1
|
Zhang X, Wu W, Li Y, Peng Z. Exploring the role and therapeutic potential of lipid metabolism in acute kidney injury. Ren Fail 2024; 46:2403652. [PMID: 39319697 PMCID: PMC11425701 DOI: 10.1080/0886022x.2024.2403652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
Acute kidney injury (AKI) is a prevalent condition, yet no specific treatment is available. Extensive research has revealed the pivotal role of lipid-related alterations in AKI. Lipid metabolism plays an essential role in the sustenance of the kidneys. In addition to their energy-supplying function, lipids contribute to the formation of renal biomembranes and the establishment of the renal microenvironment. Moreover, lipids or their metabolites actively participate in signal transduction, which governs various vital biological processes, such as proliferation, differentiation, apoptosis, autophagy, and epithelial-mesenchymal transition. While previous studies have focused predominantly on abnormalities in lipid metabolism in chronic kidney disease, this review focuses on lipid metabolism anomalies in AKI. We explore the significance of lipid metabolism products as potential biomarkers for the early diagnosis and classification of AKI. Additionally, this review assesses current preclinical investigations on the modulation of lipid metabolism in the progression of AKI. Finally, on the basis of existing research, this review proposes future directions, highlights challenges, and presents novel targets and innovative ideas for the treatment of and intervention in AKI.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Wen Wu
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
- Department of Critical Care Medicine, Yichang Central People's Hospital, Yichang, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
- Department of Critical Care Medicine, Center of Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Liu S, Wang Y, Ying L, Li H, Zhang K, Liang N, Luo G, Xiao L. Quercetin Mitigates Lysophosphatidylcholine (LPC)-Induced Neutrophil Extracellular Traps (NETs) Formation through Inhibiting the P2X7R/P38MAPK/NOX2 Pathway. Int J Mol Sci 2024; 25:9411. [PMID: 39273358 PMCID: PMC11395007 DOI: 10.3390/ijms25179411] [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: 07/18/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are three-dimensional reticular structures that release chromatin and cellular contents extracellularly upon neutrophil activation. As a novel effector mechanism of neutrophils, NETs possess the capacity to amplify localized inflammation and have been demonstrated to contribute to the exacerbation of various inflammatory diseases, including cardiovascular diseases and tumors. It is suggested that lysophosphatidylcholine (LPC), as the primary active component of oxidized low-density lipoprotein, represents a significant risk factor for various inflammatory diseases, such as cardiovascular diseases and neurodegenerative diseases. However, the specific mechanism of NETs formation induced by LPC remains unclear. Quercetin has garnered considerable attention due to its anti-inflammatory properties, serving as a prevalent flavonoid in daily diet. However, little is currently known about the underlying mechanisms by which quercetin inhibits NETs formation and alleviates associated diseases. In our study, we utilized LPC-treated primary rat neutrophils to establish an in vitro model of NETs formation, which was subsequently subjected to treatment with a combination of quercetin or relevant inhibitors/activators. Compared to the control group, the markers of NETs and the expression of P2X7R/P38MAPK/NOX2 pathway-associated proteins were significantly increased in cells treated with LPC alone. Quercetin intervention decreased the LPC-induced upregulation of the P2X7R/P38MAPK/NOX2 pathway and effectively reduced the expression of NETs markers. The results obtained using a P2X7R antagonist/activator and P38MAPK inhibitor/activator support these findings. In summary, quercetin reversed the upregulation of the LPC-induced P2X7R/P38MAPK/NOX2 pathway, further mitigating NETs formation. Our study investigated the potential mechanism of LPC-induced NETs formation, elucidated the inhibitory effect of quercetin on NETs formation, and offered new insights into the anti-inflammatory properties of quercetin.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gang Luo
- Xiangya School of Public Health, Central South University, Changsha 410013, China; (S.L.); (Y.W.); (L.Y.); (H.L.); (K.Z.); (N.L.)
| | - Lin Xiao
- Xiangya School of Public Health, Central South University, Changsha 410013, China; (S.L.); (Y.W.); (L.Y.); (H.L.); (K.Z.); (N.L.)
