1
|
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
|
2
|
Wang YQ, Ren Y, Gale RP, Niu LT, Huang XJ. Sphingosine-1 phosphate receptor 1 (S1PR1) expression maintains stemness of acute myeloid leukemia stem cells. Cancer Lett 2024; 600:217158. [PMID: 39111385 DOI: 10.1016/j.canlet.2024.217158] [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: 04/16/2024] [Revised: 07/24/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
Acute myeloid leukemia (AML) arises from leukemia stem cells (LSCs) and is maintained by cells which have acquired features of stemness. We compared transcription profiles of AML cells with/without stem cell features defined as in vitro clonogenicity and serial engraftment in immune-deficient mice xenograft model. We used multi-parameter flow cytometry (MPFC) to separate CD34+ bone marrow-derived leukemia cells into sphingosine-1 phosphate receptor 1 (S1PR1)+ and S1PR1- fractions. Cells in the S1PR1+ fraction demonstrated significantly higher clonogenicity and higher engraftment potential compared with those in the S1PR1- fraction. In contrast, CD34+ bone marrow cells from normal samples showed reduced clonogenicity in the S1PR1+ fraction compared with the S1PR1- fraction. Inhibition of S1PR1 expression in an AML cell line reduced the colony-forming potential of KG1 cells. Transcriptomic analyses and rescue experiments indicated PI3K/AKT pathway and MYBL2 are downstream mediators of S1PR1-associated stemness. These findings implicate S1PR1 as a functional biomarker of LSCs and suggest its potential as a therapeutic target in AML treatment.
Collapse
Affiliation(s)
- Yu-Qing Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yue Ren
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Robert Peter Gale
- Centre for Hematology Research, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Li-Ting Niu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
3
|
Taylor O, DeGroff N, El-Hodiri H, Gao C, Fischer AJ. Sphingosine-1-phosphate signaling regulates the ability of Müller glia to become neurogenic, proliferating progenitor-like cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606815. [PMID: 39149287 PMCID: PMC11326190 DOI: 10.1101/2024.08.06.606815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The purpose of these studies is to investigate how Sphingosine-1-phosphate (S1P) signaling regulates glial phenotype, dedifferentiation of Müller glia (MG), reprogramming into proliferating MG-derived progenitor cells (MGPCs), and neuronal differentiation of the progeny of MGPCs. We found that S1P-related genes are highly expressed by retinal neurons and glia, and levels of expression were dynamically regulated following retinal damage. S1PR1 is highly expressed by resting MG and is rapidly downregulated following acute retinal damage. Drug treatments that activate S1PR1 or increase levels of S1P suppressed the formation of MGPCs, whereas treatments that inhibit S1PR1 or decreased levels of S1P stimulated the formation of MGPCs. Inhibition of S1PR1 or SPHK1 significantly enhanced the neuronal differentiation of the progeny of MGPCs. Further, ablation of microglia from the retina, wherein the formation of MGPCs in damaged retinas is impaired, has a significant impact upon expression patterns of S1P-related genes in MG. Inhibition of S1PR1 and SPHK1 partially rescued the formation of MGPCs in damaged retinas missing microglia. Finally, we show that TGFβ/Smad3 signaling in the resting retina maintains S1PR1 expression in MG. We conclude that the S1P signaling is dynamically regulated in MG and MGPCs and activation of S1P signaling depends, in part, on signals produced by reactive microglia.
Collapse
Affiliation(s)
- Olivia Taylor
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nicholas DeGroff
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Heithem El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Chengyu Gao
- Campus Chemical Instrument Center, Mass Spectrometry & Proteomics Facility, The Ohio State University, Columbus, OH 43210, USA
| | - Andy J. Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
4
|
Hao W, Luo D, Jiang Y, Wan S, Li X. An overview of sphingosine-1-phosphate receptor 2: Structure, biological function, and small-molecule modulators. Med Res Rev 2024; 44:2331-2362. [PMID: 38665010 DOI: 10.1002/med.22044] [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: 01/03/2024] [Revised: 04/02/2024] [Accepted: 04/14/2024] [Indexed: 08/09/2024]
Abstract
Over the past decade, there has been a notable increase in research on sphingosine-1-phosphate receptor 2 (S1PR2), which is a type of G-protein-coupled receptor. Upon activation by S1P or other ligands, S1PR2 initiates downstream signaling pathways such as phosphoinositide 3-kinase (PI3K), Mitogen-activated protein kinase (MAPK), Rho/Rho-associated coiled-coil containing kinases (ROCK), and others, contributing to the diverse biological functions of S1PR2 and playing a pivotal role in various physiological processes and disease progressions, such as multiple sclerosis, fibrosis, inflammation, and tumors. Due to the extensive biological functions of S1PR2, many S1PR2 modulators, including agonists and antagonists, have been developed and discovered by pharmaceutical companies (e.g., Novartis and Galapagos NV) and academic medicinal chemists for disease diagnosis and treatment. However, few reviews have been published that comprehensively overview the functions and regulators of S1PR2. Herein, we provide an in-depth review of the advances in the function of S1PR2 and its modulators. We first summarize the structure and biological function of S1PR2 and its pathological role in human diseases. We then focus on the discovery approach, design strategy, development process, and biomedical application of S1PR2 modulators. Additionally, we outline the major challenges and future directions in this field. Our comprehensive review will aid in the discovery and development of more effective and clinically applicable S1PR2 modulators.
