1
|
Chen S, Hu Z, Tang J, Zhu H, Zheng Y, Xiao J, Xu Y, Wang Y, Luo Y, Mo X, Wu Y, Guo J, Zhang Y, Luo H. High temperature and humidity in the environment disrupt bile acid metabolism, the gut microbiome, and GLP-1 secretion in mice. Commun Biol 2024; 7:465. [PMID: 38632312 PMCID: PMC11024098 DOI: 10.1038/s42003-024-06158-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: 06/23/2023] [Accepted: 04/05/2024] [Indexed: 04/19/2024] Open
Abstract
High temperature and humidity in the environment are known to be associated with discomfort and disease, yet the underlying mechanisms remain unclear. We observed a decrease in plasma glucagon-like peptide-1 levels in response to high-temperature and humidity conditions. Through 16S rRNA gene sequencing, alterations in the gut microbiota composition were identified following exposure to high temperature and humidity conditions. Notably, changes in the gut microbiota have been implicated in bile acid synthesis. Further analysis revealed a decrease in lithocholic acid levels in high-temperature and humidity conditions. Subsequent in vitro experiments demonstrated that lithocholic acid increases glucagon-like peptide-1 secretion in NCI-H716 cells. Proteomic analysis indicated upregulation of farnesoid X receptor expression in the ileum. In vitro experiments revealed that the combination of lithocholic acid with farnesoid X receptor inhibitors resulted in a significant increase in GLP-1 levels compared to lithocholic acid alone. In this study, we elucidate the mechanism by which reduced lithocholic acid suppresses glucagon-like peptide 1 via farnesoid X receptor activation under high-temperature and humidity condition.
Collapse
Affiliation(s)
- Song Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zongren Hu
- Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
| | - Jianbang Tang
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, China
| | | | - Yuhua Zheng
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiedong Xiao
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, China
| | - Yao Wang
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Luo
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoying Mo
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yalan Wu
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianwen Guo
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Immunology Programme, The Life Science Institute, National University of Singapore, Singapore, Singapore.
| | - Huanhuan Luo
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Liang ZQ, Bian Y, Gu JF, Yin G, Sun RL, Liang Y, Wan LL, Yin QH, Wang X, Gao J, Zhao F, Tang DC. Exploring the anti-metastatic effects of Astragalus mongholicus Bunge-Curcuma aromatica Salisb. on colorectal cancer: A network-based metabolomics and pharmacology approach. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154772. [PMID: 37015187 DOI: 10.1016/j.phymed.2023.154772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignancy that can significantly diminish patients' quality of life. Astragalus mongholicus Bunge-Curcuma aromatica Salisb. (AC) is an ancient Chinese medicinal combination used for the treatment of CRC. However, the core ingredients and targets involved in regulating lipid and amino acid metabolism in CRC remain unknown. We aimed to explore the key components and pharmacological mechanisms of AC in the treatment of CRC through a comprehensive analysis of network metabolomics, network pharmacology, molecular docking, and biological methods. METHODS Ultra-performance liquid chromatography/mass spectrometry (MS) was used for quality control. Gas chromatography/MS and liquid chromatography/MS were used to detect metabolites in the feces and serum of CRC mice. A network pharmacology approach and molecular docking were used to explore the potential genes involved in the CRC-target-component network. The effect of AC on tumor immunity was investigated using flow cytometry and polymerase chain reaction. RESULTS AC, high-dose AC, and 5-fluorouracil treatment reduced liver metastasis and tumor mass. Compared with the CRC group, 2 amino acid metabolites and 14 lipid metabolites (LPC, PC, PE) were upregulated and 15 amino acid metabolites and 9 lipid metabolites (TG, PE, PG, 12-HETE) were downregulated. Subsequently, through network analysis, four components and six hub genes were identified for molecular docking. AC can bind to ALDH1B1, ALDH2, CAT, GOT2, NOS3, and ASS1 through beta-Elemene, canavanine, betaine, and chrysanthemaxanthin. AC promoted the responses of M1 macrophages and down-regulated the responses of M2 macrophages, Treg cells, and the gene expression of related factors. CONCLUSION Our research showed that AC effectively inhibited the growth and metastasis of tumors and regulated metabolism and immunity in a CRC mouse model. Thus, AC may be an effective alternative treatment option for CRC.