| |
Collapse
|
3
|
Kume H, Harigane R, Rikimaru M. Involvement of Lysophospholipids in Pulmonary Vascular Functions and Diseases. Biomedicines 2024; 12:124. [PMID: 38255229 PMCID: PMC10813361 DOI: 10.3390/biomedicines12010124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/26/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Extracellular lysophospholipids (lysophosphatidic acid, lysophosphatidylcholine, sphingosine 1-phosphate, etc.), which are synthesized from phospholipids in the cell membrane, act as lipid mediators, and mediate various cellular responses in constituent cells in the respiratory system, such as contraction, proliferation, migration, and cytoskeletal organization. In addition to these effects, the expression of the adhesion molecules is enhanced by these extracellular lysophospholipids in pulmonary endothelial cells. These effects are exerted via specific G protein-coupled receptors. Rho, Ras, and phospholipase C (PLC) have been proven to be their signaling pathways, related to Ca2+ signaling due to Ca2+ dynamics and Ca2+ sensitization. Therefore, lysophospholipids probably induce pulmonary vascular remodeling through phenotype changes in smooth muscle cells, endothelial cells, and fibroblasts, likely resulting in acute respiratory distress syndrome due to vascular leak, pulmonary hypertension, and pulmonary fibrosis. Moreover, lysophospholipids induce the recruitment of inflammatory cells to the lungs via the enhancement of adhesion molecules in endothelial cells, potentially leading to the development of asthma. These results demonstrate that lysophospholipids may be novel therapeutic targets not only for injury, fibrosis, and hypertension in the lung, but also for asthma. In this review, we discuss the mechanisms of the effects of lysophospholipids on the respiratory system, and the possibility of precision medicine targeting lysophospholipids as treatable traits of these diseases.
Collapse
Affiliation(s)
- Hiroaki Kume
- Department of Infectious Diseases and Respiratory Medicine, Fukushima Medical University Aizu Medical Center, 21-2 Maeda, Tanisawa, Kawahigashi, Aizuwakamatsu City 969-3492, Fukushima, Japan; (R.H.); (M.R.)
| | | | | |
Collapse
|
4
|
He S, Wang Y, Liu Z, Zhang J, Hao X, Wang X, Zhou Z, Wang R, Zhao Y. Metabolomic signatures associated with pathological angiogenesis in moyamoya disease. Clin Transl Med 2023; 13:e1492. [PMID: 38037492 PMCID: PMC10689969 DOI: 10.1002/ctm2.1492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023] Open
Affiliation(s)
- Shihao He
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yanru Wang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Ziqi Liu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Junze Zhang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Xiaokuan Hao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Xilong Wang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Zhenyu Zhou
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Rong Wang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Yuanli Zhao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurosurgeryPeking Union Medical College HospitalPeking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
5
|
Luo X, Liu M, Wang S, Chen Y, Bao X, Lv Y, Zhang S, Xu B, Weng X, Bai X, Zeng M, Zhao C, Li J, Jia H, Yu B. Combining metabolomics and OCT to reveal plasma metabolic profiling and biomarkers of plaque erosion and plaque rupture in STEMI patients. Int J Cardiol 2023; 390:131223. [PMID: 37517782 DOI: 10.1016/j.ijcard.2023.131223] [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: 10/20/2022] [Revised: 01/15/2023] [Accepted: 02/26/2023] [Indexed: 08/01/2023]
Abstract
OBJECTIVE Plaque erosion (PE) and plaque rupture (PR) are the main subtypes of ST-segment elevation myocardial infarction (STEMI), the differences of metabolic patterns between PE and PR remain largely unknown. METHODS 132 STEMI patients were divided into training set (PR, n = 36; PE, n = 36) and test set (PR, n = 30; PE, n = 30), the plasma from patients were analyzed by liquid chromatography quadruple time-of-flight mass spectrometry. RESULTS We identified 56 and 28 differences in training and test set, respectively. Among these metabolites, it was found that docosahexaenoic acid (DHA), salicylic acid and proline were recognized in both tests. Receiver Operating Characteristic (ROC) analysis showed that the area under curve of docosahexaenoic acid (DHA) was 0.81 and 0.75 in training and test samples, respectively; proline was 0.67 and 0.74 in training and test samples, respectively; salicylic acid was 0.70 and 0.73 in training and test samples, respectively. CONCLUSIONS DHA, salicylic acid, and proline could be used as non-invasive biomarkers to differentiate PE and PR.
