1
|
Qiu H, Liu J, You J, Zhou O, Hao C, Shu Y, Ma D, Zou W, Zhang L, Liu E, Luo Z, Ren L, Geng G, Zou L, Peng D, Fu Z. Inhibition of sphingosine 1-phosphate receptor 3 ameliorates bleomycin-induced pulmonary fibrosis by suppressing macrophage M2 polarization. Genes Dis 2025; 12:101244. [PMID: 40092491 PMCID: PMC11907442 DOI: 10.1016/j.gendis.2024.101244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/11/2024] [Accepted: 01/21/2024] [Indexed: 03/19/2025] Open
Abstract
Pulmonary fibrosis is a devastating lung disease without effective treatment options. Sphingosine-1-phosphate receptor 3 (S1pr3), a receptor for the lipid signaling molecule sphingosine-1-phosphate, has been shown to mediate the development of pulmonary fibrosis, although the underlying mechanism is not fully understood. Here, we found increased expression of S1pr3 in the lung during the process of bleomycin-induced pulmonary fibrosis in mice and specific overexpression of S1pr3 in the infiltrated M2 macrophages. We constructed LysM-Cre + /S1pr3 flox/flox mice, in which S1pr3 was conditionally depleted in myeloid cells, and this depletion protected mice from bleomycin-induced lung injury and fibrosis, with reduced M2 macrophage accumulation in the lung. Increased S1pr3 expression was found in bone marrow-derived macrophages after alternatively activated by IL4 ex vivo, while loss of S1pr3 attenuated IL-4-induced M2 polarization in bone marrow-derived macrophages by repressing the PI3K/Akt-Stat3 signaling pathway. Moreover, the S1pr3 inhibitors CAY10444 and TY52156 exerted protective effects on pulmonary fibrosis in mice. Taken together, our research showed that inhibition of S1pr3 ameliorates bleomycin-induced pulmonary fibrosis by reducing macrophage M2 polarization via the PI3K/Akt-Stat3 signaling pathway, indicating that S1pr3 may be a potential target for pulmonary fibrosis treatment.
Collapse
Affiliation(s)
- Huijun Qiu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Jiang Liu
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jingyi You
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Ou Zhou
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Chang Hao
- Department of Otolaryngology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yi Shu
- Center of Clinical Molecular Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Deyu Ma
- Center of Clinical Molecular Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Wenjing Zou
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Linghuan Zhang
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Enmei Liu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Zhengxiu Luo
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Luo Ren
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Gang Geng
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Lin Zou
- Center of Clinical Molecular Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Clinical Research Unit, Institute of Pediatric Infection, Immunity and Critical Care Medicine, Children's Hospital of Shanghai Jiaotong University Medical School, Shanghai 200062, China
| | - Danyi Peng
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Zhou Fu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| |
Collapse
|
2
|
Jiang ZM, Fang ZY, Yang X, Ji XX, Zhao YY, Lin BY, Weng ZB, Liu EH. Glycyrrhetinic acid ameliorates gastric mucosal injury by modulating gut microbiota and its metabolites via Thbs1/PI3K-Akt/p53 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156745. [PMID: 40305972 DOI: 10.1016/j.phymed.2025.156745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 03/03/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Dysbiosis of the gut microbiota is pivotal in the development of gastric mucosa injury (GMI). Glycyrrhetinic acid (GA) is a bioactive triterpenoid compound abundantly present in licorice roots. Although GA's potential in mitigating GMI is recognized, its precise mechanism remains elusive, particularly concerning the role of gut microbiota. PURPOSE This study aimed to explore the protective effects and mechanisms of GA in preventing HCl/ethanol-induced GMI in rats. RESULTS This study demonstrated the protective effects of GA on gastric mucosa, evidenced by enhanced morphology and structure as revealed through H&E staining. Utilizing fecal microbiota transplantation, GA was found to significantly mitigate oxidative damage, inflammation, and expression of apoptosis-related genes in GMI rats by a gut microbiota-dependent mechanism. 16S rRNA sequencing and metabolomics profiling revealed that GA ameliorated HCl/ethanol-triggered intestinal dysbiosis and imbalances in sphingolipid, arginine, and tryptophan metabolism. By promoting the prevalence of Bifidobacterium longum subsp. infantis (B. infantis) in the gut microbiota, GA improved metabolic disturbances linked to injury. Furthermore, its action mechanism was related to the inhibition of the Thbs1/PI3K-Akt/p53 signaling pathway. CONCLUSION The administration of GA improves GMI by mitigating intestinal dysbiosis and fostering colonization of B. infantis. GA offers therapeutic potential for GMI by modulating the Thbs1/PI3K-Akt/p53 pathway, thus alleviating inflammatory responses associated with gut microbiota imbalance.
Collapse
Affiliation(s)
- Zheng-Meng Jiang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture / School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhi-Yue Fang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture / School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xing Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Xing-Xing Ji
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture / School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuan-Yuan Zhao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture / School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Bin-Yan Lin
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture / School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Ze-Bin Weng
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture / School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - E-Hu Liu
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture / School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
3
|
Rufail ML, Bassi R, Giussani P. Sphingosine-1-Phosphate Metabolic Pathway in Cancer: Implications for Therapeutic Targets. Int J Mol Sci 2025; 26:1056. [PMID: 39940821 PMCID: PMC11817292 DOI: 10.3390/ijms26031056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Cancer biology revolves around understanding how cells undergo uncontrolled proliferation leading to the formation of malignant tumors. Key aspects include self-sufficiency in growth signals, the lack of response to signals of growth inhibition, the evasion of apoptosis, sustained angiogenesis, the evasion of immune response, the capacity to invade and metastasize, and alterations in cellular metabolism. A vast amount of research, which is exponentially growing, over the past few decades highlights the role of sphingolipids in cancer. They act not only as structural membrane components but also as bioactive molecules that regulate cell fate in different physio-pathological conditions. In cancer, sphingolipid metabolism is dysregulated, contributing to tumor progression, metastasis, and drug resistance. In this review, we outline the impact of sphingosine-1-phosphate (S1P) as a key bioactive sphingolipid in cancer. We give an overview of its metabolism summarizing the role of S1P as an intracellular and extracellular mediator through specific plasma membrane receptors in different cancers. We also describe previous findings on how the disruption in the balance between S1P and ceramide (Cer) is common in cancer cells and can contribute to tumorigenesis and resistance to chemotherapy. We finally consider the potential of targeting the metabolic pathways of S1P as well as its receptors and transporters as a promising therapeutic approach in cancer treatments.
Collapse
Affiliation(s)
- Miguel L. Rufail
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Rosaria Bassi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, LITA Segrate, Via Fratelli Cervi, 93, 20054 Segrate, Italy
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, LITA Segrate, Via Fratelli Cervi, 93, 20054 Segrate, Italy
| |
Collapse
|
4
|
Yang L, Yue Y, Wang Z, Jiang Y, Xue Z, Zhang Y. Elucidating the Mechanisms of Acquired Palbociclib Resistance via Comprehensive Metabolomics Profiling. Curr Issues Mol Biol 2025; 47:24. [PMID: 39852139 PMCID: PMC11763656 DOI: 10.3390/cimb47010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/12/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Palbociclib is a cyclin-dependent kinase 4/6 inhibitor and a commonly used antitumor drug. Many cancers are susceptible to palbociclib resistance, however, the underlying metabolism mechanism and extent of resistance to palbociclib are unknown. In this study, LC-MS metabolomics was used to investigate the metabolite changes of colorectal cancer SW620 cells that were resistant to palbociclib. The study indicated that there were 76 metabolite expression differences between SW620 cells with palbociclib resistance and the parental SW620 cells involving amino acids, glutathione, ABC transporters, and so on. MetaboAnalyst 6.0 metabolic pathway analysis showed that arginine synthesis, β-alanine metabolism, and purine metabolism were disrupted. These results may provide potential clues to the metabolism mechanism of drug resistance in cancer cells that are resistant to palbociclib. Our study has the potential to contribute to the study of anti-palbociclib resistance.
Collapse
Affiliation(s)
- Lulu Yang
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, China; (L.Y.); (Y.J.)
| | - Yajun Yue
- General Management Department of Laboratory Base, National Institute of Metrology, Beijing 100029, China;
| | - Zhendong Wang
- Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, China; (Z.W.); (Y.Z.)
| | - You Jiang
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, China; (L.Y.); (Y.J.)
| | - Zhichao Xue
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, China; (L.Y.); (Y.J.)
| | - Yongzhuo Zhang
- Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, China; (Z.W.); (Y.Z.)
| |
Collapse
|
5
|
Yilmaz S, Cizmecioglu O. PI3K Signaling at the Crossroads of Lipid Metabolism and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:139-164. [PMID: 39616584 DOI: 10.1007/5584_2024_832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
The proto-oncogenic PI3K pathway is crucial for the integration of growth factor signaling and metabolic pathways to facilitate the coordination for cell growth. Since transformed cells have the ability to upregulate their anabolic pathways and selectively modulate a subset of metabolites functioning as anti- or pro-tumorigenic signal mediators, the question of how the levels of these metabolites are regulated has also become the center of attention for cancer researchers. Apart from its well-defined roles in glucose metabolism and peptide anabolism, the PI3K pathway appears to be a significant regulator of lipid metabolism and a potentiator of proto-oncogenic bioactive lipid metabolite signaling. In this review, we aim to describe the crosstalk between the PI3K pathway and bioactive lipid species of the three main lipid classes.
Collapse
Affiliation(s)
- Sevval Yilmaz
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Onur Cizmecioglu
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey.
| |
Collapse
|
6
|
Ye Y, Huang L, Wang K, Sun Y, Zhou Z, Deng T, Liu Y, Wang R, Wu R, Yao C. Transplantation of engineered endothelial progenitor cells with H19 overexpression promotes arterial reendothelialization and inhibits neointimal hyperplasia. J Tissue Eng 2025; 16:20417314251315959. [PMID: 39974657 PMCID: PMC11837068 DOI: 10.1177/20417314251315959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 01/12/2025] [Indexed: 02/21/2025] Open
Abstract
Endothelial injury is a key factor initiating in-stent restenosis (ISR) following peripheral artery stent implantation. Genetically modified endothelial progenitor cells (EPCs) can promote reendothelialization of injured arteries and inhibit neointimal hyperplasia. However, the role of engineered EPCs overexpressing lncRNA H19 in these processes remains unclear. We constructed EPCs overexpressing lncRNA H19 and investigated their effects and mechanisms in promoting reendothelialization and inhibiting neointimal hyperplasia both in vitro and in vivo. Compared to the normal control group, ISR patients exhibited a significant reduction in circulating EPCs. Engineered EPCs overexpressing lncRNA H19 promoted reendothelialization and inhibited neointimal hyperplasia in injured arteries. Exogenous overexpression of lncRNA H19 significantly upregulated the endothelial repair-related gene S1PR3 in EPCs, while the opposite was also observed. Additionally, engineered EPCs overexpressing S1PR3 promoted reendothelialization and inhibited neointimal hyperplasia in injured arteries. S1PR3 overexpression enhanced EPCs proliferation, migration, and tube formation in vitro; these effects were lost with S1PR3 inhibition. Binding sites for H3K27 acetylation were identified on the S1PR3 promoter. Mechanistically, we found that lncRNA H19 directly interacted with HDAC2, a known H3K27ac deacetylase, disrupting its binding to H3K27 acetylation. Our findings suggest that lncRNA H19 positively regulates S1PR3 expression by disrupting HDAC2 / H3K27ac binding, thereby promoting reendothelialization of injured arteries and inhibiting neointimal hyperplasia.
Collapse
Affiliation(s)
- Yanchen Ye
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lin Huang
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University
- Institute of Interventional Radiology, Sun Yat-sen University, Zhuhai, China
| | - Kangjie Wang
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunhao Sun
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihao Zhou
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tang Deng
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunyan Liu
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rui Wang
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ridong Wu
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chen Yao
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
7
|
Dai R, Zhuo H, Chen Y, Zhang K, Dong Y, Chen C, Wang W. Retracted article: Mechanism of isosorbide dinitrate combined with exercise training rehabilitation to mobilize endothelial progenitor cells in patients with coronary heart disease. Bioengineered 2024; 15:2000258. [PMID: 34738489 PMCID: PMC10826619 DOI: 10.1080/21655979.2021.2000258] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/29/2022] Open
Abstract
Ruozhu Dai, Huilin Zhuo, Yangchun Chen, Kelian Zhang, Yongda Dong, Chengbo Chen and Wei Wang. Mechanism of isosorbide dinitrate combined with exercise training rehabilitation to mobilize endothelial progenitor cells in patients with coronary heart disease. Bioengineered. 2021 Nov. doi: 10.1080/21655979.2021.2000258.Since publication, significant concerns have been raised about the compliance with ethical policies for human research and the integrity of the data reported in the article.When approached for an explanation, the authors provided some original data but were not able to provide all the necessary supporting information. As verifying the validity of published work is core to the scholarly record's integrity, we are retracting the article. All authors listed in this publication have been informed.We have been informed in our decision-making by our editorial policies and the COPE guidelines.The retracted article will remain online to maintain the scholarly record, but it will be digitally watermarked on each page as 'Retracted.'
