types of βγ subunits, and that LPC-induced INSC may be mediated by βγ subunits of Gi/o, which are less effective in inducing IK(ACh).
Collapse
Affiliation(s)
- Libing Li
- Department of Pharmacology, School of Medicine, Fukushima Medical University
| | | | | | | |
Collapse
|
30
|
Novel effects of FTY720 on perinuclear reorganization of keratin network induced by sphingosylphosphorylcholine: Involvement of protein phosphatase 2A and G-protein-coupled receptor-12. Eur J Pharmacol 2016; 775:86-95. [PMID: 26872988 DOI: 10.1016/j.ejphar.2016.02.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 01/31/2016] [Accepted: 02/08/2016] [Indexed: 01/09/2023]
Abstract
Sphingosylphosphorylcholine (SPC) evokes perinuclear reorganization of keratin 8 (K8) filaments and regulates the viscoelasticity of metastatic cancer cells leading to enhanced migration. Few studies have addressed the compounds modulating the viscoelasticity of metastatic cancer cells. We studied the effects of sphingosine (SPH), sphingosine 1-phosphate (S1P), FTY720 and FTY720-phosphate (FTY720P) on SPC-induced K8 phosphorylation and reorganization using Western blot and confocal microscopy, and also evaluated the elasticity of PANC-1 cells by atomic force microscopy. FTY720, FTY720P, SPH, and S1P concentration-dependently inhibited SPC-evoked phosphorylation and reorganization of K8, and migration of PANC-1 cells. SPC triggered reduction and narrow distribution of elastic constant K and conversely, FTY720 blocked them. A common upstream regulator of JNK and ERK, protein phosphatase 2A (PP2A) expression was reduced by SPC, but was restored by FTY720 and FTY72P. Butyryl forskolin, a PP2A activator, suppressed SPC-induced K8 phosphorylation and okadaic acid, a PP2A inhibitor, induced K8 phosphorylation. Gene silencing of PP2A also led to K8 phosphorylation, reorganization and migration. We also investigated the involvement of GPR12, a high-affinity SPC receptor, in SPC-evoked keratin phosphorylation and reorganization. GPR12 siRNA suppressed the SPC-triggered phosphorylation and reorganization of K8. GPR12 overexpression stimulated keratin phosphorylation and reorganization even without SPC. FTY720 and FTY720P suppressed the GPR12-induced phosphorylation and reorganization of K8. The collective data indicates that FTY720 and FTY720P suppress SPC-induced phosphorylation and reorganization of K8 in PANC-1 cells by restoring the expression of PP2A via GPR12. These findings might be helpful in the development of compounds that modulate the viscoelasticity of metastatic cancer cells and various SPC actions.
Collapse
|
31
|
Molecular mechanisms of target recognition by lipid GPCRs: relevance for cancer. Oncogene 2015; 35:4021-35. [PMID: 26640151 DOI: 10.1038/onc.2015.467] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/02/2015] [Accepted: 11/02/2015] [Indexed: 12/18/2022]
|
32
|
Gazos-Lopes F, Oliveira MM, Hoelz LVB, Vieira DP, Marques AF, Nakayasu ES, Gomes MT, Salloum NG, Pascutti PG, Souto-Padrón T, Monteiro RQ, Lopes AH, Almeida IC. Structural and functional analysis of a platelet-activating lysophosphatidylcholine of Trypanosoma cruzi. PLoS Negl Trop Dis 2014; 8:e3077. [PMID: 25101628 PMCID: PMC4125143 DOI: 10.1371/journal.pntd.0003077] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 06/13/2014] [Indexed: 12/15/2022] Open
Abstract
Background Trypanosoma cruzi is the causative agent of the life-threatening Chagas disease, in which increased platelet aggregation related to myocarditis is observed. Platelet-activating factor (PAF) is a potent intercellular lipid mediator and second messenger that exerts its activity through a PAF-specific receptor (PAFR). Previous data from our group suggested that T. cruzi synthesizes a phospholipid with PAF-like activity. The structure of T. cruzi PAF-like molecule, however, remains elusive. Methodology/Principal findings Here, we have purified and structurally characterized the putative T. cruzi PAF-like molecule by electrospray ionization-tandem mass spectrometry (ESI-MS/MS). Our ESI-MS/MS data demonstrated that the T. cruzi PAF-like molecule is actually a lysophosphatidylcholine (LPC), namely sn-1 C18:1(delta 9)-LPC. Similar to PAF, the platelet-aggregating activity of C18:1-LPC was abrogated by the PAFR antagonist, WEB 2086. Other major LPC species, i.e., C16:0-, C18:0-, and C18:2-LPC, were also characterized in all T. cruzi stages. These LPC species, however, failed to induce platelet aggregation. Quantification of T. cruzi LPC species by ESI-MS revealed that intracellular amastigote and trypomastigote forms have much higher levels of C18:1-LPC than epimastigote and metacyclic trypomastigote forms. C18:1-LPC was also found to be secreted by the parasite in extracellular vesicles (EV) and an EV-free fraction. A three-dimensional model of PAFR was constructed and a molecular docking study was performed to predict the interactions between the PAFR model and PAF, and each LPC species. Molecular docking data suggested that, contrary to other LPC species analyzed, C18:1-LPC is predicted to interact with the PAFR model in a fashion similar to PAF. Conclusions/Significance Taken together, our data indicate that T. cruzi synthesizes a bioactive C18:1-LPC, which aggregates platelets via PAFR. We propose that C18:1-LPC might be an important lipid mediator in the progression of Chagas disease and its biosynthesis could eventually be exploited as a potential target for new therapeutic interventions. Chagas disease, caused by the parasite Trypanosoma cruzi, was exclusively confined to Latin America but it has recently spread to other regions of