Collapse
Affiliation(s)
- Xing Luo
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Minghao Liu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Shengfang Wang
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Yuwu Chen
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Xiaoyi Bao
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Ying Lv
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Shan Zhang
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Biyi Xu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Xiuzhu Weng
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Xiaoxuan Bai
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Ming Zeng
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Chen Zhao
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Ji Li
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Haibo Jia
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China.
| | - Bo Yu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China.
| |
Collapse
|
6
|
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
|
7
|
Su D, Liao L, Zeng Q, Liao Z, Liu Y, Jin C, Zhu G, Chen C, Yang M, Ai Z, Song Y. Study on the new anti-atherosclerosis activity of different Herba patriniae through down-regulating lysophosphatidylcholine of the glycerophospholipid metabolism pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 94:153833. [PMID: 34798520 DOI: 10.1016/j.phymed.2021.153833] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/14/2021] [Accepted: 10/25/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Atherosclerosis (AS) is a multifactor cardiovascular disease characterized by chronic inflammation. The safety of long-term medication is the focus of clinical treatment selection and application. It is urgent to develop more high-efficiency and low side effects drugs to treat AS. Therefore, the screening of anti-AS drugs with high efficiency and low toxicity from phytomedicine has attracted more and more attention. PURPOSE The aim of this study was to explore the new pharmacological effect of Herba patriniae against AS, to find the best origin and extraction part of Herba patriniae, furthermore, to reveal its potential action mechanism. METHODS Apolipoprotein E gene-knockout (ApoE-/-) mice were orally administered with different extracts of Patrinia villosa Juss (PVJ) and Patrinia scabiosaefolia Fisch (PSF). Their anti-AS effect was comprehensively evaluated by small animal ultrasound, HE staining, Oil-Red O staining, platelet aggregation rate and blood lipid level. Lipid metabolomics and network pharmacology were used to study the mechanism of drug action. Finally, the expression of related proteins were detected by western blots and immunofluorescence. RESULTS PVJ EtOAc extract and PSF EtOAc extract could significantly reduce vascular plaque, liver inflammation, platelet aggregation and blood lipid levels in AS model. By comparison, the effect of PVJEE was better than that of PSFEE. Furthermore, the results of differential metabolites indicated that PVJEE may inhibit the apoptosis of vascular endothelial cells, proliferation and migration of smooth muscle cells by reversing lysophosphatidylcholine (LPC) in the glycerophospholipid metabolic pathway, so as to play an anti-AS role. This result was double verified by KEGG based metabolic pathway enrichment analysis and related protein expression study. CONCLUSION By changing glycerophospholipid metabolism pathway, Herba patriniae can significantly regulate lipid metabolism and inflammatory level, showing the development potential of anti-AS, which provides new candidate drugs and good prospects for the safe treatment of AS. In addition, through comparison, this study also confirmed that PVJEE was the best origin and extraction part of anti-AS.