Collapse
Affiliation(s)
- Wanting Hao
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Dongdong Luo
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Yuqi Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shengbiao Wan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Xiaoyang Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Marine Biomedical Research, Institute of Qingdao, Qingdao, China
| |
Collapse
|
5
|
Dong D, Yu X, Tao X, Wang Q, Zhao L. S1P/S1PR1 signaling is involved in the development of nociceptive pain. Front Pharmacol 2024; 15:1407347. [PMID: 39045057 PMCID: PMC11263082 DOI: 10.3389/fphar.2024.1407347] [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: 03/26/2024] [Accepted: 06/20/2024] [Indexed: 07/25/2024] Open
Abstract
Background Pain is a complex perception involving unpleasant somatosensory and emotional experiences. However, the underlying mechanisms that mediate its different components remain unclear. Sphingosine-1-phosphate (S1P), a metabolite of sphingomyelin and a potent lipid mediator, initiates signaling via G protein-coupled receptors (S1PRs) on cell surfaces. It serves as a second messenger in cellular processes such as proliferation and apoptosis. Nevertheless, the neuropharmacology of sphingolipid signaling in pain conditions within the central nervous system remains largely unexplored and controversial. Methods Chronic nociceptive pain models were induced in vivo by intraplantar injection of 20 μL complete Freund's adjuvant (CFA) into the left hind paws. We assessed S1P and S1PR1 expression in the spinal cords of CFA model mice. Functional antagonists of S1PR1 or S1PR1-specific siRNA were administered daily following CFA model establishment. Paw withdrawal response frequency (PWF) and paw withdrawal latency (PWL) were measured to evaluate mechanical allodynia and thermal hyperalgesia, respectively. RT-PCR assessed interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α levels. Western blotting and immunofluorescence were used to analyze glial fibrillary acidic protein (GFAP), ionized calcium-binding adapter molecule (Iba1), STAT3, ERK, and p38 MAPK protein expression. Results In the chronic nociceptive pain model induced by CFA, S1P and S1PR1 expression levels were significantly elevated, leading to activation of spinal cord glial cells. S1PR1 activation also promoted MMP2-mediated cleavage of mature IL-1β. Additionally, S1PR1 activation upregulated phosphorylation of STAT3, ERK, and p38 MAPK in glial cells, profoundly impacting downstream signaling pathways and contributing to chronic nociceptive pain. Conclusion The S1P/S1PR1 axis plays a pivotal role in the cellular and molecular mechanisms underlying nociceptive pain. This signaling pathway modulates glial cell activation and the expression of pain-related genes (STAT3, ERK, p38 MAPK) and inflammatory factors in the spinal dorsal horn. These findings underscore the potential of targeting the S1P system for developing novel analgesic therapies.
Collapse
Affiliation(s)
- Daosong Dong
- Department of Pain, The First Hospital of China Medical University, Shenyang, China
| | - Xue Yu
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Ministry of Education, Shenyang, China
| | - Xueshu Tao
- Department of Pain, The First Hospital of China Medical University, Shenyang, China
| | - Qian Wang
- Medical Oncology, Department of Gastrointestinal Cancer, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Lin Zhao
- Department of Pain, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Janneh AH. Sphingolipid Signaling and Complement Activation in Glioblastoma: A Promising Avenue for Therapeutic Intervention. BIOCHEM 2024; 4:126-143. [PMID: 38894892 PMCID: PMC11185840 DOI: 10.3390/biochem4020007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Glioblastoma is the most common and aggressive type of malignant brain tumor with a poor prognosis due to the lack of effective treatment options. Therefore, new treatment options are required. Sphingolipids are essential components of the cell membrane, while complement components are integral to innate immunity, and both play a critical role in regulating glioblastoma survival signaling. This review focuses on recent studies investigating the functional roles of sphingolipid metabolism and complement activation signaling in glioblastoma. It also discusses how targeting these two systems together may emerge as a novel therapeutic approach.