Collapse
Affiliation(s)
- Zhong Qing Liang
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Yong Bian
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China; Laboratory Animal Center, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Jun Fei Gu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Gang Yin
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Ruo Lan Sun
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Yan Liang
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Lin Lu Wan
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Qi Hang Yin
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Xu Wang
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Jin Gao
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China; School of Acupuncture and Tuina, School of Health and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing 210046, Jiangsu, China
| | - Fan Zhao
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - De Cai Tang
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China.
| |
Collapse
|
3
|
Imamura Y, Makita Y, Masuno K, Oh H. Inhibitory Mechanism of IL-6 Production by Orento in Oral Squamous Cell Carcinoma Cell Line CAL27 Stimulated by Pathogen-Associated Molecular Patterns from Periodontopathogenic Porphyromonas gingivalis. Int J Mol Sci 2022; 24:ijms24010697. [PMID: 36614140 PMCID: PMC9821341 DOI: 10.3390/ijms24010697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/24/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Orento is a traditional Japanese medicinal kampo preparation that is also prescribed in oral care. In oral squamous cell carcinoma cell line CAL27, orento significantly inhibited periodontopathogenic bacterium Porphyromonas gingivalis lipopolysaccharide (LPS) and lipoproteins (PAMP)-stimulated production of interleukin (IL)-6. This suggests that orento negatively regulates PAMP-mediated toll-like receptor (TLR) signaling. Orento significantly suppressed PAMP-stimulated activation of the IL-6 promoter, indicating that orento may suppress the production of IL-6 by PAMP at the transcriptional level. Orento also suppressed TLR-mediated activation of transcription factor nuclear factor-kappa B (NF-kB) that was stimulated by PAMP. This finding indicates that orento may suppress the function and activation of factors involved in TLR signaling, thereby suppressing NF-kB-dependent expression of various genes. Orento suppressed IL-1 receptor-associated kinase (IRAK4), IRAK1, and c-Jun N-terminal kinase (JNK) phosphorylation in PAMP-stimulated CAL27 cells. This result indicates that orento is involved in the initiation of TLR signaling by PAMP and suppresses the downstream signaling pathways of myeloid differentiation primary response gene 88 (MyD88) such as mitogen-activated protein kinase (MAPK) and NF-kB cascades. These findings suggest that orento has an inhibitory effect on the production of inflammatory cytokines.
Collapse
Affiliation(s)
- Yasuhiro Imamura
- Department of Pharmacology, Matsumoto Dental University, Nagano 399-0781, Japan
| | - Yoshimasa Makita
- Department of Chemistry, Osaka Dental University, Osaka 573-1121, Japan
| | - Kazuya Masuno
- Center of Innovation in Dental Education, Osaka Dental University, Osaka 573-1121, Japan
| | - Hourei Oh
- Center of Innovation in Dental Education, Osaka Dental University, Osaka 573-1121, Japan
- Correspondence: ; Tel.: +81-72-864-3171
| |
Collapse
|
4
|
Luo Y, Li J, Huang C, Wang X, Long D, Cao Y. Graphene oxide links alterations of anti-viral signaling pathways with lipid metabolism via suppressing TLR3 in vascular smooth muscle cells. Mol Omics 2022; 18:779-790. [PMID: 35912640 DOI: 10.1039/d2mo00086e] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Vascular smooth muscle cells (VSMCs), the main cells constructing blood vessels, are important in the regulation of the pathophysiology of vascular systems; however, relatively few studies have investigated the influence of nanomaterials (NMs) on VSMCs. In this study, we found that the interaction between graphene oxide and human VSMCs led to the cytotoxicity and morphological changes of cells. Because transcriptomic data suggested that graphene oxide decreased anti-viral signaling pathways via decreasing Toll-like receptor 3 (TLR3), we further verified that graphene oxide decreased interferon induced protein with tetratricopeptide repeats 1 (IFIT1) and the radical S-adenosyl methionine domain containing 2 (RSAD2), and TLR3-downstream genes involved in anti-viral responses. Due to the involvement of RSAD2 in lipid dysfunction, we also verified that graphene oxide disrupted lipid homeostasis and increased adipose triglyceride lipase (ATGL). Adding TLR3 agonist polyinosinic:polycytidylic acid (Poly IC) partially increased TLR3-downstream protein interleukin-8 (IL-8) and some lipid classes, particularly lysophosphatidylcholine (LPC) and lysophosphatidylethanolamine (LPE), in graphene oxide-exposed VSMCs. In mice receiving repeated intravenous injection of graphene oxide, significantly decreased TLR3, IFIT1 and RSAD2 but increased ATGL proteins were observed in aortas. We conclude that graphene oxide altered anti-viral signaling pathways and lipid metabolism via decreasing TLR3 in VSMCs.