Collapse
Affiliation(s)
- Dan Su
- Key Laboratory of depression animal model based on TCM syndrome, Jiangxi Administration of traditional Chinese Medicine, Key Laboratory of TCM for prevention and treatment of brain diseases with cognitive impairment, Jiangxi Province, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330006, China
| | - Liangliang Liao
- Key Laboratory of depression animal model based on TCM syndrome, Jiangxi Administration of traditional Chinese Medicine, Key Laboratory of TCM for prevention and treatment of brain diseases with cognitive impairment, Jiangxi Province, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330006, China
| | - Qiang Zeng
- College of Pharmacy,Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Zhou Liao
- Key Laboratory of depression animal model based on TCM syndrome, Jiangxi Administration of traditional Chinese Medicine, Key Laboratory of TCM for prevention and treatment of brain diseases with cognitive impairment, Jiangxi Province, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330006, China
| | - Yali Liu
- Key Laboratory of depression animal model based on TCM syndrome, Jiangxi Administration of traditional Chinese Medicine, Key Laboratory of TCM for prevention and treatment of brain diseases with cognitive impairment, Jiangxi Province, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330006, China
| | - Chen Jin
- Key Laboratory of depression animal model based on TCM syndrome, Jiangxi Administration of traditional Chinese Medicine, Key Laboratory of TCM for prevention and treatment of brain diseases with cognitive impairment, Jiangxi Province, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330006, China
| | - Genhua Zhu
- Key Laboratory of depression animal model based on TCM syndrome, Jiangxi Administration of traditional Chinese Medicine, Key Laboratory of TCM for prevention and treatment of brain diseases with cognitive impairment, Jiangxi Province, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330006, China
| | - Changlian Chen
- Key Laboratory of depression animal model based on TCM syndrome, Jiangxi Administration of traditional Chinese Medicine, Key Laboratory of TCM for prevention and treatment of brain diseases with cognitive impairment, Jiangxi Province, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330006, China
| | - Ming Yang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330006, China
| | - Zhifu Ai
- Key Laboratory of depression animal model based on TCM syndrome, Jiangxi Administration of traditional Chinese Medicine, Key Laboratory of TCM for prevention and treatment of brain diseases with cognitive impairment, Jiangxi Province, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330006, China..
| | - Yonggui Song
- Key Laboratory of depression animal model based on TCM syndrome, Jiangxi Administration of traditional Chinese Medicine, Key Laboratory of TCM for prevention and treatment of brain diseases with cognitive impairment, Jiangxi Province, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330006, China..
| |
Collapse
|
8
|
Yan HC, Sun Y, Zhang MY, Zhang SE, Sun JD, Dyce PW, Klinger FG, De Felici M, Shen W, Cheng SF. YAP regulates porcine skin-derived stem cells self-renewal partly by repressing Wnt/β-catenin signaling pathway. Histochem Cell Biol 2021; 157:39-50. [PMID: 34586448 DOI: 10.1007/s00418-021-02034-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2021] [Indexed: 01/02/2023]
Abstract
Skin-derived stem cells (SDSCs) are a class of adult stem cells (ASCs) that have the ability to self-renew and differentiate. The regulation mechanisms involved in the differentiation of SDSCs are a hot topic. In this paper, we explore the link between the transcriptional regulator yes-associated protein (YAP) and the fate of porcine SDSCs (pSDSCs). We found that lysophosphatidylcholine (LPC) activates YAP, promotes pSDSCs pluripotency, and counteracts transdifferentiation of pSDSCs into porcine primordial germ cell-like cells (pPGCLCs). YAP promotes the pluripotent state of pSDSCs by maintaining the high expression of the pluripotency genes Oct4 and Sox2. The overexpression of YAP prevented the differentiation of pSDSCs, and the depletion of YAP by small interfering RNA (siRNAs) suppressed the self-renewal of pSDSCs. In addition, we found that YAP regulates the fate of pSDSCs through a mechanism related to the Wnt/β-catenin signaling pathway. When an activator of the Wnt/β-catenin signaling pathway, CHIR99021, was added to pSDSCs overexpressing YAP, the ability of pSDSCs to differentiate was partially restored. Conversely, when XAV939, an inhibitor of the Wnt/β-catenin signaling pathway, was added to YAP knockdown pSDSCs a higher self-renewal ability resulted. Taken together, our results suggested that YAP and the Wnt/β-catenin signaling pathway interact to regulate the fate of pSDSCs.