Collapse
Affiliation(s)
- Ruozhu Dai
- Department of Cardiology, Quanzhou First Hospital, Quanzhou, Fujian Province,
China
| | - Huilin Zhuo
- Department of Cardiology, Quanzhou First Hospital, Quanzhou, Fujian Province,
China
| | - Yangchun Chen
- Shanghai Lung Hospital Affiliated to Tongji University, Shanghai, China
| | - Kelian Zhang
- Department of Cardiology, Quanzhou First Hospital, Quanzhou, Fujian Province,
China
| | - Yongda Dong
- Department of Cardiology, Quanzhou First Hospital, Quanzhou, Fujian Province,
China
| | - Chengbo Chen
- Department of Cardiology, Quanzhou First Hospital, Quanzhou, Fujian Province,
China
| | - Wei Wang
- Department of Cardiology, Quanzhou First Hospital, Quanzhou, Fujian Province,
China
| |
Collapse
|
8
|
Limbu KR, Chhetri RB, Kim S, Shrestha J, Oh YS, Baek DJ, Park EY. Targeting sphingosine 1-phosphate and sphingosine kinases in pancreatic cancer: mechanisms and therapeutic potential. Cancer Cell Int 2024; 24:353. [PMID: 39462385 PMCID: PMC11514880 DOI: 10.1186/s12935-024-03535-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Pancreatic cancer is known to be the most lethal cancer. Fewer new treatments are being developed for pancreatic cancer as compared to other cancers. The bioactive lipid S1P, which is mainly regulated by sphingosine kinase 1 (SK1) and sphingosine kinase 2 (SK2) enzymes, plays significant roles in pancreatic cancer initiation and exacerbation. S1P controls many signaling pathways to modulate the progression of pancreatic cancer through the G-coupled receptor S1PR1-5. Several papers reporting amelioration of pancreatic cancer via modulation of S1P levels or downstream signaling pathways have previously been published. In this paper, for the first time, we have reviewed the results of previous studies to understand how S1P and its receptors contribute to the development of pancreatic cancer, and whether S1P can be a therapeutic target. In addition, we have also reviewed papers dealing with the effects of SK1 and SK2, which are kinases that regulate the level of S1P, on the pathogenesis of pancreatic cancer. We have also listed available drugs that particularly focus on S1P, S1PRs, SK1, and SK2 for the treatment of pancreatic cancer. Through this review, we would like to suggest that the SK/S1P/S1PR signaling system can be an important target for treating pancreatic cancer, where a new treatment target is desperately warranted.
Collapse
Affiliation(s)
- Khem Raj Limbu
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | | | - Subin Kim
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | - Jitendra Shrestha
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, Seongnam, 13135, South Korea
| | - Dong Jae Baek
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| | - Eun-Young Park
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| |
Collapse
|
9
|
Bhat OM, Mir RA, Nehvi IB, Wani NA, Dar AH, Zargar MA. Emerging role of sphingolipids and extracellular vesicles in development and therapeutics of cardiovascular diseases. IJC HEART & VASCULATURE 2024; 53:101469. [PMID: 39139609 PMCID: PMC11320467 DOI: 10.1016/j.ijcha.2024.101469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024]
Abstract
Sphingolipids are eighteen carbon alcohol lipids synthesized from non-sphingolipid precursors in the endoplasmic reticulum (ER). The sphingolipids serve as precursors for a vast range of moieties found in our cells that play a critical role in various cellular processes, including cell division, senescence, migration, differentiation, apoptosis, pyroptosis, autophagy, nutrition intake, metabolism, and protein synthesis. In CVDs, different subclasses of sphingolipids and other derived molecules such as sphingomyelin (SM), ceramides (CERs), and sphingosine-1-phosphate (S1P) are directly related to diabetic cardiomyopathy, dilated cardiomyopathy, myocarditis, ischemic heart disease (IHD), hypertension, and atherogenesis. Several genome-wide association studies showed an association between genetic variations in sphingolipid pathway genes and the risk of CVDs. The sphingolipid pathway plays an important role in the biogenesis and secretion of exosomes. Small extracellular vesicles (sEVs)/ exosomes have recently been found as possible indicators for the onset of CVDs, linking various cellular signaling pathways that contribute to the disease progression. Important features of EVs like biocompatibility, and crossing of biological barriers can improve the pharmacokinetics of drugs and will be exploited to develop next-generation drug delivery systems. In this review, we have comprehensively discussed the role of sphingolipids, and sphingolipid metabolites in the development of CVDs. In addition, concise deliberations were laid to discuss the role of sEVs/exosomes in regulating the pathophysiological processes of CVDs and the exosomes as therapeutic targets.
Collapse
Affiliation(s)
- Owais Mohmad Bhat
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - Rakeeb Ahmad Mir
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | | | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - Abid Hamid Dar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - M Afzal Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| |
Collapse
|
10
|
Poteryaeva ON, Usynin IF. Molecular mechanisms of the regulatory action of high-density lipoproteins on the endothelial function. BIOMEDITSINSKAIA KHIMIIA 2024; 70:206-217. [PMID: 39239895 DOI: 10.18097/pbmc20247004206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Endothelial dysfunction underlies the pathogenesis of many diseases, primarily cardiovascular diseases. Epidemiological studies have shown an inverse dependence between the plasma level of high-density lipoproteins (HDL) and cardiovascular diseases. The results of experimental studies indicate that the antiatherogenic effect of HDL is associated not only with their participation in the reverse transport of excess cholesterol, but also with their regulatory effect on the functions of cells of various organs and tissues, including endothelial cells. The purpose of this review is to consider recent data on the participation of plasma receptors and related intracellular signaling pathways in the mechanism of protective effect of HDL on endothelial cell functions. Understanding the mechanisms of cell function regulation under the influence of HDL is an important step for the development of new ways of pharmacological correction of impaired endothelial functions and creation of effective endothelial protection drugs.
Collapse
Affiliation(s)
- O N Poteryaeva
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| | - I F Usynin
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| |
Collapse
|
11
|
Suarez-Castillejo C, Calvo N, Preda L, Córdova Díaz R, Toledo-Pons N, Martínez J, Pons J, Vives-Borràs M, Pericàs P, Ramón L, Iglesias A, Cànaves-Gómez L, Valera Felices JL, Morell-García D, Núñez B, Sauleda J, Sala-Llinàs E, Alonso-Fernández A. Cardiopulmonary Complications after Pulmonary Embolism in COVID-19. Int J Mol Sci 2024; 25:7270. [PMID: 39000378 PMCID: PMC11242326 DOI: 10.3390/ijms25137270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/14/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024] Open
Abstract
Although pulmonary embolism (PE) is a frequent complication in COVID-19, its consequences remain unknown. We performed pulmonary function tests, echocardiography and computed tomography pulmonary angiography and identified blood biomarkers in a cohort of consecutive hospitalized COVID-19 patients with pneumonia to describe and compare medium-term outcomes according to the presence of PE, as well as to explore their potential predictors. A total of 141 patients (56 with PE) were followed up during a median of 6 months. Post-COVID-19 radiological lung abnormalities (PCRLA) and impaired diffusing capacity for carbon monoxide (DLCOc) were found in 55.2% and 67.6% cases, respectively. A total of 7.3% had PE, and 6.7% presented an intermediate-high probability of pulmonary hypertension. No significant difference was found between PE and non-PE patients. Univariate analysis showed that age > 65, some clinical severity factors, surfactant protein-D, baseline C-reactive protein, and both peak red cell distribution width and Interleukin (IL)-10 were associated with DLCOc < 80%. A score for PCRLA prediction including age > 65, minimum lymphocyte count, and IL-1β concentration on admission was constructed with excellent overall performance. In conclusion, reduced DLCOc and PCRLA were common in COVID-19 patients after hospital discharge, but PE did not increase the risk. A PCRLA predictive score was developed, which needs further validation.
Collapse
Affiliation(s)
- Carla Suarez-Castillejo
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Néstor Calvo
- Servicio de Radiodiagnóstico, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Luminita Preda
- Servicio de Radiodiagnóstico, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Rocío Córdova Díaz
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Nuria Toledo-Pons
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Joaquín Martínez
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Jaume Pons
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Cardiología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Miquel Vives-Borràs
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Cardiología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
| | - Pere Pericàs
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Cardiología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Luisa Ramón
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Amanda Iglesias
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura Cànaves-Gómez
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Jose Luis Valera Felices
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Morell-García
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Análisis Clínicos, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Belén Núñez
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Jaume Sauleda
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ernest Sala-Llinàs
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alberto Alonso-Fernández
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
12
|
Wang L, Zhang X, Ma C, Wu N. 1-Phosphate receptor agonists: A promising therapeutic avenue for ischemia-reperfusion injury management. Int Immunopharmacol 2024; 131:111835. [PMID: 38508097 DOI: 10.1016/j.intimp.2024.111835] [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: 02/01/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024]
Abstract
Ischemia-reperfusion injury (IRI) - a complex pathological condition occurring when blood supply is abruptly restored to ischemic tissues, leading to further tissue damage - poses a significant clinical challenge. Sphingosine-1-phosphate receptors (S1PRs), a specialized set of G-protein-coupled receptors comprising five subtypes (S1PR1 to S1PR5), are prominently present in various cell membranes, including those of lymphocytes, cardiac myocytes, and endothelial cells. Increasing evidence highlights the potential of targeting S1PRs for IRI therapeutic intervention. Notably, preconditioning and postconditioning strategies involving S1PR agonists like FTY720 have demonstrated efficacy in mitigating IRI. As the synthesis of a diverse array of S1PR agonists continues, with FTY720 being a prime example, the body of experimental evidence advocating for their role in IRI treatment is expanding. Despite this progress, comprehensive reviews delineating the therapeutic landscape of S1PR agonists in IRI remain limited. This review aspires to meticulously elucidate the protective roles and mechanisms of S1PR agonists in preventing and managing IRI affecting various organs, including the heart, kidney, liver, lungs, intestines, and brain, to foster novel pharmacological approaches in clinical settings.
Collapse
Affiliation(s)
- Linyuan Wang
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China; The Central Laboratory of The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaowen Zhang
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Nan Wu
- The Central Laboratory of The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
13
|
Wille A, Weske S, von Wnuck Lipinski K, Wollnitzke P, Schröder NH, Thomas N, Nowak MK, Deister-Jonas J, Behr B, Keul P, Levkau B. Sphingosine-1-phosphate promotes osteogenesis by stimulating osteoblast growth and neovascularization in a vascular endothelial growth factor-dependent manner. J Bone Miner Res 2024; 39:357-372. [PMID: 38477738 PMCID: PMC11240155 DOI: 10.1093/jbmr/zjae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 03/14/2024]
Abstract
Sphingosine-1-phosphate (S1P) plays multiple roles in bone metabolism and regeneration. Here, we have identified a novel S1P-regulated osteoanabolic mechanism functionally connecting osteoblasts (OBs) to the highly specialized bone vasculature. We demonstrate that S1P/S1PR3 signaling in OBs stimulates vascular endothelial growth factor a (VEGFa) expression and secretion to promote bone growth in an autocrine and boost osteogenic H-type differentiation of bone marrow endothelial cells in a paracrine manner. VEGFa-neutralizing antibodies and VEGF receptor inhibition by axitinib abrogated OB growth in vitro and bone formation in male C57BL/6J in vivo following S1P stimulation and S1P lyase inhibition, respectively. Pharmacological S1PR3 inhibition and genetic S1PR3 deficiency suppressed VEGFa production, OB growth in vitro, and inhibited H-type angiogenesis and bone growth in male mice in vivo. Together with previous work on the osteoanabolic functions of S1PR2 and S1PR3, our data suggest that S1P-dependent bone regeneration employs several nonredundant positive feedback loops between OBs and the bone vasculature. The identification of this yet unappreciated aspect of osteoanabolic S1P signaling may have implications for regular bone homeostasis as well as diseases where the bone microvasculature is affected such as age-related osteopenia and posttraumatic bone regeneration.
Collapse
Affiliation(s)
- Annalena Wille
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Sarah Weske
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Karin von Wnuck Lipinski
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Philipp Wollnitzke
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Nathalie H Schröder
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Nadine Thomas
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Melissa K Nowak
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Jennifer Deister-Jonas
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Björn Behr
- Department of Plastic Surgery, University Hospital BG Bergmannsheil, 44789 Bochum, Germany
| | - Petra Keul
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
14
|
Polenz CK, Scipione CA, Hyduk SJ, Althagafi MG, Ibrahim HM, Cybulsky MI. Plasma S1P Orchestrates the Reverse Transendothelial Migration of Aortic Intimal Myeloid Cells in Mice. Arterioscler Thromb Vasc Biol 2024; 44:883-897. [PMID: 38328936 DOI: 10.1161/atvbaha.123.320227] [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: 09/29/2023] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Myeloid cells (MCs) reside in the aortic intima at regions predisposed to atherosclerosis. Systemic inflammation triggers reverse transendothelial migration (RTM) of intimal MCs into the arterial blood, which orchestrates a protective immune response that clears intracellular pathogens from the arterial intima. Molecular pathways that regulate RTM remain poorly understood. S1P (sphingosine-1-phosphate) is a lipid mediator that regulates immune cell trafficking by signaling via 5 G-protein-coupled receptors (S1PRs [S1P receptors]). We investigated the role of S1P in the RTM of aortic intimal MCs. METHODS Intravenous injection of lipopolysaccharide was used to model a systemic inflammatory stimulus that triggers RTM. CD11c+ intimal MCs in the lesser curvature of the ascending aortic arch were enumerated by en face confocal microscopy. Local gene expression was evaluated by transcriptomic analysis of microdissected intimal cells. RESULTS In wild-type C57BL/6 mice, lipopolysaccharide induced intimal cell expression of S1pr1, S1pr3, and Sphk1 (a kinase responsible for S1P production). Pharmacological modulation of multiple S1PRs blocked lipopolysaccharide-induced RTM and modulation of S1PR1 and S1PR3 reduced RTM in an additive manner. Cre-mediated deletion of S1pr1 in MCs blocked lipopolysaccharide-induced RTM, confirming a role for myeloid-specific S1PR1 signaling. Global or hematopoietic deficiency of Sphk1 reduced plasma S1P levels, the abundance of CD11c+ MCs in the aortic intima, and blunted lipopolysaccharide-induced RTM. In contrast, plasma S1P levels, the abundance of intimal MCs, and lipopolysaccharide-induced RTM were rescued in Sphk1-/- mice transplanted with Sphk1+/+ or mixed Sphk1+/+ and Sphk1-/- bone marrow. Stimulation with lipopolysaccharide increased endothelial permeability and intimal MC exposure to circulating factors such as S1P. CONCLUSIONS Functional and expression studies support a novel role for S1P signaling in the regulation of lipopolysaccharide-induced RTM and the homeostatic maintenance of aortic intimal MCs. Our data provide insight into how circulating plasma mediators help orchestrate intimal MC dynamics.