Collapse
Affiliation(s)
- Yingmei Luo
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China.
- College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing 210037, China
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, China
| | - Juan Li
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, China
| | - Chaobo Huang
- College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing 210037, China
| | - Xuefeng Wang
- Department of Obstetrics and Gynecology, The third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, 510632, China
| | - Dingxin Long
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Yi Cao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China.
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, China
| |
Collapse
|
5
|
Lysophosphatidylcholine: Potential Target for the Treatment of Chronic Pain. Int J Mol Sci 2022; 23:ijms23158274. [PMID: 35955410 PMCID: PMC9368269 DOI: 10.3390/ijms23158274] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 12/26/2022] Open
Abstract
The bioactive lipid lysophosphatidylcholine (LPC), a major phospholipid component of oxidized low-density lipoprotein (Ox-LDL), originates from the cleavage of phosphatidylcholine by phospholipase A2 (PLA2) and is catabolized to other substances by different enzymatic pathways. LPC exerts pleiotropic effects mediated by its receptors, G protein-coupled signaling receptors, Toll-like receptors, and ion channels to activate several second messengers. Lysophosphatidylcholine (LPC) is increasingly considered a key marker/factor positively in pathological states, especially inflammation and atherosclerosis development. Current studies have indicated that the injury of nervous tissues promotes oxidative stress and lipid peroxidation, as well as excessive accumulation of LPC, enhancing the membrane hyperexcitability to induce chronic pain, which may be recognized as one of the hallmarks of chronic pain. However, findings from lipidomic studies of LPC have been lacking in the context of chronic pain. In this review, we focus in some detail on LPC sources, biochemical pathways, and the signal-transduction system. Moreover, we outline the detection methods of LPC for accurate analysis of each individual LPC species and reveal the pathophysiological implication of LPC in chronic pain, which makes it an interesting target for biomarkers and the development of medicine regarding chronic pain.
Collapse
|
6
|
Diray-Arce J, Angelidou A, Jensen KJ, Conti MG, Kelly RS, Pettengill MA, Liu M, van Haren SD, McCulloch SD, Michelloti G, Idoko O, Kollmann TR, Kampmann B, Steen H, Ozonoff A, Lasky-Su J, Benn CS, Levy O. Bacille Calmette-Guérin vaccine reprograms human neonatal lipid metabolism in vivo and in vitro. Cell Rep 2022; 39:110772. [PMID: 35508141 PMCID: PMC9157458 DOI: 10.1016/j.celrep.2022.110772] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 11/26/2021] [Accepted: 04/11/2022] [Indexed: 12/24/2022] Open
Abstract
Vaccines have generally been developed with limited insight into their molecular impact. While systems vaccinology enables characterization of mechanisms of action, these tools have yet to be applied to infants, who are at high risk of infection and receive the most vaccines. Bacille Calmette-Guérin (BCG) protects infants against disseminated tuberculosis (TB) and TB-unrelated infections via incompletely understood mechanisms. We employ mass-spectrometry-based metabolomics of blood plasma to profile BCG-induced infant responses in Guinea-Bissau in vivo and the US in vitro. BCG-induced lysophosphatidylcholines (LPCs) correlate with both TLR-agonist- and purified protein derivative (PPD, mycobacterial antigen)-induced blood cytokine production in vitro, raising the possibility that LPCs contribute to BCG immunogenicity. Analysis of an independent newborn cohort from The Gambia demonstrates shared vaccine-induced metabolites, such as phospholipids and sphingolipids. BCG-induced changes to the plasma lipidome and LPCs may contribute to its immunogenicity and inform the development of early life vaccines.