the world. Chagas disease affects 8–10 million people and kills thousands of them every year. Lysophosphatidylcholine (LPC) is a major bioactive phospholipid of human plasma low-density lipoproteins (LDL). Platelet-activating factor (PAF) is a phospholipid similar to LPC and a potent intercellular mediator. Both PAF and LPC have been reported to act on mammalian cells through PAF receptor (PAFR). Previous data from our group suggested that T. cruzi produces a phospholipid with PAF activity. Here, we describe the structural and functional analysis of different species of LPC from T. cruzi, including a LPC with a fatty acid chain of 18 carbon atoms and one double bond (C18:1-LPC). We also show that C18:1-LPC is able to induce rabbit platelet aggregation, which is abrogated by a PAFR antagonist. In addition, a three-dimensional model of human PAFR was constructed. Contrary to other T. cruzi LPC molecules, C18:1-LPC is predicted to interact with the PAFR model in a fashion similar to PAF. Further studies are needed to validate the biosynthesis of T. cruzi C18:1-LPC as a potential drug target in Chagas disease.
Collapse
Affiliation(s)
- Felipe Gazos-Lopes
- The Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, Texas, United States of America
| | - Mauricio M. Oliveira
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco I, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucas V. B. Hoelz
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco G, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle P. Vieira
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco I, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alexandre F. Marques
- The Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, Texas, United States of America
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Ernesto S. Nakayasu
- The Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, Texas, United States of America
| | - Marta T. Gomes
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco I, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco H, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nasim G. Salloum
- The Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, Texas, United States of America
| | - Pedro G. Pascutti
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco G, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thaïs Souto-Padrón
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco I, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Q. Monteiro
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco H, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Angela H. Lopes
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco I, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail: (AHL); (ICA)
| | - Igor C. Almeida
- The Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, Texas, United States of America
- * E-mail: (AHL); (ICA)
| |
Collapse
|
33
|
Liu P, Duan JA, Guo JM, Qian DW, Shang EX, Tang YP, Su SL. Plasma metabolic profiling of normal and dysmenorrhea syndrome rats and the effects of Xiang-Fu-Si-Wu Decoction intervention. PHARMACEUTICAL BIOLOGY 2014; 52:603-613. [PMID: 24262062 DOI: 10.3109/13880209.2013.858269] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 10/17/2013] [Indexed: 06/02/2023]
Abstract
CONTEXT Primary dysmenorrhea (PDM), a common, clinically heterogeneous endocrine disorder affecting young women, is associated with endocrinopathy and metabolic abnormalities. The Xiang-Fu-Si-Wu Decoction (XFSWD) is a traditional Chinese medicine preparation used to treat PDM. OBJECTIVE In the current study, a plasma metabonomics method based on the ultra-high-performance liquid chromatography-quantitative time-of-flight-mass spectrometry (UHPLC-Q-TOF-MS) system was employed to examine the mechanism of XFSWD action in PDM. MATERIALS AND METHODS Estradiol benzoate (0.01 g/kg/d) and oxytocin (5 mL/kg) were used to create the dysmenorrhea rat model. Based on the chromatographic data of plasma samples at different time-points following oral administration of XFSWD mixed in water (37.8 g crude herbs/kg) on day 7, partial least square (PLS) and discriminate analysis (DA) were applied to visualize group differentiation and marker selection. RESULTS Systemic changes occurring in PDM reflect alterations in not only uterus function but also whole-body metabolism. The XFSWD was effective as a therapeutic agent for PDM by reflect metabolic pathway. Prostaglandins and lysophospholipids were identified as two marker types for oxytocin-induced dysmenorrhea syndrome, including LysoPC(18:4), LysoPE(22:2/0:0), LysoPC(17:0), PGJ₂, 11-deoxy-11-methylene-PGD₂, 15-deoxy-δ-12,14-PGJ₂, LysoPC(20:3), etc. Specifically, the concentrations of prostaglandins compounds (PGJ₂, 11-deoxy-11-methylene-PGD₂, 15-deoxy-δ-12,14-PGJ₂) were increased while those of lysophospholipid compounds [lysoPC(18:4), LysoPE(22:2/0:0), LysoPC(17:0)] were decreased to a significant extent (p < 0.05) in dysmenorrheal rats. Upon treatment with the XFSWD at 12 h, the concentrations of lysophospholipids showed no significant differences (P > 0.05) between the model and normal groups. The lysophospholipid levels were restored. Lysophospholipids were the key factors in phospholipid metabolism. Thus, disruption of phospholipids metabolism appears critical for the development of dysmenorrhea. The XFSWD exerted its effects by interfering with the sphingolipid metabolic pathway. DISCUSSION AND CONCLUSIONS The metabonomics method presents a promising tool to treat PDM in animal models, and may be applicable for clinical treatment of the human disease in the future.