Collapse
Affiliation(s)
- Chanele K Polenz
- Departments of Laboratory Medicine and Pathobiology (C.K.P., C.A.S., M.G.A., H.M.I., M.I.C.), University of Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
| | - Corey A Scipione
- Departments of Laboratory Medicine and Pathobiology (C.K.P., C.A.S., M.G.A., H.M.I., M.I.C.), University of Toronto, Canada
- Immunology (C.A.S., M.I.C.), University of Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
| | - Sharon J Hyduk
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
| | - Marwan G Althagafi
- Departments of Laboratory Medicine and Pathobiology (C.K.P., C.A.S., M.G.A., H.M.I., M.I.C.), University of Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
| | - Hisham M Ibrahim
- Departments of Laboratory Medicine and Pathobiology (C.K.P., C.A.S., M.G.A., H.M.I., M.I.C.), University of Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
| | - Myron I Cybulsky
- Departments of Laboratory Medicine and Pathobiology (C.K.P., C.A.S., M.G.A., H.M.I., M.I.C.), University of Toronto, Canada
- Immunology (C.A.S., M.I.C.), University of Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Canada (C.K.P., C.A.S., S.J.H., M.G.A., H.M.I., M.I.C.)
- Peter Munk Cardiac Centre, University Health Network, Toronto, Canada (M.I.C.)
| |
Collapse
|
15
|
Alkafaas SS, Elsalahaty MI, Ismail DF, Radwan MA, Elkafas SS, Loutfy SA, Elshazli RM, Baazaoui N, Ahmed AE, Hafez W, Diab M, Sakran M, El-Saadony MT, El-Tarabily KA, Kamal HK, Hessien M. The emerging roles of sphingosine 1-phosphate and SphK1 in cancer resistance: a promising therapeutic target. Cancer Cell Int 2024; 24:89. [PMID: 38419070 PMCID: PMC10903003 DOI: 10.1186/s12935-024-03221-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 01/09/2024] [Indexed: 03/02/2024] Open
Abstract
Cancer chemoresistance is a problematic dilemma that significantly restrains numerous cancer management protocols. It can promote cancer recurrence, spreading of cancer, and finally, mortality. Accordingly, enhancing the responsiveness of cancer cells towards chemotherapies could be a vital approach to overcoming cancer chemoresistance. Tumour cells express a high level of sphingosine kinase-1 (SphK1), which acts as a protooncogenic factor and is responsible for the synthesis of sphingosine-1 phosphate (S1P). S1P is released through a Human ATP-binding cassette (ABC) transporter to interact with other phosphosphingolipids components in the interstitial fluid in the tumor microenvironment (TME), provoking communication, progression, invasion, and tumor metastasis. Also, S1P is associated with several impacts, including anti-apoptotic behavior, metastasis, mesenchymal transition (EMT), angiogenesis, and chemotherapy resistance. Recent reports addressed high levels of S1P in several carcinomas, including ovarian, prostate, colorectal, breast, and HCC. Therefore, targeting the S1P/SphK signaling pathway is an emerging therapeutic approach to efficiently attenuate chemoresistance. In this review, we comprehensively discussed S1P functions, metabolism, transport, and signaling. Also, through a bioinformatic framework, we pointed out the alterations of SphK1 gene expression within different cancers with their impact on patient survival, and we demonstrated the protein-protein network of SphK1, elaborating its sparse roles. Furthermore, we made emphasis on different machineries of cancer resistance and the tight link with S1P. We evaluated all publicly available SphK1 inhibitors and their inhibition activity using molecular docking and how SphK1 inhibitors reduce the production of S1P and might reduce chemoresistance, an approach that might be vital in the course of cancer treatment and prognosis.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Mohamed I Elsalahaty
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Doha F Ismail
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mustafa Ali Radwan
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt
- Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, 197101, Russia
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Nanotechnology Research Center, British University, Cairo, Egypt
| | - Rami M Elshazli
- Biochemistry and Molecular Genetics Unit, Department of Basic Sciences, Faculty of Physical Therapy, Horus University-Egypt, New Damietta, 34517, Egypt
| | - Narjes Baazaoui
- Biology Department, College of Sciences and Arts Muhayil Assir, King Khalid University, Abha 61421, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Wael Hafez
- NMC Royal Hospital, 16th Street, 35233, Khalifa, Abu Dhabi, United Arab Emirates
- Medical Research Division, Department of Internal Medicine, The National Research Centre, Cairo 11511, Egypt
| | - Mohanad Diab
- Burjeel Hospital Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Mohamed Sakran
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Hani K Kamal
- Anatomy and Histology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
16
|
Morvaridzadeh M, Zoubdane N, Heshmati J, Alami M, Berrougui H, Khalil A. High-Density Lipoprotein Metabolism and Function in Cardiovascular Diseases: What about Aging and Diet Effects? Nutrients 2024; 16:653. [PMID: 38474781 DOI: 10.3390/nu16050653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Cardiovascular diseases (CVDs) have become the leading global cause of mortality, prompting a heightened focus on identifying precise indicators for their assessment and treatment. In this perspective, the plasma levels of HDL have emerged as a pivotal focus, given the demonstrable correlation between plasma levels and cardiovascular events, rendering them a noteworthy biomarker. However, it is crucial to acknowledge that HDLs, while intricate, are not presently a direct therapeutic target, necessitating a more nuanced understanding of their dynamic remodeling throughout their life cycle. HDLs exhibit several anti-atherosclerotic properties that define their functionality. This functionality of HDLs, which is independent of their concentration, may be impaired in certain risk factors for CVD. Moreover, because HDLs are dynamic parameters, in which HDL particles present different atheroprotective properties, it remains difficult to interpret the association between HDL level and CVD risk. Besides the antioxidant and anti-inflammatory activities of HDLs, their capacity to mediate cholesterol efflux, a key metric of HDL functionality, represents the main anti-atherosclerotic property of HDL. In this review, we will discuss the HDL components and HDL structure that may affect their functionality and we will review the mechanism by which HDL mediates cholesterol efflux. We will give a brief examination of the effects of aging and diet on HDL structure and function.
Collapse
Affiliation(s)
- Mojgan Morvaridzadeh
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Nada Zoubdane
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Javad Heshmati
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Mehdi Alami
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Hicham Berrougui
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Abdelouahed Khalil
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| |
Collapse
|
17
|
He Y, Li Y, Zhang Y, Chen L, Luo J, Bi L, Liu L, Wang X, Lv M. Transplantation of miR-193b-3p-Transfected BMSCs Improves Neurological Impairment after Traumatic Brain Injury through S1PR3-Mediated Regulation of the PI3K/AKT/mTOR Signaling Pathway. Crit Rev Eukaryot Gene Expr 2024; 34:1-16. [PMID: 39072405 DOI: 10.1615/critreveukaryotgeneexpr.2024053225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The aim of the present study was to explore the molecular mechanisms by which miR-193b-3p-trans-fected bone marrow mesenchymal stem cells (BMSCs) transplantation improves neurological impairment after traumatic brain injury (TBI) through sphingosine-1-phosphate receptor 3 (S1PR3)-mediated regulation of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway at the cellular and animal levels. BMSCs were transfected with miR-193b-3p. A TBI cell model was established by oxygen-glucose deprivation (OGD)-induced HT22 cells, and a TBI animal model was established by controlled cortical impact (CCI). Cell apoptosis was detected by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL), and cell activity was detected by a cell counting kit 8 (CCK-8) assay. Western blot analysis and quantitative real-time polymerase chain reaction (qRT-PCR) were used to detect the expression of related proteins and genes. In this study, transfection of miR-193b-3p into BMSCs significantly enhanced BMSCs proliferation and differentiation. Transfection of miR-193b-3p reduced the levels of the interleukin-6 (IL-6), IL-1β, and tumor necrosis factor-alpha (TNF-α) inflammatory factors in cells and mouse models, and it inhibited neuronal apoptosis, which alleviated OGD-induced HT22 cell damage and neural function damage in TBI mice. Downstream experiments showed that miR-193b-3p targeting negatively regulated the expression of S1PR3, promoted the activation of the PI3K/AKT/mTOR signaling pathway, and inhibited the levels of apoptosis and inflammatory factors, which subsequently improved OGD-induced neuronal cell damage and nerve function damage in TBI mice. However, S1PR3 overexpression or inhibition of the PI3K/AKT/mTOR signaling pathway using the IN-2 inhibitor weakened the protective effect of miR-193b-3p-transfected BMSCs on HT22 cells. Transplantation of miR-193b-3p-transfected BMSCs inhibits neurological injury and improves the progression of TBI in mice through S1PR3-mediated regulation of the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Yinghong He
- Rehabilitation Medicine, Qujing No. 1 Hospital, Qujing 655000, Yunnan, China
| | - Yuanmou Li
- Rehabilitation Medicine, Qujing No. 1 Hospital, Qujing 655000, Yunnan, China
| | - Yan Zhang
- Rehabilitation Medicine, Qujing No. 1 Hospital, Qujing 655000, Yunnan, China
| | - Lixia Chen
- Rehabilitation Medicine, Qujing No. 1 Hospital, Qujing 655000, Yunnan, China
| | - Juan Luo
- Rehabilitation Medicine, Qujing No. 1 Hospital, Qujing 655000, Yunnan, China
| | - Liqiao Bi
- Rehabilitation Medicine, Qujing No. 1 Hospital, Qujing 655000, Yunnan, China
| | - Limei Liu
- Rehabilitation Medicine, Shizong County People's Hospital, Shizong 655700, Yunnan, China
| | - Xuelian Wang
- Rehabilitation Medicine, Qujing No. 1 Hospital, Qujing 655000, Yunnan, China
| | | |
Collapse
|
18
|
Yang Q, Tang D, Gan C, Bai M, Song X, Jiang W, Li Q, Chen Y, Zhang A, Wang M. Novel variants in CRB2 targeting the malfunction of slit diaphragm related to focal segmental glomerulosclerosis. Pediatr Nephrol 2024; 39:149-165. [PMID: 37452832 DOI: 10.1007/s00467-023-06087-6] [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: 04/22/2023] [Revised: 06/01/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Focal segmental glomerulosclerosis (FSGS) is a leading cause of steroid-resistant nephrotic syndrome (SRNS) that predominantly affects the podocytes. While mutations in genes causing pediatric SRNS have enhanced our understanding of FSGS, the disease's etiology remains complex and poorly understood. METHODS Whole exome sequencing (WES) was performed on a 9-year-old girl with SRNS associated with FSGS (SRNS-FSGS). We analyzed the expression of CRB2, slit diaphragm (SD)-associated proteins, and sphingosine 1-phosphate receptor 1 (S1PR1) in the proband and CRB2 knock-down podocytes. RESULTS In this study, we identified two novel compound heterozygous mutations in the Crumbs homolog 2 (CRB2) gene (c.2905delinsGCCACCTCGCGCTGGCTG, p.T969Afs*179 and c.3268C > G, p.R1090G) in a family with early-onset SRNS-FSGS. Our findings demonstrate that these CRB2 abnormalities were the underlying cause of SRNS-FSGS. CRB2 defects led to the dysfunction of podocyte SD-related proteins, including podocin, nephrin, and zonula occludens-1 (ZO-1), by reducing the phosphorylation level of S1PR1. Interestingly, the podocytic cytoskeleton remained unaffected, as demonstrated by normal expression and localization of synaptopodin. Our study also revealed a secondary decrease in CRB2 expression in idiopathic FSGS patients, indicating that CRB2 mutations may cause FSGS through a previously unknown mechanism involving SD-related proteins. CONCLUSIONS Overall, our findings shed new light on the pathogenesis of SRNS-FSGS and revealed that the novel pathogenic mutations in CRB2 contribute to the development of FSGS through a previously unknown mechanism involving SD-related proteins. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Qing Yang
- Pediatric Research Institute, Department of Nephrology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dan Tang
- Department of Pediatrics, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, 621000, Sichuan, China
| | - Chun Gan
- Pediatric Research Institute, Department of Nephrology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Mi Bai
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Xiaomei Song
- Pediatric Research Institute, Department of Nephrology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Wei Jiang
- Pediatric Research Institute, Department of Nephrology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Qiu Li
- Pediatric Research Institute, Department of Nephrology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yaxi Chen
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| | - Aihua Zhang
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Mo Wang
- Pediatric Research Institute, Department of Nephrology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
19
|
Wang W, Zhao Y, Zhu G. The role of sphingosine-1-phosphate in the development and progression of Parkinson's disease. Front Cell Neurosci 2023; 17:1288437. [PMID: 38179204 PMCID: PMC10764561 DOI: 10.3389/fncel.2023.1288437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Parkinson's disease (PD) could be viewed as a proteinopathy caused by changes in lipids, whereby modifications in lipid metabolism may lead to protein alterations, such as the accumulation of alpha-synuclein (α-syn), ultimately resulting in neurodegeneration. Although the loss of dopaminergic neurons in the substantia nigra is the major clinical manifestation of PD, the etiology of it is largely unknown. Increasing evidence has highlighted the important role of lipids in the pathophysiology of PD. Sphingosine-1-phosphate (S1P), a signaling lipid, has been suggested to have a potential association with the advancement and worsening of PD. Therefore, better understanding the mechanisms and regulatory proteins is of high interest. Most interestingly, S1P appears to be an important target to offers a new strategy for the diagnosis and treatment of PD. In this review, we first introduce the basic situation of S1P structure, function and regulation, with a special focus on the several pathways. We then briefly describe the regulation of S1P signaling pathway on cells and make a special focused on the cell growth, proliferation and apoptosis, etc. Finally, we discuss the function of S1P as potential therapeutic target to improve the clinical symptoms of PD, and even prevent the progression of the PD. In the context of PD, the functions of S1P modulators have been extensively elucidated. In conclusion, S1P modulators represent a novel and promising therapeutic principle and therapeutic method for PD. However, more research is required before these drugs can be considered as a standard treatment option for PD.