Collapse
Affiliation(s)
- Alhaji H Janneh
- Hollings Cancer Center, Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
7
|
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
|
8
|
Beilankouhi EAV, Valilo M, Dastmalchi N, Teimourian S, Safaralizadeh R. The Function of Autophagy in the Initiation, and Development of Breast Cancer. Curr Med Chem 2024; 31:2974-2990. [PMID: 37138421 DOI: 10.2174/0929867330666230503145319] [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: 12/09/2020] [Revised: 02/26/2021] [Accepted: 03/15/2021] [Indexed: 05/05/2023]
Abstract
Autophagy is a significant catabolic procedure that increases in stressful conditions. This mechanism is mostly triggered after damage to the organelles, the presence of unnatural proteins, and nutrient recycling in reaction to these stresses. One of the key points in this article is that cleaning and preserving damaged organelles and accumulated molecules through autophagy in normal cells helps prevent cancer. Since dysfunction of autophagy is associated with various diseases, including cancer, it has a dual function in tumor suppression and expansion. It has newly become clear that the regulation of autophagy can be used for the treatment of breast cancer, which has a promising effect of increasing the efficiency of anticancer treatment in a tissue- and cell-type-specific manner by affecting the fundamental molecular mechanisms. Regulation of autophagy and its function in tumorigenesis is a vital part of modern anticancer techniques. This study discusses the current advances related to the mechanisms that describe essential modulators of autophagy involved in the metastasis of cancers and the development of new breast cancer treatments.
Collapse
Affiliation(s)
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Narges Dastmalchi
- Department of Biology, University College of Nabi Akram, Tabriz, Iran
| | - Shahram Teimourian
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
9
|
Tao YP, Zhu HY, Shi QY, Wang CX, Hua YX, Hu HY, Zhou QY, Zhou ZL, Sun Y, Wang XM, Wang Y, Zhang YL, Guo YJ, Wang ZY, Che X, Xu CW, Zhang XC, Heger M, Tao SP, Zheng X, Xu Y, Ao L, Liu AJ, Liu SB, Cheng SQ, Pan WW. S1PR1 regulates ovarian cancer cell senescence through the PDK1-LATS1/2-YAP pathway. Oncogene 2023; 42:3491-3502. [PMID: 37828220 PMCID: PMC10656284 DOI: 10.1038/s41388-023-02853-w] [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: 02/28/2023] [Revised: 09/18/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023]
Abstract
Cell senescence deters the activation of various oncogenes. Induction of senescence is, therefore, a potentially effective strategy to interfere with vital processes in tumor cells. Sphingosine-1-phosphate receptor 1 (S1PR1) has been implicated in various cancer types, including ovarian cancer. The mechanism by which S1PR1 regulates ovarian cancer cell senescence is currently elusive. In this study, we demonstrate that S1PR1 was highly expressed in human ovarian cancer tissues and cell lines. S1PR1 deletion inhibited the proliferation and migration of ovarian cancer cells. S1PR1 deletion promoted ovarian cancer cell senescence and sensitized ovarian cancer cells to cisplatin chemotherapy. Exposure of ovarian cancer cells to sphingosine-1-phosphate (S1P) increased the expression of 3-phosphatidylinositol-dependent protein kinase 1 (PDK1), decreased the expression of large tumor suppressor 1/2 (LATS1/2), and induced phosphorylation of Yes-associated protein (p-YAP). Opposite results were obtained in S1PR1 knockout cells following pharmacological inhibition. After silencing LATS1/2 in S1PR1-deficient ovarian cancer cells, senescence was suppressed and S1PR1 expression was increased concomitantly with YAP expression. Transcriptional regulation of S1PR1 by YAP was confirmed by chromatin immunoprecipitation. Accordingly, the S1PR1-PDK1-LATS1/2-YAP pathway regulates ovarian cancer cell senescence and does so through a YAP-mediated feedback loop. S1PR1 constitutes a druggable target for the induction of senescence in ovarian cancer cells. Pharmacological intervention in the S1PR1-PDK1-LATS1/2-YAP signaling axis may augment the efficacy of standard chemotherapy.