Collapse
Affiliation(s)
- Pei Liu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine , Nanjing, PR China
| | | | | | | | | | | | | |
Collapse
|
34
|
Su YS, Sun WH, Chen CC. Molecular mechanism of inflammatory pain. World J Anesthesiol 2014; 3:71-81. [DOI: 10.5313/wja.v3.i1.71] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 09/20/2013] [Accepted: 11/03/2013] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammatory pain resulting from arthritis, nerve injury and tumor growth is a serious public health issue. One of the major challenges in chronic inflammatory pain research is to develop new pharmacologic treatments with long-term efficacy and few side effects. The mediators released from inflamed sites induce complex changes in peripheral and central processing by directly acting on transducer receptors located on primary sensory neurons to transmit pain signals or indirectly modulating pain signals by activating receptors coupled with G-proteins and second messengers. High local proton concentration (acidosis) is thought to be a decisive factor in inflammatory pain and other mediators such as prostaglandin, bradykinin, and serotonin enhance proton-induced pain. Proton-sensing ion channels [transient receptor potential V1 (TRPV1) and the acid-sensing ion channel (ASIC) family] are major receptors for direct excitation of nociceptive sensory neurons in response to acidosis or inflammation. G-protein-coupled receptors activated by prostaglandin, bradykinin, serotonin, and proton modulate functions of TRPV1, ASICs or other ion channels, thus leading to inflammation- or acidosis-linked hyperalgesia. Although detailed mechanisms remain unsolved, clearly different types of pain or hyperalgesia could be due to complex interactions between a distinct subset of inflammatory mediator receptors expressed in a subset of nociceptors. This review describes new directions for the development of novel therapeutic treatments in pain.
Collapse
|
35
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol 2013; 170:1459-581. [PMID: 24517644 PMCID: PMC3892287 DOI: 10.1111/bph.12445] [Citation(s) in RCA: 509] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. G protein-coupled receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
36
|
Davenport AP, Alexander SPH, Sharman JL, Pawson AJ, Benson HE, Monaghan AE, Liew WC, Mpamhanga CP, Bonner TI, Neubig RR, Pin JP, Spedding M, Harmar AJ. International Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list: recommendations for new pairings with cognate ligands. Pharmacol Rev 2013; 65:967-86. [PMID: 23686350 PMCID: PMC3698937 DOI: 10.1124/pr.112.007179] [Citation(s) in RCA: 231] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In 2005, the International Union of Basic and Clinical Pharmacology Committee on Receptor Nomenclature and Drug Classification (NC-IUPHAR) published a catalog of all of the human gene sequences known or predicted to encode G protein-coupled receptors (GPCRs), excluding sensory receptors. This review updates the list of orphan GPCRs and describes the criteria used by NC-IUPHAR to recommend the pairing of an orphan receptor with its cognate ligand(s). The following recommendations are made for new receptor names based on 11 pairings for class A GPCRs: hydroxycarboxylic acid receptors [HCA₁ (GPR81) with lactate, HCA₂ (GPR109A) with 3-hydroxybutyric acid, HCA₃ (GPR109B) with 3-hydroxyoctanoic acid]; lysophosphatidic acid receptors [LPA₄ (GPR23), LPA₅ (GPR92), LPA₆ (P2Y5)]; free fatty acid receptors [FFA4 (GPR120) with omega-3 fatty acids]; chemerin receptor (CMKLR1; ChemR23) with chemerin; CXCR7 (CMKOR1) with chemokines CXCL12 (SDF-1) and CXCL11 (ITAC); succinate receptor (SUCNR1) with succinate; and oxoglutarate receptor [OXGR1 with 2-oxoglutarate]. Pairings are highlighted for an additional 30 receptors in class A where further input is needed from the scientific community to validate these findings. Fifty-seven human class A receptors (excluding pseudogenes) are still considered orphans; information has been provided where there is a significant phenotype in genetically modified animals. In class B, six pairings have been reported by a single publication, with 28 (excluding pseudogenes) still classified as orphans. Seven orphan receptors remain in class C, with one pairing described by a single paper. The objective is to stimulate research into confirming pairings of orphan receptors where there is currently limited information and to identify cognate ligands for the remaining GPCRs. Further information can be found on the IUPHAR Database website (http://www.iuphar-db.org).