Collapse
Affiliation(s)
- Wang Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guoxue Zhu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
20
|
Yuan Z, Tian Y, Zhang C, Wang M, Xie J, Wang C, Huang J. Integration of systematic review, lipidomics with experiment verification reveals abnormal sphingolipids facilitate diabetic retinopathy by inducing oxidative stress on RMECs. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159382. [PMID: 37659619 DOI: 10.1016/j.bbalip.2023.159382] [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: 05/05/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023]
Abstract
OBJECTIVE This study aims to explore the potential biomarkers in the development of diabetes mellitus (DM) into diabetic retinopathy (DR). METHODS Systematic review of diabetic metabolomics was used to screen the differential metabolites and related pathways during the development of DM. Non-targeted lipidomics of rat plasma was performed to explore the differential metabolites in the development of DM into DR in vivo. To verify the effects of differential metabolites in inducing retinal microvascular endothelial cells (RMECs) injury by increasing oxidative stress, high glucose medium containing differential metabolites was used to induce rat RMECs injury and cell viability, malondialdehyde (MDA) contents, superoxide dismutase (SOD) activities, reactive oxygen species (ROS) levels and mitochondrial membrane potential (MMP) were evaluated in vitro. Network pharmacology was performed to explore the potential mechanism of differential metabolites in inducing DR. RESULTS Through the systematic review, 148 differential metabolites were obtained and the sphingolipid metabolic pathway attracted our attention. Plasma non-targeted lipidomics found that sphingolipids were accompanied by the development of DM into DR. In vitro experiments showed sphinganine and sphingosine-1-phosphate aggravated rat RMECs injury induced by high glucose, further increased MDA and ROS levels, and further decreased SOD activities and MMP. Network pharmacology revealed sphinganine and sphingosine-1-phosphate may induce DR by regulating the AGE-RAGE and HIF-1 signaling pathways. CONCLUSIONS Integrated systematic review, lipidomics and experiment verification reveal that abnormal sphingolipid metabolism facilitates DR by inducing oxidative stress on RMECs. Our study could provide the experimental basis for finding potential biomarkers for the diagnosis and treatment of DR.
Collapse
Affiliation(s)
- Zhenshuang Yuan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yue Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Cong Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingshuang Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jiaqi Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Can Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Jianmei Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
21
|
Xiao S, Peng K, Li C, Long Y, Yu Q. The role of sphingosine-1-phosphate in autophagy and related disorders. Cell Death Discov 2023; 9:380. [PMID: 37852968 PMCID: PMC10584985 DOI: 10.1038/s41420-023-01681-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/03/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023] Open
Abstract
S1P, also referred to as sphingosine-1-phosphate, is a lipid molecule with bioactive properties involved in numerous cellular processes such as cell growth, movement, programmed cell death, self-degradation, cell specialization, aging, and immune system reactions. Autophagy is a meticulously controlled mechanism in which cells repurpose their elements to maintain cellular balance. There are five stages in autophagy: initiation, nucleation, elongation and maturation, fusion, and degradation. New research has provided insight into the complex connection between S1P and autophagy, uncovering their interaction in both normal and abnormal circumstances. Gaining knowledge about the regulatory mechanism of S1P signaling on autophagy can offer a valuable understanding of its function in well-being and illness, potentially leading to innovative therapeutic concepts for diverse ailments. Hence, this review analyzes the essential stages in mammalian autophagy, with a specific emphasis on recent research exploring the control of each stage by S1P. Additionally, it sheds light on the roles of S1P-induced autophagy in various disorders.
Collapse
Affiliation(s)
- Siqi Xiao
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Kaixin Peng
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Congxin Li
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Yuanyuan Long
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Qin Yu
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China.
| |
Collapse
|
22
|
Du J, Wu W, Zhu B, Tao W, Liu L, Cheng X, Zhao M, Wu J, Li Y, Pei K. Recent advances in regulating lipid metabolism to prevent coronary heart disease. Chem Phys Lipids 2023; 255:105325. [PMID: 37414117 DOI: 10.1016/j.chemphyslip.2023.105325] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/01/2023] [Accepted: 07/01/2023] [Indexed: 07/08/2023]
Abstract
The pathogenesis of coronary heart disease is a highly complex process, with lipid metabolism disorders being closely linked to its development. Therefore, this paper analyzes the various factors that influence lipid metabolism, including obesity, genes, intestinal microflora, and ferroptosis, through a comprehensive review of basic and clinical studies. Additionally, this paper delves deeply into the pathways and patterns of coronary heart disease. Based on these findings, it proposes various intervention pathways and therapeutic methods, such as the regulation of lipoprotein enzymes, lipid metabolites, and lipoprotein regulatory factors, as well as the modulation of intestinal microflora and the inhibition of ferroptosis. Ultimately, this paper aims to offer new ideas for the prevention and treatment of coronary heart disease.
Collapse
Affiliation(s)
- Jingchun Du
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Wu
- Key laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Boran Zhu
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weiwei Tao
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lina Liu
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaolan Cheng
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Min Zhao
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Yunlun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Ke Pei
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
23
|
Stewart CM, Bo Y, Fu K, Chan M, Kozak R, Apperley KYP, Laroche G, Daniel R, Beauchemin AM, Kobinger G, Kobasa D, Côté M. Sphingosine Kinases Promote Ebola Virus Infection and Can Be Targeted to Inhibit Filoviruses, Coronaviruses, and Arenaviruses Using Late Endocytic Trafficking to Enter Cells. ACS Infect Dis 2023; 9:1064-1077. [PMID: 37053583 DOI: 10.1021/acsinfecdis.2c00416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Entry of enveloped viruses in host cells requires the fusion of viral and host cell membranes, a process that is facilitated by viral fusion proteins protruding from the viral envelope. These viral fusion proteins need to be triggered by host factors, and for some viruses, this event occurs inside endosomes and/or lysosomes. Consequently, these 'late-penetrating viruses' must be internalized and delivered to entry-conducive intracellular vesicles. Because endocytosis and vesicular trafficking are tightly regulated cellular processes, late-penetrating viruses also depend on specific host proteins for efficient delivery to the site of fusion, suggesting that these could be targeted for antiviral therapy. In this study, we investigated a role for sphingosine kinases (SKs) in viral entry and found that chemical inhibition of sphingosine kinase 1 (SK1) and/or SK2 and knockdown of SK1/2 inhibited entry of Ebola virus (EBOV) into host cells. Mechanistically, inhibition of SK1/2 prevented EBOV from reaching late-endosomes and lysosomes that contain the EBOV receptor, Niemann Pick C1 (NPC1). Furthermore, we present evidence that suggests that the trafficking defect caused by SK1/2 inhibition occurs independently of sphingosine-1-phosphate (S1P) signaling through cell-surface S1P receptors. Lastly, we found that chemical inhibition of SK1/2 prevents entry of other late-penetrating viruses, including arenaviruses and coronaviruses, and inhibits infection by replication-competent EBOV and SARS-CoV-2 in Huh7.5 cells. In sum, our results highlight an important role played by SK1/2 in endocytic trafficking, which can be targeted to inhibit entry of late-penetrating viruses and could serve as a starting point for the development of broad-spectrum antiviral therapeutics.
Collapse
Affiliation(s)
- Corina M Stewart
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Yuxia Bo
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Kathy Fu
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Mable Chan
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
- Department of Infectious Diseases and Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Robert Kozak
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| | - Kim Yang-Ping Apperley
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Geneviève Laroche
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Redaet Daniel
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - André M Beauchemin
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Gary Kobinger
- Galveston National Laboratory, Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas 77550, United States
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
- Department of Infectious Diseases and Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
24
|
Frost K, Naylor AJ, McGettrick HM. The Ying and Yang of Sphingosine-1-Phosphate Signalling within the Bone. Int J Mol Sci 2023; 24:6935. [PMID: 37108099 PMCID: PMC10139073 DOI: 10.3390/ijms24086935] [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: 02/06/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Bone remodelling is a highly active and dynamic process that involves the tight regulation of osteoblasts, osteoclasts, and their progenitors to allow for a balance of bone resorption and formation to be maintained. Ageing and inflammation are risk factors for the dysregulation of bone remodelling. Once the balance between bone formation and resorption is lost, bone mass becomes compromised, resulting in disorders such as osteoporosis and Paget's disease. Key molecules in the sphingosine-1-phosphate signalling pathway have been identified for their role in regulating bone remodelling, in addition to its more recognised role in inflammatory responses. This review discusses the accumulating evidence for the different, and, in certain circumstances, opposing, roles of S1P in bone homeostasis and disease, including osteoporosis, Paget's disease, and inflammatory bone loss. Specifically, we describe the current, often conflicting, evidence surrounding S1P function in osteoblasts, osteoclasts, and their precursors in health and disease, concluding that S1P may be an effective biomarker of bone disease and also an attractive therapeutic target for disease.
Collapse
Affiliation(s)
| | - Amy J. Naylor
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | | |
Collapse
|
25
|
Liu L, Liang Z, Ma S, Li L, Liu X. Radioprotective countermeasures for radiation injury (Review). Mol Med Rep 2023; 27:66. [PMID: 36799170 PMCID: PMC9926870 DOI: 10.3892/mmr.2023.12953] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 01/10/2023] [Indexed: 02/09/2023] Open
Abstract
A series of physiological and pathological changes occur after radiotherapy and accidental exposure to ionizing radiation (IR). These changes cause serious damage to human tissues and can lead to death. Radioprotective countermeasures are radioprotective agents that prevent and reduce IR injury or have therapeutic effects. Based on a good understanding of radiobiology, a number of protective agents have achieved positive results in early clinical trials. The present review grouped known radioprotective agents according to biochemical categories and potential clinical use, and reviewed radiation countermeasures, i.e., radioprotectors, radiation mitigators and radiotherapeutic agents, with an emphasis on their current status and research progress. The aim of the present review is to facilitate the selection and application of suitable radioprotectors for clinicians and researchers, to prevent or reduce IR injury.
Collapse
Affiliation(s)
- Lianchang Liu
- National Health Commission Key Laboratory of Radiobiology, School of Public Health of Jilin University, Jilin, Changchun 130021, P.R. China,School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,Department of Intervention, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Zhenzhen Liang
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Shumei Ma
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou, Zhejiang 325035, P.R. China
| | - Lan Li
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou, Zhejiang 325035, P.R. China,Correspondence to: Professor Lan Li, School of Public Health and Management, Wenzhou Medical University, 1 North Zhongxin Road, Chashan, Wenzhou, Zhejiang 325035, P.R. China, E-mail:
| | - Xiaodong Liu
- National Health Commission Key Laboratory of Radiobiology, School of Public Health of Jilin University, Jilin, Changchun 130021, P.R. China,School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou, Zhejiang 325035, P.R. China,Professor Xiaodong Liu, National Health Commission Key Laboratory of Radiobiology, School of Public Health of Jilin University, 1163 Xinmin Road, Changchun, Jilin 130021, P.R. China, E-mail:
| |
Collapse
|
26
|
Choi DH, Liu HW, Jung YH, Ahn J, Kim JA, Oh D, Jeong Y, Kim M, Yoon H, Kang B, Hong E, Song E, Chung S. Analyzing angiogenesis on a chip using deep learning-based image processing. LAB ON A CHIP 2023; 23:475-484. [PMID: 36688448 DOI: 10.1039/d2lc00983h] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Angiogenesis, the formation of new blood vessels from existing vessels, has been associated with more than 70 diseases. Although numerous studies have established angiogenesis models, only a few indicators can be used to analyze angiogenic structures. In the present study, we developed an image-processing pipeline based on deep learning to analyze and quantify angiogenesis. We utilized several image-processing algorithms to quantify angiogenesis, including a deep learning-based cell nuclear segmentation algorithm and image skeletonization. This method could quantify and measure changes in blood vessels in response to biochemical gradients using 16 indicators, including length, width, number, and nuclear distribution. Moreover, this procedure is highly efficient for the three-dimensional quantitative analysis of angiogenesis and can be applied to diverse angiogenesis investigations.