Collapse
Affiliation(s)
- Hong-Chen Yan
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yu Sun
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Ming-Yu Zhang
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Shu-Er Zhang
- Animal Husbandry General Station of Shandong Province, Jinan, 250010, China
| | - Jia-Dong Sun
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Paul W Dyce
- Department of Animal Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Francesca Gioia Klinger
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Wei Shen
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Shun-Feng Cheng
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
9
|
Zhang S, Li C, Feng T, Cao S, Zhou H, Li L, Hu Q, Mao X, Ji S. A Metabolic Profiling Study of Realgar-Induced Acute Kidney Injury in Mice. Front Pharmacol 2021; 12:706249. [PMID: 34497512 PMCID: PMC8419260 DOI: 10.3389/fphar.2021.706249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/11/2021] [Indexed: 11/13/2022] Open
Abstract
Realgar has been used as a type of mineral drug that contains arsenic for thousands of years. Previous studies have shown that Realgar-induced acute kidney injury is associated with abnormal metabolism, but the underlying mechanism is poorly understood. The aim of this study is to investigate the metabolic changes in serum and kidney tissues of mice exposed to Realgar by using a metabolomic approach and explore the molecular mechanisms of acute kidney injury induced by Realgar. Forty mice were randomly divided into four groups: Control group, 0.5-, 1.0, and 2.0 g/kg Realgar group. After 1 week, the body weight and kidney weight of the mice were measured. The serum and kidney samples were used for LC-MS spectroscopic metabolic profiling. Principal component analysis (PCA), correlation analysis, and pathway analysis were used to detect the nephrotoxic effects of Realgar. Body weight decreased significantly in the 2.0 g/kg group, and the kidney weight index also showed a dose-dependent increase in Realgar. The PCA score plot showed the serum and kidney tissue metabolic profile of mice exposed to 2.0 g/kg Realgar separated from the control group, while the lower-doses of 0.5 g/kg and 1.0 g/kg Realgar shown a similar view to the Control group. Thirty-three metabolites and seventeen metabolites were screened and identified in the serum and kidney of mice in a dose-dependent manner. respectively. Correlation analysis showed a strong correlation among these metabolites. Amino acid metabolism, lipid metabolism, glutathione metabolism, and purine metabolism pathways were found to be mainly associated with Realgar nephrotoxicity. This work illustrated the metabolic alterations in Realgar-induced nephrotoxic mice through a metabolomic approach.
Collapse
Affiliation(s)
- Sheng Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.,NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Chao Li
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China.,Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tingting Feng
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Shuai Cao
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Heng Zhou
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Limin Li
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Qing Hu
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Xiuhong Mao
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Shen Ji
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.,NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
He K, Chen C, Deng J, Hou YJ, Xiang Z, Yang Y. In situ detection and imaging of lysophospholipids in zebrafish using matrix-assisted laser desorption/ionization Fourier transform ion cyclotron resonance mass spectrometry. JOURNAL OF MASS SPECTROMETRY : JMS 2021; 56:e4637. [PMID: 32789983 DOI: 10.1002/jms.4637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 06/11/2023]
Abstract
In this paper, a matrix-assisted laser desorption/ionization (MALDI) Fourier transform ion cyclotron resonance (FTICR) mass spectrometry (MS) (MALDI-FTICR-MS) imaging method was developed to rapid and in situ detect the spatial distribution of lysophospholipids (LPLs) in zebrafish. The combination of MALDI with ultrahigh-resolution FTICR-MS achieves the MS imaging of LPLs with a mass resolution up to 50 000, which allows accurate identification and clear spatial visualization of LPLs in complex biological tissues. A series of lysophosphatidylcholines (LPCs) was detected using positive ion detection mode, and their concentration differences and spatial distributions were clearly visualized in different parts of zebrafish tissue. The method is rapid, simple, and efficient, being a desirable way to understand the spatial distribution of LPLs in biosome.