Collapse
Affiliation(s)
- Anthony P Davenport
- Clinical Pharmacology Unit, University of Cambridge, Level 6, Centre for Clinical Investigation, Box 110, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lu X, Zhang N, Meng B, Dong S, Hu Y. Involvement of GPR12 in the regulation of cell proliferation and survival. Mol Cell Biochem 2012; 366:101-10. [PMID: 22430950 DOI: 10.1007/s11010-012-1287-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 03/02/2012] [Indexed: 01/27/2023]
Abstract
GPR12, a member of the orphan G-protein-coupled receptor family, constitutively activates the Gs protein and increases intracellular cyclic AMP concentrations. GPR12 can be activated by its known ligand-sphingosylphosphorylcholine, which regulates cellular physiological activities, including proliferation, neurite extension, cell clustering, and maintenance of meiotic arrest. However, signaling pathways involved in the GPR12-mediated physiological and biochemical changes are still not clearly illustrated. In the present study, heterologous GPR12 expression was demonstrated to promote proliferation and survival in human embryonic kidney 293 cells. Immunochemical analysis showed that Ki67, a prototypic cell cycle-related nuclear protein, might participate in the regulation of GPR12-mediated cell proliferation. Activation of extracellular signal-regulated protein kinase signaling and increased total Erk1/2 and B-cell lymphoma/leukemia-2 expression were also observed in HEK293 cells overexpressing human GPR12. In addition, we found that GPR12 promoted cell survival under serum deprivation, indicating that GPR12 may play a role in cell proliferation and survival.
Collapse
Affiliation(s)
- Xiaoming Lu
- Institutes for Advanced Interdisciplinary Research, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | | | | | | | | |
Collapse
|
38
|
Jancic CC, Cabrini M, Gabelloni ML, Rodríguez Rodrigues C, Salamone G, Trevani AS, Geffner J. Low extracellular pH stimulates the production of IL-1β by human monocytes. Cytokine 2011; 57:258-68. [PMID: 22154780 DOI: 10.1016/j.cyto.2011.11.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 10/06/2011] [Accepted: 11/17/2011] [Indexed: 10/14/2022]
Abstract
The development of acidic environments is a hallmark of inflammatory processes of different etiology. We have previously shown that transient exposure to acidic conditions, similar to those encountered in vivo, induces the activation of neutrophils and the phenotypic maturation of dendritic cells. We here report that extracellular acidosis (pH 6.5) selectively stimulates the production and the secretion of IL-1β by human monocytes without affecting the production of TNF-α, IL-6 and the expression of CD40, CD80, CD86, and HLA-DR. Stimulation of IL-1β production by pH 6.5-treated monocytes was shown to be dependent on caspase-1 activity, and it was also observed using peripheral blood mononuclear cells instead of isolated monocytes. Contrasting with the results in monocytes, we found that pH 6.5 did not stimulate any production of IL-1β by macrophages. Changes in intracellular pH seem to be involved in the stimulation of IL-1β production. In fact, monocytes cultured at pH 6.5 undergo a fall in the values of intracellular pH while the inhibitor of the Na+/H+ exchanger, 5-(N-ethyl-N-isopropyl)amiloride induced both, a decrease in the values of intracellular pH and the stimulation of IL-1β production. Real time quantitative PCR assays indicated that monocytes cultured either at pH 6.5 or in the presence of 5-(N-ethyl-N-isopropyl)amiloride expressed higher levels of pro-IL-1β mRNA suggesting that low values of intracellular pH enhance the production of IL-1β, at least in part, by stimulating the synthesis of its precursor.
Collapse
Affiliation(s)
- Carolina Cristina Jancic
- Instituto de Investigaciones Hematológicas (IIHEMA), Academia Nacional de Medicina, Buenos Aires, Argentina.