Collapse
Affiliation(s)
- Dong-Hee Choi
- School of Mechanical Engineering, Korea University, Seoul, 02841, Korea.
| | - Hui-Wen Liu
- School of Mechanical Engineering, Korea University, Seoul, 02841, Korea.
| | - Yong Hun Jung
- School of Mechanical Engineering, Korea University, Seoul, 02841, Korea.
| | - Jinchul Ahn
- School of Mechanical Engineering, Korea University, Seoul, 02841, Korea.
| | - Jin-A Kim
- School of Mechanical Engineering, Korea University, Seoul, 02841, Korea.
| | - Dongwoo Oh
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Korea
| | - Yeju Jeong
- School of Mechanical Engineering, Korea University, Seoul, 02841, Korea.
| | - Minseop Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Korea
| | - Hongjin Yoon
- School of Mechanical Engineering, Korea University, Seoul, 02841, Korea.
| | - Byengkyu Kang
- School of Mechanical Engineering, Korea University, Seoul, 02841, Korea.
| | - Eunsol Hong
- School of Mechanical Engineering, Korea University, Seoul, 02841, Korea.
| | | | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul, 02841, Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Korea
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| |
Collapse
|
27
|
Levesque MV, Hla T. Signal Transduction and Gene Regulation in the Endothelium. Cold Spring Harb Perspect Med 2023; 13:a041153. [PMID: 35667710 PMCID: PMC9722983 DOI: 10.1101/cshperspect.a041153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Extracellular signals act on G-protein-coupled receptors (GPCRs) to regulate homeostasis and adapt to stress. This involves rapid intracellular post-translational responses and long-lasting gene-expression changes that ultimately determine cellular phenotype and fate changes. The lipid mediator sphingosine 1-phosphate (S1P) and its receptors (S1PRs) are examples of well-studied GPCR signaling axis essential for vascular development, homeostasis, and diseases. The biochemical cascades involved in rapid S1P signaling are well understood. However, gene-expression regulation by S1PRs are less understood. In this review, we focus our attention to how S1PRs regulate nuclear chromatin changes and gene transcription to modulate vascular and lymphatic endothelial phenotypic changes during embryonic development and adult homeostasis. Because S1PR-targeted drugs approved for use in the treatment of autoimmune diseases cause substantial vascular-related adverse events, these findings are critical not only for general understanding of stimulus-evoked gene regulation in the vascular endothelium, but also for therapeutic development of drugs for autoimmune and perhaps vascular diseases.
Collapse
Affiliation(s)
- Michel V Levesque
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
28
|
iTRAQ-Based Proteomic Analysis of APP Transgenic Mouse Urine Exosomes. Int J Mol Sci 2022; 24:ijms24010672. [PMID: 36614115 PMCID: PMC9820663 DOI: 10.3390/ijms24010672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is a common dementia disease in the elderly. To get a better understanding of the pathophysiology, we performed a proteomic analysis of the urine exosomes (U-exo) in AD model mice (J20). The polymer precipitation method was used to isolate U-exo from the urine of 3-month-old J20 and wild-type (WT) mice. Neuron-derived exosome (N-exo) was isolated from U-exo by immunoprecipitation. iTRAQ-based MALDI TOF MS/MS was used for proteomic analysis. The results showed that compared to WT, the levels of 61 and 92 proteins were increased in the J20 U-exo and N-exo, respectively. Gene ontology enrichment analysis demonstrated that the sphingolipid catabolic process, ceramide catabolic process, membrane lipid catabolic process, Aβ clearance, and Aβ metabolic process were highly enriched in U-exo and N-exo. Among these, Asah1 was shown to be the key protein in lipid metabolism, and clusterin, ApoE, neprilysin, and ACE were related to Aβ metabolism and clearance. Furthermore, protein-protein interaction analysis identified four protein complexes where clusterin and ApoE participated as partner proteins. Thus, J20 U-exo and N-exo contain proteins related to lipid- and Aβ-metabolism in the early stages of AD, providing a new insight into the underlying pathological mechanism of early AD.
Collapse
|
29
|
miR-196a-5p Correlates with Chronic Atrophic Gastritis Progression to Gastric Cancer and Induces Malignant Biological Behaviors of Gastric Cancer Cells by Targeting ACER2. Mol Biotechnol 2022:10.1007/s12033-022-00589-8. [PMID: 36513872 DOI: 10.1007/s12033-022-00589-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/19/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND As the prognosis of early gastric cancer (EGC) is significantly better than that of advanced gastric cancer (AGC), the development of biomarkers to monitor the progression of chronic atrophic gastritis (CAG) to gastric cancer (GC) is essential. METHODS Stomach tissue miRNA and mRNA sequences from patients with chronic non-atrophic gastritis (CNAG), CAG, precancerous lesions of gastric cancer (PLGC), and GC were analyzed. A publicly available GC-related miRNA microarray dataset was obtained from the Gene Expression Omnibus database. Spearman's correlation and differential gene analyses, and clinical validation were used to identify novel miRNAs correlating with CAG progression to GC. miRNA targets were predicted using weighted gene co-expression analysis and databases. A dual-luciferase reporter assay was performed to check for direct interaction between miR-196a-5p and ACER2. The CCK-8 and wound healing assays, and flow cytometry were performed to evaluate cell proliferation, migration, and apoptosis. RESULTS miR-196a-5p was correlated with CAG progression to GC. Overexpression of miR-196a-5p promoted GC cell proliferation and migration and inhibited apoptosis, whereas suppression of miR-196a-5p exerted the opposite effect. Based on the prediction and luciferase assays, ACER2 was identified as the target of miR-196a-5p. ACER2 was downregulated in GC cell lines. Knockdown of ACER2 increased GC cell proliferation rates and migration ability and inhibited apoptosis, while ACER2 overexpression led to the opposite effect. CONCLUSIONS miR-196a-5p correlated with CAG progression to GC and induced malignant biological behaviors of GC cells by targeting ACER2, providing a novel monitoring biomarker and target for GC prevention.
Collapse
|
30
|
Wu J, Luo Y, Shen Y, Hu Y, Zhu F, Wu J, Liu Y. Integrated Metabonomics and Network Pharmacology to Reveal the Action Mechanism Effect of Shaoyao Decoction on Ulcerative Colitis. Drug Des Devel Ther 2022; 16:3739-3776. [PMID: 36324421 PMCID: PMC9620839 DOI: 10.2147/dddt.s375281] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background Traditional Chinese medicine (TCM) has the advantage of multi-component and multi-target, which becomes a hot spot in the treatment of numerous diseases. Shaoyao decoction (SYD) is a TCM prescription, which is mainly used to treat damp-heat dysentery clinically, with small side effects and low cost. However, its mechanism remains elusive. The purpose of this study is to explore the mechanism of SYD in the treatment of mice with ulcerative colitis (UC) induced by dextran sulfate sodium (DSS) through metabolomics and network pharmacology, and verify through molecular docking and immunohistochemistry, so as to provide a scientific basis for the role of SYD in the treatment of UC. Materials and Methods Firstly, DSS-induced UC models were established and then untargeted metabolomics analysis of feces, livers, serum and urine was performed to determine biomarkers and metabolic pathways closely related to the role of SYD. Besides, network pharmacology was applied to screen the active components and UC-related targets, which was verified by molecular docking. Finally, metabonomics and network pharmacology were combined to draw the metabolite-pathway-target network and verified by immunohistochemistry. Results Metabolomics results showed that a total of 61 differential metabolites were discovered in SYD-treated UC with 3 main metabolic pathways containing glycerophospholipid metabolism, sphingolipid metabolism and biosynthesis of unsaturated fatty acids, as well as 8 core targets involving STAT3, IL1B, IL6, IL2, AKT1, IL4, ICAM1 and CCND1. Molecular docking demonstrated that the first five targets had strong affinity with quercetin, wogonin, kaempferol and baicalein. Combined with metabolomics and network pharmacology, sphingolipid signaling pathway, PI3K/AKT-mTOR signaling pathway and S1P3 pathway were identified as the main pathways. Conclusion SYD can effectively ameliorate various symptoms and alleviate intestinal mucosal damage and metabolic disorder in DSS induced UC mice. Its effect is mainly related to sphingolipid metabolism, PI3K/AKT-mTOR signaling pathway and S1P3 pathway.
Collapse
Affiliation(s)
- Jin Wu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Yiting Luo
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Yan Shen
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, People’s Republic of China
| | - Yuyao Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, People’s Republic of China
| | - Fangyuan Zhu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Jiaqian Wu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Yingchao Liu
- Academic Affairs Office, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China,Correspondence: Yingchao Liu, Academic Affairs Office, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China, Email
| |
Collapse
|
31
|
Raza Y, Atallah J, Luberto C. Advancements on the Multifaceted Roles of Sphingolipids in Hematological Malignancies. Int J Mol Sci 2022; 23:12745. [PMID: 36361536 PMCID: PMC9654982 DOI: 10.3390/ijms232112745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/07/2022] [Accepted: 10/17/2022] [Indexed: 09/19/2023] Open
Abstract
Dysregulation of sphingolipid metabolism plays a complex role in hematological malignancies, beginning with the first historical link between sphingolipids and apoptosis discovered in HL-60 leukemic cells. Numerous manuscripts have reviewed the field including the early discoveries that jumpstarted the studies. Many studies discussed here support a role for sphingolipids, such as ceramide, in combinatorial therapeutic regimens to enhance anti-leukemic effects and reduce resistance to standard therapies. Additionally, inhibitors of specific nodes of the sphingolipid pathway, such as sphingosine kinase inhibitors, significantly reduce leukemic cell survival in various types of leukemias. Acid ceramidase inhibitors have also shown promising results in acute myeloid leukemia. As the field moves rapidly, here we aim to expand the body of literature discussed in previously published reviews by focusing on advances reported in the latter part of the last decade.
Collapse
Affiliation(s)
- Yasharah Raza
- Department of Pharmacological Sciences, Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, NY 11794, USA
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Jane Atallah
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY 11794, USA
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Chiara Luberto
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY 11794, USA
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
32
|
Lai Y, Tian Y, You X, Du J, Huang J. Effects of sphingolipid metabolism disorders on endothelial cells. Lipids Health Dis 2022; 21:101. [PMID: 36229882 PMCID: PMC9563846 DOI: 10.1186/s12944-022-01701-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/05/2022] [Indexed: 11/10/2022] Open
Abstract
Many cardiovascular disorders, including atherosclerosis, hypertension, coronary heart disease, diabetes, etc., are characterized by endothelial cell dysfunction. Endothelial cell function is closely related to sphingolipid metabolism, and normal sphingolipid metabolism is critical for maintaining endothelial cell homeostasis. Sphingolipid metabolites or key enzymes in abnormal situation, including sphingosine, ceramide (Cer), sphingosine-1-phosphate (S1P), serine, sphingosine kinase (SPHK), ceramide kinase (Cerk), sphingosine-1-phosphate lyase (S1PL) etc., may have a protective or damaging effect on the function of endothelial cells. This review summarizes the effects of sphingolipid metabolites and key enzymes disordering in sphingolipid metabolism on endothelial cells, offering some insights into further research on the pathogenesis of cardiovascular diseases and corresponding therapeutic targets.
Collapse
Affiliation(s)
- Yali Lai
- School of Traditional Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Tian
- School of Traditional Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xintong You
- School of Traditional Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiangnan Du
- School of Traditional Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jianmei Huang
- School of Traditional Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
33
|
Zheng Z, Lei C, Liu H, Jiang M, Zhou Z, Zhao Y, Yu CY, Wei H. A ROS-Responsive Liposomal Composite Hydrogel Integrating Improved Mitochondrial Function and Pro-Angiogenesis for Efficient Treatment of Myocardial Infarction. Adv Healthc Mater 2022; 11:e2200990. [PMID: 35848825 DOI: 10.1002/adhm.202200990] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/07/2022] [Indexed: 01/27/2023]
Abstract
Mitochondrial dysfunction of cardiomyocytes (CMs) has been identified as a significant pathogenesis of early myocardial infarction (MI). However, only a few agents or strategies have been developed to improve mitochondrial dysfunction for the effective MI treatment. Herein, a reactive oxygen species (ROS)-responsive PAMB-G-TK/4-arm-PEG-SG hydrogel is developed for localized drug-loaded liposome delivery. Notably, the liposomes contain both elamipretide (SS-31) and sphingosine-1-phosphate (S1P), where SS-31 acts as an inhibitor of mitochondrial oxidative damage and S1P as a signaling molecule for activating angiogenesis. Liposome-encapsulated PAMB-G-TK/4-arm-PEG-SG hydrogels demonstrate myocardium-like mechanical strength and electrical conductivity, and ROS-sensitive release of SS-31 and S1P-loaded liposomes. Further liposomal release of SS-31, which can target cytochrome c in the mitochondrial inner membrane of damaged CMs, inhibits pathological ROS production, improving mitochondrial dysfunction. Meanwhile, S1P released from the liposome induces endothelial cell angiogenesis by activating the S1PR1/PI3K/Akt pathway. In a rat MI model, the resulting liposomal composite hydrogel improves cardiac function by scavenging excess ROS, improving mitochondrial dysfunction, and promoting angiogenesis. This study reports for the first time a liposomal composite hydrogel that can directly target mitochondria of damaged CMs for a feedback-regulated release of encapsulated liposomes to consume the overproduced pathological ROS for improved CM activity and enhanced MI treatment.