Collapse
Affiliation(s)
- Kaili He
- Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Guangdong Provincial Key Laboratory of Emergency Test for Dangerous Chemicals, Guangdong Institute of Analysis (China National Analytical Center Guangzhou), Guangdong Academy of Sciences, 100 Xianlie Middle Road, Guangzhou, 510070, China
- Shenyang University of Technology, Shenyang, 110870, China
| | - Chao Chen
- Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Guangdong Provincial Key Laboratory of Emergency Test for Dangerous Chemicals, Guangdong Institute of Analysis (China National Analytical Center Guangzhou), Guangdong Academy of Sciences, 100 Xianlie Middle Road, Guangzhou, 510070, China
| | - Jiewei Deng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 100 Waihuanxi Road, Guangzhou, 510006, China
| | - Ya-Jun Hou
- Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Guangdong Provincial Key Laboratory of Emergency Test for Dangerous Chemicals, Guangdong Institute of Analysis (China National Analytical Center Guangzhou), Guangdong Academy of Sciences, 100 Xianlie Middle Road, Guangzhou, 510070, China
| | - Zhangmin Xiang
- Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Guangdong Provincial Key Laboratory of Emergency Test for Dangerous Chemicals, Guangdong Institute of Analysis (China National Analytical Center Guangzhou), Guangdong Academy of Sciences, 100 Xianlie Middle Road, Guangzhou, 510070, China
| | - Yunyun Yang
- Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Guangdong Provincial Key Laboratory of Emergency Test for Dangerous Chemicals, Guangdong Institute of Analysis (China National Analytical Center Guangzhou), Guangdong Academy of Sciences, 100 Xianlie Middle Road, Guangzhou, 510070, China
| |
Collapse
|
13
|
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: 108] [Impact Index Per Article: 21.6] [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
|
14
|
Zhang W, Huo T, Li A, Wu X, Feng C, Liu J, Jiang H. Identification of neurotoxicity markers induced by realgar exposure in the mouse cerebral cortex using lipidomics. JOURNAL OF HAZARDOUS MATERIALS 2020; 389:121567. [PMID: 32061421 DOI: 10.1016/j.jhazmat.2019.121567] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/12/2019] [Accepted: 10/29/2019] [Indexed: 06/10/2023]
Abstract
Realgar is a traditional Chinese medicine containing arsenic and has neurotoxicity. This study used realgar exposure mice model, neurobehavioral tests, analytical chemistry, molecular biology and nontargeted lipidomics to explore the mechanism of realgar damages the nervous system. The arsenic contained in realgar passed through the BBB and accumulated in the brain. Neurons, synapses and myelin showed abnormal changes in the cerebral cortex. The number of autophagosomes were incresed as well as levels of MDA, Lp-PLA2, and cPLA2 but the CAT level was significant reduced. Finally, the cognition and memory of mice were decreased. Nontargeted lipidomics detected 34 lipid subclasses including 1603 lipid molecules. The levels of the LPC and LPE were significantly increased. Under the condition of variable importance for the projection (VIP)>1 and P < 0.05, only 28 lipid molecules satisfied the criteria. The lipid molecular markers SM (d36:2), PE (18:2/22:6) and PE (36:3) which were filtered by receiver operating characteristic (ROC) curve (AUC>0.8 or AUC<0.2) were used to identify the neurotoxicity induced by realgar. Therefore, realgar induces neurotoxicity through exacerbating oxidative damage and lipid dysfunction. Providing research basis for the clinical diagnosis and treatment of realgar-induced neurotoxicity.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Taoguang Huo
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Aihong Li
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Xinyu Wu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Cong Feng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Jieyu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Hong Jiang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
| |
Collapse
|
15
|