| | | | | | | | | | | | | |
Collapse
|
39
|
Chen A, Dong L, Leffler NR, Asch AS, Witte ON, Yang LV. Activation of GPR4 by acidosis increases endothelial cell adhesion through the cAMP/Epac pathway. PLoS One 2011; 6:e27586. [PMID: 22110680 PMCID: PMC3217975 DOI: 10.1371/journal.pone.0027586] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 10/20/2011] [Indexed: 01/11/2023] Open
Abstract
Endothelium-leukocyte interaction is critical for inflammatory responses. Whereas the tissue microenvironments are often acidic at inflammatory sites, the mechanisms by which cells respond to acidosis are not well understood. Using molecular, cellular and biochemical approaches, we demonstrate that activation of GPR4, a proton-sensing G protein-coupled receptor, by isocapnic acidosis increases the adhesiveness of human umbilical vein endothelial cells (HUVECs) that express GPR4 endogenously. Acidosis in combination with GPR4 overexpression further augments HUVEC adhesion with U937 monocytes. In contrast, overexpression of a G protein signaling-defective DRY motif mutant (R115A) of GPR4 does not elicit any increase of HUVEC adhesion, indicating the requirement of G protein signaling. Downregulation of GPR4 expression by RNA interference reduces the acidosis-induced HUVEC adhesion. To delineate downstream pathways, we show that inhibition of adenylate cyclase by inhibitors, 2',5'-dideoxyadenosine (DDA) or SQ 22536, attenuates acidosis/GPR4-induced HUVEC adhesion. Consistently, treatment with a cAMP analog or a G(i) signaling inhibitor increases HUVEC adhesiveness, suggesting a role of the G(s)/cAMP signaling in this process. We further show that the cAMP downstream effector Epac is important for acidosis/GPR4-induced cell adhesion. Moreover, activation of GPR4 by acidosis increases the expression of vascular adhesion molecules E-selectin, VCAM-1 and ICAM-1, which are functionally involved in acidosis/GPR4-mediated HUVEC adhesion. Similarly, hypercapnic acidosis can also activate GPR4 to stimulate HUVEC adhesion molecule expression and adhesiveness. These results suggest that acidosis/GPR4 signaling regulates endothelial cell adhesion mainly through the G(s)/cAMP/Epac pathway and may play a role in the inflammatory response of vascular endothelial cells.
Collapse
Affiliation(s)
- Aishe Chen
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Lixue Dong
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Nancy R. Leffler
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Adam S. Asch
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, United States of America
| | - Owen N. Witte
- Howard Hughes Medical Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Li V. Yang
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
40
|
Hasegawa H, Lei J, Matsumoto T, Onishi S, Suemori K, Yasukawa M. Lysophosphatidylcholine enhances the suppressive function of human naturally occurring regulatory T cells through TGF-β production. Biochem Biophys Res Commun 2011; 415:526-31. [PMID: 22074829 DOI: 10.1016/j.bbrc.2011.10.119] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 10/26/2011] [Indexed: 11/30/2022]
Abstract
Naturally occurring CD4(+)CD25(+) regulatory T cells (nTregs) play a pivotal role in the maintenance of self-tolerance and immune homeostasis. To gain insight into the mechanism of action of nTregs in pathological and physiological immune responses, it is important to analyze bioactive molecules that modulate the maintenance and function of nTregs. From a library of bioactive lipids, we obtained lysophosphatidylcholine (LPC) as a molecule that enhanced the Foxp3 expression and suppressive function of human nTregs significantly in comparison with those of DMSO-treated nTregs (control). The expression levels of TGF-β1 mRNA and protein in LPC-treated nTregs were significantly higher than those in control nTregs. After treatment with anti-TGF-β1 antibody, the increases in Foxp3 expression and the suppressive properties of LPC-treated nTregs returned to the levels observed in control nTregs. These findings indicate that LPC enhances Foxp3 expression and the suppressive function of nTregs through TGF-β1 produced by nTregs themselves. Experimental knockdown of G2A and GPR4 showed that this LPC-induced TGF-β1 expression in nTregs was due to G2A signaling, and did not involve GPR4. Moreover, JNK was a major contributor to LPC-induced TGF-β1 expression in nTregs, although LPC activated MAPKs including ERK1/2, p38 MAPK, and JNK via G2A. LPC is a bioactive lysolipid highly abundant in the circulation. Therefore, LPC may contribute to the maintenance and function of human nTregs in vivo.
Collapse
Affiliation(s)
- Hitoshi Hasegawa
- Department of Bioregulatory Medicine, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan.
| | | | | | | | | | | |
Collapse
|
41
|
Lysophosphatidylcholine: A Novel Modulator of Trypanosoma cruzi Transmission. J Parasitol Res 2011; 2012:625838. [PMID: 22132309 PMCID: PMC3206328 DOI: 10.1155/2012/625838] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 07/29/2011] [Accepted: 09/12/2011] [Indexed: 01/18/2023] Open
Abstract
Lysophosphatidylcholine is a bioactive lipid that regulates a large number of cellular processes and is especially present during the deposition and infiltration of inflammatory cells and deposition of atheromatous plaque. Such molecule is also present in saliva and feces of the hematophagous organism Rhodnius prolixus, a triatominae bug vector of Chagas disease. We have recently demonstrated that LPC is a modulator of Trypanosoma cruzi transmission. It acts as a powerful chemoattractant for inflammatory cells at the site of the insect bite, which will provide a concentrated population of cells available for parasite infection. Also, LPC increases macrophage intracellular calcium concentrations that ultimately enhance parasite invasion. Finally, LPC inhibits NO production by macrophages stimulated by live T. cruzi, and thus interferes with the immune system of the vertebrate host. In the present paper, we discuss the main signaling mechanisms that are likely used by such molecule and their eventual use as targets to block parasite transmission and the pathogenesis of Chagas disease.