Collapse
Affiliation(s)
- Zhi Zheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Cai Lei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Hongbing Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Mingchao Jiang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Zongtao Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Yuqi Zhao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| |
Collapse
|
34
|
Foote CA, Soares RN, Ramirez-Perez FI, Ghiarone T, Aroor A, Manrique-Acevedo C, Padilla J, Martinez-Lemus LA. Endothelial Glycocalyx. Compr Physiol 2022; 12:3781-3811. [PMID: 35997082 PMCID: PMC10214841 DOI: 10.1002/cphy.c210029] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The glycocalyx is a polysaccharide structure that protrudes from the body of a cell. It is primarily conformed of glycoproteins and proteoglycans, which provide communication, electrostatic charge, ionic buffering, permeability, and mechanosensation-mechanotransduction capabilities to cells. In blood vessels, the endothelial glycocalyx that projects into the vascular lumen separates the vascular wall from the circulating blood. Such a physical location allows a number of its components, including sialic acid, glypican-1, heparan sulfate, and hyaluronan, to participate in the mechanosensation-mechanotransduction of blood flow-dependent shear stress, which results in the synthesis of nitric oxide and flow-mediated vasodilation. The endothelial glycocalyx also participates in the regulation of vascular permeability and the modulation of inflammatory responses, including the processes of leukocyte rolling and extravasation. Its structural architecture and negative charge work to prevent macromolecules greater than approximately 70 kDa and cationic molecules from binding and flowing out of the vasculature. This also prevents the extravasation of pathogens such as bacteria and virus, as well as that of tumor cells. Due to its constant exposure to shear and circulating enzymes such as neuraminidase, heparanase, hyaluronidase, and matrix metalloproteinases, the endothelial glycocalyx is in a continuous process of degradation and renovation. A balance favoring degradation is associated with a variety of pathologies including atherosclerosis, hypertension, vascular aging, metastatic cancer, and diabetic vasculopathies. Consequently, ongoing research efforts are focused on deciphering the mechanisms that promote glycocalyx degradation or limit its syntheses, as well as on therapeutic approaches to improve glycocalyx integrity with the goal of reducing vascular disease. © 2022 American Physiological Society. Compr Physiol 12: 1-31, 2022.
Collapse
Affiliation(s)
- Christopher A. Foote
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Rogerio N. Soares
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | | | - Thaysa Ghiarone
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Annayya Aroor
- Department of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Luis A. Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
35
|
Chen K, Wang Z, Liu C, Yang X, Jiang J. Sphingosine-1-phosphate Attenuates Endoplasmic Reticulum Stress-induced Cardiomyocyte Apoptosis Through Sphingosine-1-phosphate Receptor 1. Arch Med Res 2022; 53:562-573. [PMID: 35999060 DOI: 10.1016/j.arcmed.2022.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 07/03/2022] [Accepted: 08/09/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND Endoplasmic reticulum stress (ER stress) is involved in the development and progression of various forms of heart disease and may lead to myocardial apoptosis. Sphingosine-1-phosphate (S1P) possesses cardioprotective properties, including anti-apoptosis. However, little is known about the link between S1P and ER stress-induced myocardial apoptosis. This study investigated the regulatory role of S1P in ER stress-induced apoptosis in cardiomyocytes. METHODS ER stress and myocardial apoptosis were induced by transverse aortic constriction (TAC) or tunicamycin in mice, which were then treated with 2-acetyl-5-tetrahydroxybutyl imidazole (THI) or S1P. AC16 cells were treated with tunicamycin or thapsigargin, or pretreated with S1P, sphingosine-1-phosphate receptor (S1PR) subtype antagonists, S1PR1 agonist, and PI3K and MEK inhibitors. Cardiac function, the level of S1P in plasma and heart, ER stress markers, cell viability, and apoptosis were detected. RESULTS S1P reduced the expression of ER stress-related molecules and ER stress-induced myocardial apoptosis in mice subjected to TAC or an injection of tunicamycin. Furthermore, in AC16 cells exposed to thapsigargin or tunicamycin, S1P decreased the expression of ER stress-related molecules, promoting cell viability and survival. Nevertheless, the S1PR1 antagonist abrogated the protection of S1P. Subsequently, in TAC S1PR1 heterozygous (S1PR1+/-) mice, S1P had no effect on ER stress and apoptosis in cardiomyocytes. Notably, in vitro, the impact of anti-ER stress-induced myocardial apoptosis by the S1PR1 agonist was reversed by PI3K and MEK inhibitors. CONCLUSION This study is the first to demonstrate that S1P relieves ER stress-induced myocardial apoptosis via S1PR1/AKT and S1PR1/ERK1/2, which are potential therapeutic targets for heart disease.
Collapse
Affiliation(s)
- Kengquan Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhongqin Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Yang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangang Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
36
|
Fan X, Chen H, Xu C, Wang Y, Yin P, Li M, Tang Z, Jiang F, Wei W, Song J, Li G, Zhong D. Inhibiting Sphingosine 1-Phosphate Receptor Subtype 3 Attenuates Brain Damage During Ischemia-Reperfusion Injury by Regulating nNOS/NO and Oxidative Stress. Front Neurosci 2022; 16:838621. [PMID: 35242008 PMCID: PMC8886115 DOI: 10.3389/fnins.2022.838621] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/17/2022] [Indexed: 11/19/2022] Open
Abstract
Background Ischemic stroke (IS) is a common disease endangering human life and health. Cerebral ischemia triggers a series of complex harmful events, including excitotoxicity, inflammation and cell death, as well as increased nitric oxide production through the activation of nitric oxide synthase (NOS). Oxidative stress plays a major role in cerebral ischemia and reperfusion. Sphingosine 1-phosphate receptor subtype 3 (S1PR3), a member of S1P’s G protein-coupled receptors S1PR1-S1PR5, is involved in a variety of biological effects in the body, and its role in regulating oxidative stress during cerebral ischemia and reperfusion is still unclear. Methods Transient middle cerebral artery occlusion (tMCAO) mice were selected as the brain ischemia–reperfusion (I/R) injury model. Male C57/BL6 mice were treated with or without a selective S1PR3 inhibition after tMCAO, and changes in infarct volume, Nissl staining, hematoxylin-eosin (H&E) staining and NOS protein, nitric oxide (NO), superoxide dismutase (SOD), and malondialdehyde (MDA) content after tMCAO were observed. Results In the cerebral ischemia–reperfusion model, inhibition of S1PR3 improved the infarct volume and neuronal damage in mice after tMCAO. Similarly, inhibition of S1PR3 can reduce the expression of NO synthase subtype neuronal NOS (nNOS) and reduce the production of NO after cerebral ischemia. After cerebral ischemia and reperfusion, the oxidative stress response was enhanced, and after the administration of the S1PR3 inhibitor, the SOD content increased and the MDA content decreased, indicating that S1PR3 plays an important role in regulating oxidative stress response. Conclusion Inhibiting S1PR3 attenuates brain damage during I/R injury by regulating nNOS/NO and oxidative stress, which provides a potential new therapeutic target and mechanism for the clinical treatment of IS.
Collapse
Affiliation(s)
- Xuehui Fan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongping Chen
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chen Xu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingju Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pengqi Yin
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meng Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhanbin Tang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fangchao Jiang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wan Wei
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jihe Song
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Di Zhong
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
37
|
Wang ZL, Sun HH, Liu HY, Ji QX, Niu YT, Ma P, Hao G, Zhang JX, Yuan YY, Chai XL, Han J, Wang W. The water extracts of Euonymus alatus (Thunb.) Siebold attenuate diabetic retinopathy by mediating angiogenesis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114782. [PMID: 34728316 DOI: 10.1016/j.jep.2021.114782] [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: 07/15/2021] [Revised: 10/13/2021] [Accepted: 10/23/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Euonymus alatus (Thunb.) Siebold (family Celastraceae) is a deciduous woody shrub that is recorded in ShenNong BenCaoJing. It has been widely used for diabetes in traditional Chinese medicine. AIM OF THE STUDY This study aimed to identify the most effective extract of Euonymus alatus (EA) against high glucose-induced endothelial cells in vitro, evaluate its pharmacological effect on retinopathy in diabetic mice and explore its underlying mechanism by RNA sequencing. METHODS Retinal vascular endothelial cells (RF/6A) were treated with normal glucose (5.5 mmol/L glucose), high glucose (25 mmol/L glucose) or high glucose plus methanol extracts of EA (MEA), ethyl acetate extracts of EA (EEA) or water extracts of EA (WEA). The cytotoxicity and cell viability were determined by Cell Counting Kit-8 (CCK-8) assay. Cell migration was examined using the Transwell assay, and tube formation ability was measured using the Matrigel assay. Then, the KK-Ay mice were administered WEA or water for 12 weeks. The velocities of ocular blood flow were determined by Doppler ultrasound. RNA sequencing and reverse transcription quantitative PCR (RT-qPCR) were performed on WEA-stimulated RF/6A cells to reveal the underlying mechanism. RESULTS The cytotoxicity assay found that 30 μg/mL MEA, 20 μg/mL EEA and 30 μg/mL WEA had no toxic effect on RF/6A cells. The cell viability results showed that MEA, EEA and WEA all decreased cell viability. Compared with the high-glucose group, both MEA and WEA decreased the number of migrated cells, while the inhibition rate of WEA was higher. The Matrigel results showed that 30 μg/mL WEA effectively reduced the total tube length. Moreover, WEA improved the haemodynamics of the central retinal artery. RNA sequencing coupled with RT-qPCR verified that WEA regulated angiogenesis-related factors in high glucose-stimulated RF/6A cells. CONCLUSIONS WEA inhibits the migration and tube formation of RF/6A cells and improves diabetic retinopathy (DR) by mediating angiogenesis.
Collapse
Affiliation(s)
- Zheng-Lin Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China.
| | - Hui-Hui Sun
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China.
| | - Han-Ying Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China.
| | - Qing-Xuan Ji
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China.
| | - Yi-Ting Niu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China.
| | - Pan Ma
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China.
| | - Gaimei Hao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Jing-Xuan Zhang
- Institute of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Yue-Ying Yuan
- Institute of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Xin-Lou Chai
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China.
| | - Jing Han
- Institute of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China.
| | - Wei Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China.
| |
Collapse
|
38
|
Xu G, Yang Z, Sun Y, Dong H, Ma J. Interaction of microRNAs with sphingosine kinases, sphingosine-1 phosphate, and sphingosine-1 phosphate receptors in cancer. Discov Oncol 2021; 12:33. [PMID: 35201458 PMCID: PMC8777508 DOI: 10.1007/s12672-021-00430-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Sphingosine-1-phosphate (S1P), a pleiotropic lipid mediator, participates in various cellular processes during tumorigenesis, including cell proliferation, survival, drug resistance, metastasis, and angiogenesis. S1P is formed by two sphingosine kinases (SphKs), SphK1 and SphK2. The intracellularly produced S1P is delivered to the extracellular space by ATP-binding cassette (ABC) transporters and spinster homolog 2 (SPNS2), where it binds to five transmembrane G protein-coupled receptors to mediate its oncogenic functions (S1PR1-S1PR5). MicroRNAs (miRNAs) are small non-coding RNAs, 21-25 nucleotides in length, that play numerous crucial roles in cancer, such as tumor initiation, progression, apoptosis, metastasis, and angiogenesis via binding to the 3'-untranslated region (3'-UTR) of the target mRNA. There is growing evidence that various miRNAs modulate tumorigenesis by regulating the expression of SphKs, and S1P receptors. We have reviewed various roles of miRNAs, SphKs, S1P, and S1P receptors (S1PRs) in malignancies and how notable miRNAs like miR-101, miR-125b, miR-128, and miR-506, miR-1246, miR-21, miR-126, miR499a, miR20a-5p, miR-140-5p, miR-224, miR-137, miR-183-5p, miR-194, miR181b, miR136, and miR-675-3p, modulate S1P signaling. These tumorigenesis modulating miRNAs are involved in different cancers including breast, gastric, hepatocellular carcinoma, prostate, colorectal, cervical, ovarian, and lung cancer via cell proliferation, invasion, angiogenesis, apoptosis, metastasis, immune evasion, chemoresistance, and chemosensitivity. Therefore, understanding the interaction of SphKs, S1P, and S1P receptors with miRNAs in human malignancies will lead to better insights for miRNA-based cancer therapy.
Collapse
Affiliation(s)
- Guangmeng Xu
- Department of Colorectal Surgery, The Second Hospital of Jilin University, Changchun, 130000 China
| | - Zecheng Yang
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun, 130000 China
| | - Yamin Sun
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun, 130000 China
| | - Hongmei Dong
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun, 130000 China
| | - Jingru Ma
- Clinical Laboratory, The Second Hospital of Jilin University, Changchun, 130000 China
| |
Collapse
|
39
|
Mechanisms of sphingosine-1-phosphate (S1P) signaling on excessive stress-induced root resorption during orthodontic molar intrusion. Clin Oral Investig 2021; 26:1003-1016. [PMID: 34363103 DOI: 10.1007/s00784-021-04084-3] [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] [Received: 12/17/2020] [Accepted: 07/15/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVES The aim of this study was to investigate cementocyte mechanotransduction during excessive orthodontic intrusive force-induced root resorption and the role of S1P signaling in this process. MATERIALS AND METHODS Fifty-four 12-week-old male Wistar rats were randomly divided into 3 groups: control group (Control), intrusive stress application group (Stress), and intrusive stress together with S1PR2-specific antagonist injection group (Stress + JTE). A rat molar intrusion model was established on animals in the Stress and the Stress + JTE groups. The animals in the Stress + JTE group received daily intraperitoneal (i.p.) injection of S1PR2 antagonist JTE-013, while the Control and Stress groups received only the vehicle. Histomorphometric, immunohistochemical, quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analyses were performed after euthanizing of the rats. RESULTS Root resorption was promoted in the Stress group with increased volumes of resorption pits and amounts of molar intrusion compared with the Control group. The expression levels of cementogenic- and cementoclastic-related factors were affected under excessive intrusive force. Immunohistochemical staining and qRT-PCR analysis showed promoted S1P signaling activities during molar intrusion. Western blot analysis indicated decreased nuclear translocation of β-catenin under excessive intrusive force. Through the administration of JTE-013, S1P signaling activity was suppressed and excessive intrusive force-induced root resorption was reversed. The regulation of S1P signaling could also influence the nuclear translocation of β-catenin and the expressions of cementogenic- and cementoclastic-related factors. CONCLUSIONS Root resorption was promoted under excessive orthodontic intrusive force due to the disruption of cementum homeostasis. S1P signaling pathway might play an important role in cementocyte mechanotransduction in this process. CLINICAL RELEVANCE The S1P signaling might be a promising therapeutic target for novel therapeutic approaches to prevent external root resorption caused by excessive orthodontic intrusive force.