Collapse
Affiliation(s)
- Joann Diray-Arce
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Asimenia Angelidou
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Kristoffer Jarlov Jensen
- Research Center for Vitamins and Vaccines (CVIVA), Bandim Health Project, University of Southern Denmark, 2300 Copenhagen, Denmark; Bandim Health Project, Department of Clinical Research, University of Southern Denmark, 1455 Copenhagen K, Denmark; Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark, 2800 Kgs, Lyngby, Denmark
| | - Maria Giulia Conti
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Maternal and Child Health, Sapienza University of Rome, 00185 Rome, Italy
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Matthew A Pettengill
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mark Liu
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Simon D van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Olubukola Idoko
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Tobias R Kollmann
- Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia
| | - Beate Kampmann
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Hanno Steen
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Al Ozonoff
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Christine S Benn
- Research Center for Vitamins and Vaccines (CVIVA), Bandim Health Project, University of Southern Denmark, 2300 Copenhagen, Denmark; Bandim Health Project, Department of Clinical Research, University of Southern Denmark, 1455 Copenhagen K, Denmark; Danish Institute for Advanced Study, University of Southern Denmark, 5230 Odense M, Denmark
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
7
|
Belcher T, Dubois V, Rivera-Millot A, Locht C, Jacob-Dubuisson F. Pathogenicity and virulence of Bordetella pertussis and its adaptation to its strictly human host. Virulence 2021; 12:2608-2632. [PMID: 34590541 PMCID: PMC8489951 DOI: 10.1080/21505594.2021.1980987] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The highly contagious whooping cough agent Bordetella pertussis has evolved as a human-restricted pathogen from a progenitor which also gave rise to Bordetella parapertussis and Bordetella bronchiseptica. While the latter colonizes a broad range of mammals and is able to survive in the environment, B. pertussis has lost its ability to survive outside its host through massive genome decay. Instead, it has become a highly successful human pathogen by the acquisition of tightly regulated virulence factors and evolutionary adaptation of its metabolism to its particular niche. By the deployment of an arsenal of highly sophisticated virulence factors it overcomes many of the innate immune defenses. It also interferes with vaccine-induced adaptive immunity by various mechanisms. Here, we review data from invitro, human and animal models to illustrate the mechanisms of adaptation to the human respiratory tract and provide evidence of ongoing evolutionary adaptation as a highly successful human pathogen.