Collapse
|
42
|
LysoPC and PAF Trigger Arachidonic Acid Release by Divergent Signaling Mechanisms in Monocytes. J Lipids 2011; 2011:532145. [PMID: 21912747 PMCID: PMC3170782 DOI: 10.1155/2011/532145] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 04/29/2011] [Accepted: 05/28/2011] [Indexed: 11/17/2022] Open
Abstract
Oxidized low-density lipoproteins (LDLs) play an important role during the development of atherosclerosis characterized by intimal inflammation and macrophage accumulation. A key component of LDL is lysophosphatidylcholine (lysoPC). LysoPC is a strong proinflammatory mediator, and its mechanism is uncertain, but it has been suggested to be mediated via the platelet activating factor (PAF) receptor. Here, we report that PAF triggers a pertussis toxin- (PTX-) sensitive intracellular signaling pathway leading to sequential activation of sPLA(2), PLD, cPLA(2), and AA release in human-derived monocytes. In contrast, lysoPC initiates two signaling pathways, one sequentially activating PLD and cPLA(2), and a second parallel PTX-sensitive pathway activating cPLA(2) with concomitant activation of sPLA(2), all leading to AA release. In conclusion, lysoPC and PAF stimulate AA release by divergent pathways suggesting involvement of independent receptors. Elucidation of monocyte lysoPC-specific signaling mechanisms will aid in the development of novel strategies for atherosclerosis prevention, diagnosis, and therapy.
Collapse
|
43
|
Castellone RD, Leffler NR, Dong L, Yang LV. Inhibition of tumor cell migration and metastasis by the proton-sensing GPR4 receptor. Cancer Lett 2011; 312:197-208. [PMID: 21917373 DOI: 10.1016/j.canlet.2011.08.013] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 08/10/2011] [Accepted: 08/10/2011] [Indexed: 01/07/2023]
Abstract
GPR4 is a member of the proton-sensing G protein-coupled receptor family. Within tumor microenvironments, the interstitial acidic pH may activate GPR4 to regulate the behavior of tumor cells. Mouse B16F10 melanoma cells and TRAMP-C1 prostate cancer cells, genetically engineered to overexpress GPR4 or the control vector, were subject to a series of cell migration, invasion and metastasis assays. Upon GPR4 overexpression and activation in an acidic pH, the migration of B16F10 and TRAMP-C1 cells was substantially inhibited in comparison to the vector control. Similar results were observed in the Matrigel invasion and transendothelial invasion assays. At the molecular level, stimulation of GPR4 by acidosis induced the activation of RhoA and the formation of actin stress fibers. In addition, treating B16F10 cells with the known Rho activator CN01 (calpeptin) strongly inhibited cell migration, recapitulating the acidosis/GPR4-induced motility inhibition phenotype. To examine the biological effects in vivo, B16F10 melanoma cells were intravenously injected into syngeneic C57BL/6 mice and pulmonary metastasis was inhibited by approximately 80% in GPR4-overexpressing B16F10 cells in comparison to the vector control. Upon treatment with the Rho activator CN01, the phenotype of the B16F10 vector cells paralleled that of the GPR4-overexpressing cells in cell migration and metastasis assays. These findings suggest that GPR4 activation by an acidic pH inhibits tumor cell migration and invasion, and the Rho GTPase is at least partly responsible for this phenotype.
Collapse
Affiliation(s)
- Reid D Castellone
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | | | | | | |
Collapse
|
44
|
Han MS, Lim YM, Quan W, Kim JR, Chung KW, Kang M, Kim S, Park SY, Han JS, Park SY, Cheon HG, Dal Rhee S, Park TS, Lee MS. Lysophosphatidylcholine as an effector of fatty acid-induced insulin resistance. J Lipid Res 2011; 52:1234-1246. [PMID: 21447485 DOI: 10.1194/jlr.m014787] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The mechanism of FFA-induced insulin resistance is not fully understood. We have searched for effector molecules(s) in FFA-induced insulin resistance. Palmitic acid (PA) but not oleic acid (OA) induced insulin resistance in L6 myotubes through C-Jun N-terminal kinase (JNK) and insulin receptor substrate 1 (IRS-1) Ser307 phosphorylation. Inhibitors of ceramide synthesis did not block insulin resistance by PA. However, inhibition of the conversion of PA to lysophosphatidylcholine (LPC) by calcium-independent phospholipase A₂ (iPLA₂) inhibitors, such as bromoenol lactone (BEL) or palmitoyl trifluoromethyl ketone (PACOCF₃), prevented insulin resistance by PA. iPLA₂ inhibitors or iPLA₂ small interfering RNA (siRNA) attenuated JNK or IRS-1 Ser307 phosphorylation by PA. PA treatment increased LPC content, which was reversed by iPLA₂ inhibitors or iPLA₂ siRNA. The intracellular DAG level was increased by iPLA₂ inhibitors, despite ameliorated insulin resistance. Pertussis toxin (PTX), which inhibits LPC action through the G-protein coupled receptor (GPCR)/Gα(i), reversed insulin resistance by PA. BEL administration ameliorated insulin resistance and diabetes in db/db mice. JNK and IRS-1Ser307 phosphorylation in the liver and muscle of db/db mice was attenuated by BEL. LPC content was increased in the liver and muscle of db/db mice, which was suppressed by BEL. These findings implicate LPC as an important lipid intermediate that links saturated fatty acids to insulin resistance.