Collapse
|
40
|
Chen X, Ruan H, Ma Z, Hu J, Xu W, Yin L, Fu S. Polymerase Chain Reaction in the Detection of miR-455-5p and Sphingosine-1 Phosphate Proteins in Cervical Carcinoma with the Help of Gold Nanoparticles-Based. J Biomed Nanotechnol 2021; 17:1535-1544. [PMID: 34544531 DOI: 10.1166/jbn.2021.3138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study aimed to detect miR-455-5p and S1PR1 proteins using nanoparticle-assisted polymerase chain reaction (nano-PCR) to determine their correlation with cervical carcinoma prognosis. To achieve this study's goals, we selected 48 cervical carcinoma patients between January 2014 to January 2016 and subjected them to the miR-455-5p test by nano-PCR. The collected samples were then divided into two groups based on miR-455-5p levels. We had four HeLa cell groups, one group as the control, and one group overexpressed the miR-455-5p protein. A third group was miR-455-5p silent, and a separate group overexpressed both the miR-455-5p and S1PR1 proteins. Results also proved that the nano-PCR had a higher sensitivity than RT-PCR, and patients with poor prognosis had lesser miR-455-5p levels. Similarly, high levels of miR-455-5 contributed to cancer cell apoptosis and migration inhibition by targeting S1PR1 expression negatively. These two biomarkers are therefore significantly related to the prognosis of cervical carcinoma patients.
Collapse
Affiliation(s)
- Xinping Chen
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, 570228, Hainan, PR China
| | - Heqiu Ruan
- Central Laboratory, Hainan General Hospital, Hainan Hospital Affiliated to the Hainan Medical College, Hainan Provincial Key Laboratory of Cell and Molecular Genetic Translational Medicine, Haikou, Hainan Province 570311, China
| | - Zhichao Ma
- Central Laboratory, Hainan General Hospital, Hainan Hospital Affiliated to the Hainan Medical College, Hainan Provincial Key Laboratory of Cell and Molecular Genetic Translational Medicine, Haikou, Hainan Province 570311, China
| | - Junjie Hu
- Central Laboratory, Hainan General Hospital, Hainan Hospital Affiliated to the Hainan Medical College, Hainan Provincial Key Laboratory of Cell and Molecular Genetic Translational Medicine, Haikou, Hainan Province 570311, China
| | - Weihua Xu
- Central Laboratory, Hainan General Hospital, Hainan Hospital Affiliated to the Hainan Medical College, Hainan Provincial Key Laboratory of Cell and Molecular Genetic Translational Medicine, Haikou, Hainan Province 570311, China
| | - Liyan Yin
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, 570228, Hainan, PR China
| | - Shengmiao Fu
- Central Laboratory, Hainan General Hospital, Hainan Hospital Affiliated to the Hainan Medical College, Hainan Provincial Key Laboratory of Cell and Molecular Genetic Translational Medicine, Haikou, Hainan Province 570311, China
| |
Collapse
|
41
|
Ying Y, Ma X, Fang J, Chen S, Wang W, Li J, Xie H, Wu J, Xie B, Liu B, Wang X, Zheng X, Xie L. EGR2-mediated regulation of m 6A reader IGF2BP proteins drive RCC tumorigenesis and metastasis via enhancing S1PR3 mRNA stabilization. Cell Death Dis 2021; 12:750. [PMID: 34326314 PMCID: PMC8322060 DOI: 10.1038/s41419-021-04038-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/24/2022]
Abstract
Emerging discoveries of dynamic and reversible N6-methyladenosine (m6A) modification on RNA in mammals have revealed the key roles of the modification in human tumorigenesis. As known m6A readers, insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs) are upregulated in most cancers and mediates the enhancement of m6A-modified mRNAs stability. However, the mechanisms of IGF2BPs in renal cell cancer (RCC) still remain unclear. Bioinformatic analysis and RT-qPCR were performed to evaluate the expression of IGF2BPs and m6A writer Wilms tumor 1-associating protein (WTAP) in RCC samples and its correlation with patient prognosis. In vitro, in vivo biological assays were performed to investigate the functions of IGF2BPs and WTAP in RCC. Chromatin immunoprecipitation-qPCR (ChIP-qPCR) combined with bioinformatics analysis and following western blot assay, dual-luciferase reporter assays were performed to validate the regulatory relationships between transcription factor (TF) early growth response 2 (EGR2) and potential target genes IGF2BPs. RNA sequencing (RNA-seq), methylated RNA immunoprecipitation-qPCR (MERIP-qPCR), RIP-qPCR, m6A dot blot, and dual-luciferase reporter assays combined with bioinformatics analysis were employed to screen and validate the direct targets of IGF2BPs and WTAP. Here, we showed that early growth response 2 (EGR2) transcription factor could increase IGF2BPs expression in RCC. IGF2BPs in turn regulated sphingosine-1-phosphate receptor 3 (S1PR3) expression in an m6A-dependent manner by enhancing the stability of S1PR3 mRNA. They also promoted kidney tumorigenesis via PI3K/AKT pathway. Furthermore, IGF2BPs and WTAP upregulation predicted poor overall survival in RCC. Our studies showed that the EGR2/IGF2BPs regulatory axis and m6A-dependent regulation of S1PR3-driven RCC tumorigenesis, which enrich the m6A-modulated regulatory network in renal cell cancer. Together, our findings provide new evidence for the role of N6-methyladenosine modification in RCC.
Collapse
Affiliation(s)
- Yufan Ying
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China
| | - Xueyou Ma
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China
| | - Jiajie Fang
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China
| | - Shiming Chen
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China
| | - Weiyu Wang
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China
| | - Jiangfeng Li
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China
| | - Haiyun Xie
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China
| | - Jian Wu
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China
| | - Bo Xie
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China
| | - Ben Liu
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China.,Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Xiao Wang
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China.
| | - Xiangyi Zheng
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China. .,Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
| | - Liping Xie
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, Zhejiang, China. .,Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
| |
Collapse
|
42
|
Extracellular Sphingosine-1-Phosphate Downstream of EGFR Increases Human Glioblastoma Cell Survival. Int J Mol Sci 2021; 22:ijms22136824. [PMID: 34201962 PMCID: PMC8268299 DOI: 10.3390/ijms22136824] [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: 06/10/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 11/22/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a crucial mediator involved in the progression of different cancers, including glioblastoma multiforme (GBM), the most frequent and deadly human brain tumor, characterized by extensive invasiveness and rapid cell growth. Most of GBMs overexpress the epidermal growth factor receptor (EGFR), and we investigated the possible link between S1P and EGFR signaling pathways, focusing on its role in GBM survival, using the U87MG human cell line overexpressing EGFR (EGFR+). We previously demonstrated that EGFR+ cells have higher levels of extracellular S1P and increased sphingosine kinase-1 (SK1) activity than empty vector expressing cells. Notably, we demonstrated that EGFR+ cells are resistant to temozolomide (TMZ), the standard chemotherapeutic drug in GBM treatment, and the inhibition of SK1 or S1P receptors made EGFR+ cells sensitive to TMZ; moreover, exogenous S1P reverted this effect, thus involving extracellular S1P as a survival signal in TMZ resistance in GBM cells. In addition, both PI3K/AKT and MAPK inhibitors markedly reduced cell survival, suggesting that the enhanced resistance to TMZ of EGFR+ cells is dependent on the increased S1P secretion, downstream of the EGFR-ERK-SK1-S1P pathway. Altogether, our study provides evidence of a functional link between S1P and EGFR signaling pathways enhancing the survival properties of GBM cells.
Collapse
|
43
|
Zhou N, Li Z, Wang JJ, Wu QT, Li K, Zheng XK, Feng WS. Correlation analysis between extracts and endogenous metabolites to characterise the influence of salt-processing on compatibility mechanism of 'Psoraleae Fructus & Foeniculi Fructus'. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113782. [PMID: 33421603 DOI: 10.1016/j.jep.2021.113782] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 'Salt-processed Psoraleae Fructus & salt-processed Foeniculi Fructus' (sPF&sFF) is a common Chinese medicinal combination for treating diarrhoea. However, it is not clear how sPF and sFF work together, and why salt-processing is necessary. AIM OF THE STUDY To investigate the compatibility mechanism of sPF&sFF and the influence of salt-processing on it. MATERIALS AND METHODS Firstly, the metabolomics approach was appliedto screen the differential components between four (s)PF&(s)FF extracts, i.e., sPF&sFF, sPF&FF, PF&sFF, and PF&FF extracts. Then, an in vivo metabolomics study was carried out to filter critical metabolites reflecting the curative effects of (s)PF&(s)FF, and construct a metabolic network. Finally, a correlation analysis between chemical components in extracts and critical metabolites in vivo was performed to find out the synergistic and/or antagonistic effects between herbs as well as the influence of salt-processing. RESULTS Salt-processing had a direct influence on the contents of chemical components in sPF and sFF extracts, and there existed positive/negative correlations between the content change of chemical components and the effects of critical metabolites. Therefore, salt-processing indirectly affected on these correlations and was (i) conducive to the positive effects of sPF and sFF on bile acids, making sFF play a synergistic role, thereby, sPF&sFF could perform better than sPF and other three combinations and effectively relieve the symptoms of fatty diarrhoea, osmotic diuresis, malnutrition, and weight loss; (ii) conducive to the positive effects of sPF on triacylglycerol, 12(S)-hydroxyeicosatetraenoic acid, cholesterol, and arachidonic acid, and adverse to that of sFF, making sFF play an antagonistic role, thereby, sPF&sFF could prevent a series of side effects caused by over-regulation and suitably relieve the symptoms of osmotic diuresis, polyuria, malnutrition, and weight loss; and (iii) adverse to the positive effects of sPF and sFF on thromboxane A2, sphinganine and sphingosine, making sFF play a synergistic role, thereby, sPF&sFF could prevent a series of side effects and moderately relieve the symptoms of metabolic diarrhoea and polyuria. CONCLUSIONS Salt-processing indirectly affected on the correlations between chemical components in extracts and critical metabolites in vivo, and exhibited both conducive and adverse effects on the efficacy, making sPF and sFF cooperate with each other to moderately repair the metabolic disorders. Thereby, sPF&sFF could suitably relieve the diarrhoea and polyuria symptoms in the model and exert the most appropriate efficacy. Moreover, this novel strategy provided a feasible approach for further studying the compatibility mechanism of herbs.
Collapse
Affiliation(s)
- Ning Zhou
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, 450046, PR China.
| | - Zhe Li
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Jin-Jin Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China.
| | - Qi-Tong Wu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China.
| | - Kai Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Henan Research Center for Special Processing Technology of Chinese Medicine, Zhengzhou, 450046, PR China.
| | - Xiao-Ke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Henan University of Chinese Medicine, Co-construction of Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Zhengzhou, 450046, PR China.
| | - Wei-Sheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, 450046, PR China.
| |
Collapse
|
44
|
Gupta P, Taiyab A, Hussain A, Alajmi MF, Islam A, Hassan MI. Targeting the Sphingosine Kinase/Sphingosine-1-Phosphate Signaling Axis in Drug Discovery for Cancer Therapy. Cancers (Basel) 2021; 13:1898. [PMID: 33920887 PMCID: PMC8071327 DOI: 10.3390/cancers13081898] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/11/2021] [Accepted: 04/03/2021] [Indexed: 02/07/2023] Open
Abstract
Sphingolipid metabolites have emerged as critical players in the regulation of various physiological processes. Ceramide and sphingosine induce cell growth arrest and apoptosis, whereas sphingosine-1-phosphate (S1P) promotes cell proliferation and survival. Here, we present an overview of sphingolipid metabolism and the compartmentalization of various sphingolipid metabolites. In addition, the sphingolipid rheostat, a fine metabolic balance between ceramide and S1P, is discussed. Sphingosine kinase (SphK) catalyzes the synthesis of S1P from sphingosine and modulates several cellular processes and is found to be essentially involved in various pathophysiological conditions. The regulation and biological functions of SphK isoforms are discussed. The functions of S1P, along with its receptors, are further highlighted. The up-regulation of SphK is observed in various cancer types and is also linked to radio- and chemoresistance and poor prognosis in cancer patients. Implications of the SphK/S1P signaling axis in human pathologies and its inhibition are discussed in detail. Overall, this review highlights current findings on the SphK/S1P signaling axis from multiple angles, including their functional role, mechanism of activation, involvement in various human malignancies, and inhibitor molecules that may be used in cancer therapy.