Zhao PF, Wu S, Li Y, Bao G, Pei JY, Wang YW, Ma Q, Sun HJ, Damirin A. LPA receptor1 antagonists as anticancer agents suppress human lung tumours. Eur J Pharmacol 2019; 868:172886. [PMID: 31866407 DOI: 10.1016/j.ejphar.2019.172886] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 12/05/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022]
Abstract
Lysophosphatidic acid (LPA), as a bioactive lipid, plays a variety of physiological and pathological roles via activating six types of G-protein-coupled LPA receptors (LPA1-6). Our preliminary study found that LPA1 is highly expressed in lung cancer tissues compared with paracancerous tissues, but the role of LPA1 in lung carcinoma is unclear. This study aimed to elucidate the association between LPA1 and lung tumour behaviour at the cellular and animal model levels. We found that LPA promoted the migration, proliferation and colony formation of a lung cancer cell line (A549). LPA1 and LPA3 are preferentially expressed in A549 cells, and both Ki16425 (LPA1 and LPA3 antagonist) and ono7300243 (LPA1 antagonist) completely blocked the LPA-induced actions. These results were further verified by experiments of the LPA1/3 overexpression and LPA1 knockdown A549 cells. Furthermore, LPA1 overexpression and knockdown A549 cells were used to assess the in vivo tumour-bearing animal model and the mechanism underlying LPA-induced actions. In the animal model, A549 cell-derived tumour volume was significantly increased by LPA1 overexpression and significantly decreased by LPA1 knockdown respectively, suggesting that LPA1 is a regulator of in vivo tumour formation. Our results also indicated that the LPA1/Gi/MAP kinase/NF-κB pathway is involved in LPA-induced oncogenic actions in A549 cells. Thus, targeting LPA1 may be a novel strategy for treating lung carcinoma.
Collapse
Affiliation(s)
- Peng-Fei Zhao
- School of Life Sciences, Inner Mongolia University, Hohhot, 010070, China
| | - Shuang Wu
- School of Life Sciences, Inner Mongolia University, Hohhot, 010070, China; Department of Internal Medicine, Tianjin Provincial Corps Hospital of Chinese People's Armed Police Forces, Tianjin, 300252, China
| | - Yan Li
- School of Life Sciences, Inner Mongolia University, Hohhot, 010070, China
| | - Gegentuya Bao
- School of Life Sciences, Inner Mongolia University, Hohhot, 010070, China
| | - Jing-Yuan Pei
- School of Life Sciences, Inner Mongolia University, Hohhot, 010070, China
| | - Yue-Wu Wang
- School of Life Sciences, Inner Mongolia University, Hohhot, 010070, China; Research Center for New Drug Screening, Inner Mongolia Medical University, Hohhot, 010010, China
| | - Qing Ma
- School of Life Sciences, Inner Mongolia University, Hohhot, 010070, China
| | - Hong-Ju Sun
- School of Life Sciences, Inner Mongolia University, Hohhot, 010070, China
| | | |
Collapse
|
16
|
Elevated Autotaxin and LPA Levels During Chronic Viral Hepatitis and Hepatocellular Carcinoma Associate with Systemic Immune Activation. Cancers (Basel) 2019; 11:cancers11121867. [PMID: 31769428 PMCID: PMC6966516 DOI: 10.3390/cancers11121867] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022] Open
Abstract
Circulating autotaxin (ATX) is elevated in persons with liver disease, particularly in the setting of chronic hepatitis C virus (HCV) and HCV/HIV infection. It is thought that plasma ATX levels are, in part, attributable to impaired liver clearance that is secondary to fibrotic liver disease. In a discovery data set, we identified plasma ATX to be associated with parameters of systemic immune activation during chronic HCV and HCV/HIV infection. We and others have observed a partial normalization of ATX levels within months of starting interferon-free direct-acting antiviral (DAA) HCV therapy, consistent with a non-fibrotic liver disease contribution to elevated ATX levels, or HCV-mediated hepatocyte activation. Relationships between ATX, lysophosphatidic acid (LPA) and parameters of systemic immune activation will be discussed in the context of HCV infection, age, immune health, liver health, and hepatocellular carcinoma (HCC).