Collapse
Affiliation(s)
- Myoung Sook Han
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Yu-Mi Lim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Wenying Quan
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Jung Ran Kim
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University of Medicine and Science, Inchon 406-840, Korea
| | - Kun Wook Chung
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Mira Kang
- Center for Health Promotion, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Sunshin Kim
- Carcinogenesis Branch, Korean National Cancer Center, Goyang 410-769, Korea
| | - Sun Young Park
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Joong-Soo Han
- Institute of Biomedical Science, College of Medicine, Hanyang University, Seoul 133-791, Korea; Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
| | - Shin-Young Park
- Institute of Biomedical Science, College of Medicine, Hanyang University, Seoul 133-791, Korea; Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
| | - Hyae Gyeong Cheon
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University of Medicine and Science, Inchon 406-840, Korea
| | - Sang Dal Rhee
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, Daejon 305-343, Korea
| | - Tae-Sik Park
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University of Medicine and Science, Inchon 406-840, Korea.
| | - Myung-Shik Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea.
| |
Collapse
|
45
|
da Silva Cunha KC, Fuly AL, de Araujo EG. A phospholipase A₂ isolated from Lachesis muta snake venom increases the survival of retinal ganglion cells in vitro. Toxicon 2011; 57:580-5. [PMID: 21223976 DOI: 10.1016/j.toxicon.2010.12.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 12/08/2010] [Accepted: 12/30/2010] [Indexed: 12/31/2022]
Abstract
We have previously showed that a phospholipase A₂ isolated from Lachesis muta snake venom and named LM-PLA₂-I displayed particular biological activities, as hemolysis, inhibition on platelet aggregation, edema induction and myotoxicity. In the present work, we evaluated the effect of LM-PLA₂-I on the survival of axotomized rat retinal ganglion cells kept in vitro, as well as its mechanism of action. Our results clearly showed that treatment with LM-PLA₂-I increased the survival of ganglion cells (100% when compared to control cultures) and the treatment of LM-PLA₂-I with p-bromophenacyl bromide abolished this effect. This result indicates that the effect of LM-PLA₂-I on ganglion cell survival is entirely dependent on its enzymatic activity and the generation of lysophosphatidylcholine (LPC) may be a prerequisite to the observed survival. In fact, commercial LPC mimicked the effect of LM-PLA₂-I upon ganglion cell survival. To investigate the mechanism of action of LM-PLA₂-I, cultures were treated with chelerythrine chloride, BAPTA-AM, rottlerin and also with an inhibitor of c-junc kinase (JNKi). Our results showed that rottlerin and JNK inhibitor abolished the LM-PLA₂-I on ganglion cell survival. Taken together, our results showed that LM-PLA₂-I and its enzymatic product, LPC promoted survival of retinal ganglion cells through the protein kinase C pathway and strongly suggest a possible role of the PLA₂ enzyme and LPC in controlling the survival of axotomized neuronal cells.