Collapse
Affiliation(s)
- Preeti Gupta
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| | - Aaliya Taiyab
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.H.); (M.F.A.)
| | - Mohamed F. Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.H.); (M.F.A.)
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| |
Collapse
|
45
|
Li Q, Li Y, Lei C, Tan Y, Yi G. Sphingosine-1-phosphate receptor 3 signaling. Clin Chim Acta 2021; 519:32-39. [PMID: 33811927 DOI: 10.1016/j.cca.2021.03.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/18/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive lipid which regulates a series of physiological and pathological processes via binding to five S1P receptors (S1PR1-5). Although S1PR1-3 are widely expressed, the study of S1PRs, however, mainly addressed S1PR1 and S1PR2, and few studies focus on S1PR3-5. In recent years, a growing number of studies have shown that S1PR3 plays an important role in cell proliferation, differentiation, apoptosis, and migration, but its function is still controversial. This is the first comprehensive review paper about the role of S1PR3 signaling in cardiovascular function, tissue fibrosis, cancer, immune response, and neurological function. In addition, existing S1PR3 agonists and antagonists are listed at the end of the article, and we also put forward our opinion on the dispute of S1PR3 function.
Collapse
Affiliation(s)
- Qian Li
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Yi Li
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Cai Lei
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Ying Tan
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Guanghui Yi
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
46
|
Magaye RR, Savira F, Hua Y, Xiong X, Huang L, Reid C, Flynn BL, Kaye D, Liew D, Wang BH. Attenuating PI3K/Akt- mTOR pathway reduces dihydrosphingosine 1 phosphate mediated collagen synthesis and hypertrophy in primary cardiac cells. Int J Biochem Cell Biol 2021; 134:105952. [PMID: 33609744 DOI: 10.1016/j.biocel.2021.105952] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/15/2022]
Abstract
Cardiac fibrosis and myocyte hypertrophy play contributory roles in the progression of diseases such as heart Failure (HF) through what is collectively termed cardiac remodelling. The phosphoinositide 3- kinase (PI3K), protein kinase B (Akt) and mammalian target for rapamycin (mTOR) signalling pathway (PI3K/Akt- mTOR) is an important pathway in protein synthesis, cell growth, cell proliferation, and lipid metabolism. The sphingolipid, dihydrosphingosine 1 phosphate (dhS1P) has been shown to bind to high density lipids in plasma. Unlike its analog, spingosine 1 phosphate (S1P), the role of dhS1P in cardiac fibrosis is still being deciphered. This study was conducted to investigate the effect of dhS1P on PI3K/Akt signalling in primary cardiac fibroblasts and myocytes. Our findings demonstrate that inhibiting PI3K reduced collagen synthesis in neonatal cardiac fibroblasts (NCFs), and hypertrophy in neonatal cardiac myocytes (NCMs) induced by dhS1P, in vitro. Reduced activation of the PI3K/Akt- mTOR signalling pathway led to impaired translation of fibrotic proteins such as collagen 1 (Coll1) and transforming growth factor β (TGFβ) and inhibited the transcription and translation of tissue inhibitor of matrix metalloproteinase 1 (TIMP1). PI3K inhibition also affected the gene expression of S1P receptors and enzymes such as the dihydroceramide delta 4 desaturase (DEGS1) and sphingosine kinase 1 (SK1) in the de novo sphingolipid pathway. While in myocytes, PI3K inhibition reduced myocyte hypertrophy induced by dhS1P by reducing skeletal muscle α- actin (αSKA) mRNA expression, and protein translation due to increased glycogen synthase kinase 3β (GSK3β) mRNA expression. Our findings show a relationship between the PI3K/Akt- mTOR signalling cascade and exogenous dhS1P induced collagen synthesis and myocyte hypertrophy in primary neonatal cardiac cells.
Collapse
Affiliation(s)
- Ruth R Magaye
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Feby Savira
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Yue Hua
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xin Xiong
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; Shanghai Institute of Heart Failure, Research Centre for Translational Medicine, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai 200120, China
| | - Li Huang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Christopher Reid
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; School of Public Health School, Curtin University, Perth, Australia
| | - Bernard L Flynn
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - David Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Danny Liew
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Bing H Wang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia.
| |
Collapse
|
47
|
Paradiso E, Lazzaretti C, Sperduti S, Antoniani F, Fornari G, Brigante G, Di Rocco G, Tagliavini S, Trenti T, Morini D, Falbo AI, Villani MT, Nofer JR, Simoni M, Potì F, Casarini L. Sphingosine-1 phosphate induces cAMP/PKA-independent phosphorylation of the cAMP response element-binding protein (CREB) in granulosa cells. Mol Cell Endocrinol 2021; 520:111082. [PMID: 33189864 DOI: 10.1016/j.mce.2020.111082] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/20/2020] [Accepted: 11/09/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS Sphingosine-1 phosphate (S1P) is a lysosphingolipid present in the ovarian follicular fluid. The role of the lysosphingolipid in gonads of the female is widely unclear. At nanomolar concentrations, S1P binds and activates five specific G protein-coupled receptors (GPCRs), known as S1P1-5, modulating different signaling pathways. S1P1 and S1P3 are highly expressed in human primary granulosa lutein cells (hGLC), as well as in the immortalized human primary granulosa cell line hGL5. In this study, we evaluated the signaling cascade activated by S1P and its synthetic analogues in hGLC and hGL5 cells, exploring the biological relevance of S1PR-stimulation in this context. METHODS AND RESULTS hGLC and hGL5 cells were treated with a fixed dose (0.1 μM) of S1P, or by S1P1- and S1P3-specific agonists SEW2871 and CYM5541. In granulosa cells, S1P and, at a lesser extent, SEW2871 and CYM5541, potently induced CREB phosphorylation. No cAMP production was detected and pCREB activation occurred even in the presence of the PKA inhibitor H-89. Moreover, S1P-dependent CREB phosphorylation was dampened by the mitogen-activate protein kinase (MEK) inhibitor U0126 and by the L-type Ca2+ channel blocker verapamil. The complete inhibition of CREB phosphorylation occurred by blocking either S1P2 or S1P3 with the specific receptor antagonists JTE-013 and TY52156, or under PLC/PI3K depletion. S1P-dependent CREB phosphorylation induced FOXO1 and the EGF-like epiregulin-encoding gene (EREG), confirming the exclusive role of gonadotropins and interleukins in this process, but did not affect steroidogenesis. However, S1P or agonists did not modulate granulosa cell viability and proliferation in our conditions. CONCLUSIONS This study demonstrates for the first time that S1P may induce a cAMP-independent activation of pCREB in granulosa cells, although this is not sufficient to induce intracellular steroidogenic signals and progesterone synthesis. S1P-induced FOXO1 and EREG gene expression suggests that the activation of S1P-S1PR axis may cooperate with gonadotropins in modulating follicle development.
Collapse
Affiliation(s)
- Elia Paradiso
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy
| | - Clara Lazzaretti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy
| | - Samantha Sperduti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Antoniani
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Fornari
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Brigante
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Giulia Di Rocco
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Simonetta Tagliavini
- Department of Laboratory Medicine and Pathological Anatomy, Azienda Ospedaliero-Universitaria di Modena, NOCSAE, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathological Anatomy, Azienda Ospedaliero-Universitaria di Modena, NOCSAE, Modena, Italy
| | - Daria Morini
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN. Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Modena, Italy
| | - Angela Immacolata Falbo
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN. Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Modena, Italy
| | - Maria Teresa Villani
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN. Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Modena, Italy
| | - Jerzy-Roch Nofer
- Central Laboratory Facility, University Hospital Münster, Münster, Germany
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy; Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy; PR China, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Francesco Potì
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
48
|
Rostami N, Nikkhoo A, Khazaei-Poul Y, Farhadi S, Sadat Haeri M, Moghadaszadeh Ardebili S, Aghaei Vanda N, Atyabi F, Namdar A, Baghaei M, Haghnavaz N, Kazemi T, Yousefi M, Ghalamfarsa G, Sabz G, Jadidi-Niaragh F. Coinhibition of S1PR1 and GP130 by siRNA-loaded alginate-conjugated trimethyl chitosan nanoparticles robustly blocks development of cancer cells. J Cell Physiol 2020; 235:9702-9717. [PMID: 32424937 DOI: 10.1002/jcp.29781] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
There is an interconnected network between S1P/sphingosine-1-phosphate receptor 1 (S1PR1), IL-6/glycoprotein 130 (GP130), and signal transducer and activator of transcription 3 (STAT3) signaling pathways in the tumor microenvironment, which leads to cancer progression. S1P/S1PR1 and IL-6/GP130 signaling pathways phosphorylate and activate STAT3, and it then induces the expression of S1PR1 and interleukin-6 (IL-6) in a positive feedback loop leading to cancer progression. We hypothesized that blockade of this amplification loop can suppress the growth and development of cancer cells. Therefore, we silenced STAT3 upstream molecules including the S1PR1 and GP130 molecules in cancer cells using small interfering RNA (siRNA)-loaded alginate-conjugated trimethyl chitosan (ATMC) nanoparticles (NPs). The generated NPs had competent properties including the appropriate size, zeta potential, polydispersity index, morphology, high uptake of siRNA, high rate of capacity, high stability, and low toxicity. We evaluated the effects of siRNA loaded ATMC NPs on tumor hallmarks of three murine-derived cancer cell lines, including 4T1 (breast cancer), B16-F10 (melanoma), and CT26 (colon cancer). The results confirmed the tumor-suppressive effects of combinational targeting of S1PR1 and GP130. Moreover, combination therapy could potently suppress tumor growth as assessed by the chick chorioallantoic membrane assay. In this study, we targeted this positive feedback loop for the first time and applied this novel combination therapy, which provides a promising approach for cancer treatment. The development of a potent nanocarrier system with ATMC for this combination was also another aspect of this study, which should be further investigated in cancer animal models in further studies.
Collapse
Affiliation(s)
- Narges Rostami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Nikkhoo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yalda Khazaei-Poul
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shohreh Farhadi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Melika Sadat Haeri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Fatemeh Atyabi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Afshin Namdar
- Department of Oncology, Cross Cancer Institute, The University of Alberta, Edmonton, Alberta, Canada
| | - Masoumeh Baghaei
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Navideh Haghnavaz
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Gholamabas Sabz
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
49
|
Pan Y, Liu L, Zhang Q, Shi W, Feng W, Wang J, Wang Q, Li S, Li M. Activation of AMPK suppresses S1P-induced airway smooth muscle cells proliferation and its potential mechanisms. Mol Immunol 2020; 128:106-115. [PMID: 33126079 DOI: 10.1016/j.molimm.2020.09.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 01/13/2023]
Abstract
The aims of the present study were to investigate the signaling mechanisms for sphingosine-1-phosphate (S1P)-induced airway smooth muscle cells (ASMCs) proliferation and to explore the effect of activation of adenosine monophosphate-activated protein kinase (AMPK) on S1P-induced ASMCs proliferation and its underlying mechanisms. S1P phosphorylated signal transducer and activator of transcription 3 (STAT3) through binding to S1PR2/3, and this further sequentially up-regulated polo-like kinase 1 (PLK1) and inhibitor of differentiation 2 (ID2) protein expression. Pretreatment of cells with S1PR2 antagonist JTE-013, S1PR3 antagonist CAY-10444, knockdown of STAT3, PLK1 and ID2 attenuated S1P-triggered ASMCs proliferation. In addition, activation of AMPK by metformin inhibited S1P-induced ASMCs proliferation by suppressing STAT3 phosphorylation and therefore suppression of PLK1 and ID2 protein expression. Our study suggests that S1P promotes ASMCs proliferation by stimulating S1PR2/3/STAT3/PLK1/ID2 axis, and activation of AMPK suppresses ASMCs proliferation by targeting on STAT3 signaling pathway. Activation of AMPK might benefit asthma by inhibiting airway remodeling.
Collapse
Affiliation(s)
- Yilin Pan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Pulmonary and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wei Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
50
|
Ouyang J, Shu Z, Chen S, Xiang H, Lu H. The role of sphingosine 1-phosphate and its receptors in cardiovascular diseases. J Cell Mol Med 2020; 24:10290-10301. [PMID: 32803879 PMCID: PMC7521328 DOI: 10.1111/jcmm.15744] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/12/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
There are many different types of cardiovascular diseases, which impose a huge economic burden due to their extremely high mortality rates, so it is necessary to explore the underlying mechanisms to achieve better supportive and curative care outcomes. Sphingosine 1‐phosphate (S1P) is a bioactive lipid mediator with paracrine and autocrine activities that acts through its cell surface S1P receptors (S1PRs) and intracellular signals. In the circulatory system, S1P is indispensable for both normal and disease conditions; however, there are very different views on its diverse roles, and its specific relevance to cardiovascular pathogenesis remains elusive. Here, we review the synthesis, release and functions of S1P, specifically detail the roles of S1P and S1PRs in some common cardiovascular diseases, and then address several controversial points, finally, we focus on the development of S1P‐based therapeutic approaches in cardiovascular diseases, such as the selective S1PR1 modulator amiselimod (MT‐1303) and the non‐selective S1PR1 and S1PR3 agonist fingolimod, which may provide valuable insights into potential therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Jie Ouyang
- Center for Experimental Medical Research, the Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhihao Shu
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, China
| | - Shuhua Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, China
| | - Hong Xiang
- Center for Experimental Medical Research, the Third Xiangya Hospital of Central South University, Changsha, China
| | - Hongwei Lu
- Center for Experimental Medical Research, the Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|