Collapse
|
17
|
Zhao Y, Hasse S, Zhao C, Bourgoin SG. Targeting the autotaxin - Lysophosphatidic acid receptor axis in cardiovascular diseases. Biochem Pharmacol 2019; 164:74-81. [PMID: 30928673 DOI: 10.1016/j.bcp.2019.03.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/26/2019] [Indexed: 02/06/2023]
Abstract
Lysophosphatidic acid (LPA) is a well-characterized bioactive lipid mediator, which is involved in development, physiology, and pathological processes of the cardiovascular system. LPA can be produced both inside cells and in biological fluids. The majority of extracellularLPAis produced locally by the secreted lysophospholipase D, autotaxin (ATX), through its binding to various β integrins or heparin sulfate on cell surface and hydrolyzing various lysophospholipids. LPA initiates cellular signalling pathways upon binding to and activation of its G protein-coupled receptors (LPA1-6). LPA has potent effects on various blood cells and vascular cells involved in the development of cardiovascular diseases such as atherosclerosis and aortic valve sclerosis. LPA signalling drives cell migration and proliferation, cytokine production, thrombosis, fibrosis, as well as angiogenesis. For instance, LPA promotes activation and aggregation of platelets through LPA5, increases expression of adhesion molecules in endothelial cells, and enhances expression of tissue factor in vascular smooth muscle cells. Furthermore, LPA induces differentiation of monocytes into macrophages and stimulates oxidized low-density lipoproteins (oxLDLs) uptake by macrophages to form foam cells during formation of atherosclerotic lesions through LPA1-3. This review summarizes recent findings of the roles played by ATX, LPA and LPA receptors (LPARs) in atherosclerosis and calcific aortic valve disease. Targeting the ATX-LPAR axis may have potential applications for treatment of patients suffering from various cardiovascular diseases.
Collapse
Affiliation(s)
- Yang Zhao
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V4G2, Canada
| | - Stephan Hasse
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V4G2, Canada
| | - Chenqi Zhao
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada
| | - Sylvain G Bourgoin
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V4G2, Canada.
| |
Collapse
|
18
|
Law SH, Chan ML, Marathe GK, Parveen F, Chen CH, Ke LY. An Updated Review of Lysophosphatidylcholine Metabolism in Human Diseases. Int J Mol Sci 2019; 20:ijms20051149. [PMID: 30845751 PMCID: PMC6429061 DOI: 10.3390/ijms20051149] [Citation(s) in RCA: 457] [Impact Index Per Article: 76.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/12/2022] Open
Abstract
Lysophosphatidylcholine (LPC) is increasingly recognized as a key marker/factor positively associated with cardiovascular and neurodegenerative diseases. However, findings from recent clinical lipidomic studies of LPC have been controversial. A key issue is the complexity of the enzymatic cascade involved in LPC metabolism. Here, we address the coordination of these enzymes and the derangement that may disrupt LPC homeostasis, leading to metabolic disorders. LPC is mainly derived from the turnover of phosphatidylcholine (PC) in the circulation by phospholipase A2 (PLA2). In the presence of Acyl-CoA, lysophosphatidylcholine acyltransferase (LPCAT) converts LPC to PC, which rapidly gets recycled by the Lands cycle. However, overexpression or enhanced activity of PLA2 increases the LPC content in modified low-density lipoprotein (LDL) and oxidized LDL, which play significant roles in the development of atherosclerotic plaques and endothelial dysfunction. The intracellular enzyme LPCAT cannot directly remove LPC from circulation. Hydrolysis of LPC by autotaxin, an enzyme with lysophospholipase D activity, generates lysophosphatidic acid, which is highly associated with cancers. Although enzymes with lysophospholipase A1 activity could theoretically degrade LPC into harmless metabolites, they have not been found in the circulation. In conclusion, understanding enzyme kinetics and LPC metabolism may help identify novel therapeutic targets in LPC-associated diseases.
Collapse
Affiliation(s)
- Shi-Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Mei-Lin Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- Division of Thoracic Surgery, Department of Surgery, MacKay Memorial Hospital, MacKay Medical College, Taipei 10449, Taiwan.
| | - Gopal K Marathe
- Department of Studies in Biochemistry, Manasagangothri, University of Mysore, Mysore-570006, India.
| | - Farzana Parveen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chu-Huang Chen
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA.
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|