Collapse
Affiliation(s)
- Yi-Ping Tao
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Heng-Yan Zhu
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Qian-Yuan Shi
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Cai-Xia Wang
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Yu-Xin Hua
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
- Zhejiang Chinese Medicine University and Jiaxing University Master Degree Cultivation Base, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Han-Yin Hu
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
- Zhejiang Chinese Medicine University and Jiaxing University Master Degree Cultivation Base, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Qi-Yin Zhou
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
- Zhejiang Chinese Medicine University and Jiaxing University Master Degree Cultivation Base, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Zi-Lu Zhou
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Ying Sun
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Xiao-Min Wang
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Yu Wang
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Ya-Ling Zhang
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Yan-Jun Guo
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Zi-Ying Wang
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Xuan Che
- Department of Anesthesiology, Jiaxing Maternity and Child Health Care Hospital, Affiliated Women and Children Hospital, Jiaxing University, Jiaxing, Zhejiang Province, 314001, China
| | - Chun-Wei Xu
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, No. 1 Banshan East Street, Gongshu District, Hangzhou, 310022, China
| | - Xian-Chao Zhang
- Institute of Information Network and Artificial Intelligence, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, the Netherlands
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Su-Ping Tao
- Department of Gynecology and Obstetrics, Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| | - Xin Zheng
- Department of Gynecology and Obstetrics, Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| | - Ying Xu
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Lei Ao
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China
| | - Ai-Jun Liu
- Department of Pathology, the 7th Medical Center, General Hospital of PLA, Beijing, 100700, China
| | - Sheng-Bing Liu
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China.
| | - Shu-Qun Cheng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China.
| | - Wei-Wei Pan
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China.
- G60 STI Valley Industry & Innovation Institute, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, China.
| |
Collapse
|
10
|
Kurano M, Uranbileg B, Yatomi Y. Apolipoprotein M bound sphingosine 1-phosphate suppresses NETosis through activating S1P1 and S1P4. Biomed Pharmacother 2023; 166:115400. [PMID: 37657263 DOI: 10.1016/j.biopha.2023.115400] [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: 06/29/2023] [Revised: 08/23/2023] [Accepted: 08/26/2023] [Indexed: 09/03/2023] Open
Abstract
The pleiotropic effects of high-density lipoprotein (HDL), including its protective properties against sepsis, are attributed to the sphingosine 1-phosphate and apolipoprotein M (ApoM) that are carried on the lipoproteins. In this study, we attempted to elucidate the possible mechanisms underlying the sepsis coagulopathic state by considering the modulation of NETosis. Our results revealed that in a lipopolysaccharide-induced sepsis mouse model, the levels of NETosis markers, such as plasma DNA and histone, were elevated in ApoM-knockout (KO) mice and attenuated in ApoM-overexpressing mice. In ApoM-KO mice, the survival rate decreased and the occurrence rates of coagulopathy and organ injury increased following the administration of histone. Treatment with a conditioned medium of ApoM-overexpressing cells attenuated the observed NETosis in HL-60S cells that differentiated into neutrophils and were inhibited through the suppression of S1P1 or S1P4. The attenuation of PKCδ and PKCα/β by S1P1 and S1P4 activation may also be involved. In ApoM-overexpressing mice, coagulopathy and organ injuries were attenuated following an injection of histone; these effects were partially inhibited by S1P1, 3, S1P4, or S1P1 antagonists. Furthermore, the exogenous administration of ApoM protected ApoM-KO mice that were challenged with histone from developing NETosis. In conclusion, the ApoM/S1P axis protects against NETosis through the attenuation of PKC activation by S1P1 and S1P4. The development of drugs targeting the ApoM/S1P axis may be beneficial for the treatment of pathological conditions involving uncontrolled NETosis, such as sepsis.