Collapse
Affiliation(s)
- Karinne Cristinne da Silva Cunha
- Programa de Pós-Graduação em Neurociências, Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | | | | |
Collapse
|
46
|
Samadi N, Bekele R, Capatos D, Venkatraman G, Sariahmetoglu M, Brindley DN. Regulation of lysophosphatidate signaling by autotaxin and lipid phosphate phosphatases with respect to tumor progression, angiogenesis, metastasis and chemo-resistance. Biochimie 2010; 93:61-70. [PMID: 20709140 DOI: 10.1016/j.biochi.2010.08.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 08/03/2010] [Accepted: 08/04/2010] [Indexed: 12/21/2022]
Abstract
Evidence from clinical, animal and cell culture studies demonstrates that increased autotaxin (ATX) expression is responsible for enhancing tumor progression, cell migration, metastases, angiogenesis and chemo-resistance. These effects depend mainly on the rapid formation of lysophosphatidate (LPA) by ATX. Circulating LPA has a half-life of about 3 min in mice and it is degraded by the ecto-activities of lipid phosphate phosphatases (LPPs). These enzymes also hydrolyze extracellular sphingosine 1-phosphate (S1P), a potent signal for cell division, survival and angiogenesis. Many aggressive tumor cells express high ATX levels and low LPP activities. This favors the formation of locally high LPA and S1P concentrations. Furthermore, LPPs attenuate signaling downstream of the activation of G-protein coupled receptors and receptor tyrosine kinases. Therefore, we propose that the low expression of LPPs in many tumor cells makes them hypersensitive to growth promoting and survival signals that are provided by LPA, S1P, platelet-derived growth factor (PDGF) and epidermal growth factor (EGF). One of the key signaling pathways in this respect appears to be activation of phospholipase D (PLD) and phosphatidate (PA) production. This is required for the transactivations of the EGFR and PDGFR and also for LPA-induced cell migration. PA also increases the activities of ERK, mTOR, myc and sphingosine kinase-1 (SK-1), which provide individual signals for cells division, survival, chemo-resistance and angiogenesis. This review focuses on the balance of signaling by bioactive lipids including LPA, phosphatidylinositol 3,4,5-trisphosphate, PA and S1P versus the action of ceramides. We will discuss how these lipid mediators interact to produce an aggressive neoplastic phenotype.
Collapse
Affiliation(s)
- Nasser Samadi
- Signal Transduction Research Group, Department of Biochemistry, School of Molecular and Systems Medicine, University of Alberta, Edmonton, T6G 2S2 Alberta, Canada
| | | | | | | | | | | |
Collapse
|
47
|
Rat CC chemokine receptor 4 is the functional homologue of human CC chemokine receptor 4 and can interact with human CCL17 and CCL22. CHINESE SCIENCE BULLETIN-CHINESE 2010. [DOI: 10.1007/s11434-010-0157-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
48
|
Kim DS, Park SH, Kwon SB, Kwon NS, Park KC. Sphingosylphosphorylcholine inhibits melanin synthesis via pertussis toxin-sensitive MITF degradation. J Pharm Pharmacol 2010; 62:181-7. [DOI: 10.1211/jpp.62.02.0005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Abstract
Objectives
Sphingolipids act as structural components in cell membranes, and form lipid intermediates that have functional roles as signalling molecules in various cellular processes. Our previous findings have suggested that sphingolipid metabolites are deeply involved in the regulation of melanogenic processes. In this study we aimed to examine sphingosylphosphorylcholine-mediated signalling pathways related to melanogenesis.
Methods
We determined the hypopigmenting effects and the related signalling pathways of sphingosylphosphorylcholine in Mel-Ab cells. In particular, we analysed the involvement of the G-protein-coupled receptor in sphingosylphosphorylcholine-induced MITF degradation.
Key findings
Western blotting revealed that sphingosylphosphorylcholine induced the activation of extracellular signal-regulated kinase (ERK), as well as Akt. Moreover, the specific Akt pathway inhibitor LY294002 blocked the hypopigmenting effect of sphingosylphosphorylcholine and abrogated the sphingosylphosphorylcholine-mediated down-regulation of microphthalmia-associated transcription factor (MITF), showing that the Akt pathway is involved in sphingosylphosphorylcholine-mediated melanin inhibition. Treatment with the proteasome inhibitor MG132 blocked the decrease in MITF by sphingosylphosphorylcholine, but sphingosylphosphorylcholine did not decrease the level of MITF mRNA, indicating that the reduction in the level of MITF results from MITF degradation. Furthermore, pre-incubation of Mel-Ab cells with pertussis toxin completely abolished the hypopigmenting effects and the activation of ERK and Akt by sphingosylphosphorylcholine, suggesting that the effects of sphingosylphosphorylcholine are mainly dependent on the G-protein-coupled receptor).
Conclusions
Together, these results suggest that sphingosylphosphorylcholine reduces melanin synthesis via pertussis toxin-sensitive ERK and Akt activation, and subsequent MITF degradation.
Collapse
Affiliation(s)
- Dong-Seok Kim
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Seo-Hyoung Park
- Department of Dermatology, Seoul National University, Bundang Hospital, Kyoungki-Do, Republic of Korea
| | - Sun-Bang Kwon
- Department of Dermatology, Seoul National University, Bundang Hospital, Kyoungki-Do, Republic of Korea
| | - Nyoun Soo Kwon
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Kyoung-Chan Park
- Department of Dermatology, Seoul National University, Bundang Hospital, Kyoungki-Do, Republic of Korea
| |
Collapse
|
49
|
|
50
|
Orphan 7TM receptors. Br J Pharmacol 2009. [DOI: 10.1111/j.1476-5381.2009.00501_2.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|