Collapse
Affiliation(s)
- Thomas Belcher
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Violaine Dubois
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Alex Rivera-Millot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Camille Locht
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Françoise Jacob-Dubuisson
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
8
|
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
|
9
|
Zhao H, He Y. The Inhibitory Effect of Lysophosphatidylcholine on Proangiogenesis of Human CD34 + Cells Derived Endothelial Progenitor Cells. Front Mol Biosci 2021; 8:682367. [PMID: 34179086 PMCID: PMC8223510 DOI: 10.3389/fmolb.2021.682367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022] Open
Abstract
Increasing evidence reveals that lysophosphatidylcholine (LPC) is closely related to endothelial dysfunction. The present study aimed to investigate the mechanism of LPC in inhibiting the proangiogenesis and vascular inflammation of human endothelial progenitor cells (EPCs) derived from CD34+ cells. The early EPCs were derived from CD34+ hematopoietic stem cells whose purity was identified using flow cytometry analysis. The surface markers (CD34, KDR, CD31; VE-cadherin, vWF, eNOS) of EPCs were examined by flow cytometry analysis and immunofluorescence. RT-qPCR was used to detect the mRNA expression of inflammatory cytokines (CCL2, IL-8, CCL4) and genes associated with angiogenesis (VEGF, ANG-1, ANG-2) in early EPCs after treatment of LPC (10 μg/ml) or phosphatidylcholine (PC, 10 μg/ml, control). The angiogenesis of human umbilical vein endothelial cells (HUVECs) incubated with the supernatants of early EPCs was detected by a tube formation assay. The mRNA and protein levels of key factors on the PKC pathway (phosphorylated PKC, TGF-β1) were measured by RT-qPCR and western blot. The localization of PKC-β1 in EPCs was determined by immunofluorescence staining. We found that LPC suppressed the expression of CCL2, CCL4, ANG-1, ANG-2, promoted IL-8 expression and had no significant effects on VEGF expression in EPCs. EPCs promoted the angiogenesis of HUVECs, which was significantly inhibited by LPC treatment. Moreover, LPC was demonstrated to promote the activation of the PKC signaling pathway in EPCs. In conclusion, LPC inhibits proangiogenesis of human endothelial progenitor cells derived from CD34+ hematopoietic stem cells.
Collapse
Affiliation(s)
- Haijun Zhao
- Department of Pain, The First Hospital of Jilin University, Changchun, China
| | - Yanhui He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Akhade AS, Atif SM, Lakshmi BS, Dikshit N, Hughes KT, Qadri A, Subramanian N. Type 1 interferon-dependent repression of NLRC4 and iPLA2 licenses down-regulation of Salmonella flagellin inside macrophages. Proc Natl Acad Sci U S A 2020; 117:29811-29822. [PMID: 33177235 PMCID: PMC7703570 DOI: 10.1073/pnas.2002747117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Inflammasomes have been implicated in the detection and clearance of a variety of bacterial pathogens, but little is known about whether this innate sensing mechanism has any regulatory effect on the expression of stimulatory ligands by the pathogen. During infection with Salmonella and many other pathogens, flagellin is a major activator of NLRC4 inflammasome-mediated macrophage pyroptosis and pathogen eradication. Salmonella switches to a flagellin-low phenotype as infection progresses to avoid this mechanism of clearance by the host. However, the host cues that Salmonella perceives to undergo this switch remain unclear. Here, we report an unexpected role of the NLRC4 inflammasome in promoting expression of its microbial ligand, flagellin, and identify a role for type 1 IFN signaling in switching of Salmonella to a flagellin-low phenotype. Early in infection, activation of NLRC4 by flagellin initiates pyroptosis and concomitant release of lysophospholipids which in turn enhance expression of flagellin by Salmonella thereby amplifying its ability to elicit cell death. TRIF-dependent production of type 1 IFN, however, later represses NLRC4 and the lysophospholipid biosynthetic enzyme iPLA2, causing a decline in intracellular lysophospholipids that results in down-regulation of flagellin expression by Salmonella These findings reveal a previously unrecognized immune-modulating regulatory cross-talk between endosomal TLR signaling and cytosolic NLR activation with significant implications for the establishment of infection with Salmonella.
Collapse
Affiliation(s)
| | - Shaikh M Atif
- Hybridoma Laboratory, National Institute of Immunology, 110067 New Delhi, India
| | | | - Neha Dikshit
- Hybridoma Laboratory, National Institute of Immunology, 110067 New Delhi, India
| | - Kelly T Hughes
- Department of Biology, University of Utah, Salt Lake City, UT 84112
| | - Ayub Qadri
- Hybridoma Laboratory, National Institute of Immunology, 110067 New Delhi, India;
| | - Naeha Subramanian
- Institute for Systems Biology, Seattle, WA 98109;
- Department of Immunology, University of Washington, Seattle, WA 98109
- Department of Global Health, University of Washington, Seattle, WA 98109
| |
Collapse
|