Collapse
Affiliation(s)
- Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan.
| | - Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Pu X, Cao X, Liu H, Huang W, Zhang L, Jiang T. Isorhamnetin attenuates the proliferation, invasion, migration and fibrosis of keloid fibroblasts by targeting S1PR1. Exp Ther Med 2023; 26:310. [PMID: 37273758 PMCID: PMC10236135 DOI: 10.3892/etm.2023.12009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/14/2023] [Indexed: 06/06/2023] Open
Abstract
Isorhamnetin (IH) is a type of flavonoid with multiple biological activities, including cardioprotective, antitumor, anti-inflammatory and antioxidant activities. However, the role and potential mechanism of IH in keloids are still not completely understood. The aim of the present study was to explore how IH affects keloid progression. In the present study, cell proliferation was evaluated using the Cell Counting Kit-8 assay and immunofluorescence. Wound healing and Transwell assays were performed to assess cell migration and invasion, respectively. The expression levels of fibrosis-related proteins were measured using western blot analysis and immunofluorescence. In addition, the binding between IH and sphingosine-1-phosphate receptor-1 (S1PR1) was analyzed using the TargetNet database, and molecular docking was performed using Zinc, PubChem, AutoDockTools 1.5.6 and Discovery Studio 4.5 software. The expression levels of proteins in the PI3K/AKT pathway were detected by western blot analysis. The results showed that IH inhibited the proliferation, invasion, migration and fibrosis of keloid fibroblasts. The binding of IH and S1PR1 was verified and molecular docking was performed. Notably, IH significantly suppressed the expression levels of S1PR1, phosphorylated (p)-PI3K and p-AKT. Furthermore, the silencing of S1PR1 suppressed the cell proliferation, migration, invasion and fibrosis of keloid fibroblasts, as well as the expression of the PI3K/AKT pathway proteins. Conversely, S1PR1 upregulation reversed the inhibitory effects of IH on keloid fibroblast proliferation, migration, invasion and fibrosis. In conclusion, the results revealed that IH suppressed the proliferation, migration, invasion and fibrosis of keloid fibroblasts by targeting the S1PR1/PI3K/AKT pathway, suggesting that IH may be a promising drug for the treatment of keloids.
Collapse
Affiliation(s)
- Xiaoshu Pu
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China
| | - Xiaolei Cao
- General Surgery Department, The People's Hospital of Shunqing District, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China
| | - Hongyan Liu
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China
| | - Wenlian Huang
- Department of Critical Care Medicine, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China
| | - Lanfang Zhang
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China
| | - Ting Jiang
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
12
|
Wu X, Wabitsch M, Yang J, Sakharkar MK. Effects of adipocyte-conditioned cell culture media on S1P treatment of human triple-negative breast cancer cells. PLoS One 2023; 18:e0286111. [PMID: 37220155 DOI: 10.1371/journal.pone.0286111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a potent sphingolipid metabolite that regulates a wide range of biological functions such as cell proliferation, cell apoptosis and angiogenesis. Its cellular level is elevated in breast cancer, which, in turn, would promote cancer cell proliferation, survival, growth and metastasis. However, the cellular concentration of S1P is normally in the low nanomolar range, and our previous studies showed that S1P selectively induced apoptosis of breast cancer cells at high concentrations (high nanomolar to low micromolar). Thus, local administration of high-concentration S1P alone or in combination of chemotherapy agents could be used to treat breast cancer. The breast mainly consists of mammary gland and connective tissue stroma (adipose), which are dynamically interacting each other. Thus, in the current study, we evaluated how normal adipocyte-conditioned cell culture media (AD-CM) and cancer-associated adipocyte-conditioned cell culture media (CAA-CM) would affect high-concentration S1P treatment of triple-negative breast cancer (TNBC) cells. Both AD-CM and CAA-CM may suppress the anti-proliferative effect and reduce nuclear alteration/apoptosis caused by high-concentration S1P. This implicates that adipose tissue is likely to be detrimental to local high-concentration S1P treatment of TNBC. Because the interstitial concentration of S1P is about 10 times higher than its cellular level, we undertook a secretome analysis to understand how S1P would affect the secreted protein profile of differentiated SGBS adipocytes. At 100 nM S1P treatment, we identified 36 upregulated and 21 downregulated secretome genes. Most of these genes are involved in multiple biological processes. Further studies are warranted to identify the most important secretome targets of S1P in adipocytes and illustrate the mechanism on how these target proteins affect S1P treatment of TNBC.
Collapse
Affiliation(s)
- Xiyuan Wu
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Martin Wabitsch
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Jian Yang
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | | |
Collapse
|
13
|
Inhibitory Effect of Astragalus Polysaccharide on Premetastatic Niche of Lung Cancer through the S1PR1-STAT3 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:4010797. [PMID: 36714534 PMCID: PMC9883101 DOI: 10.1155/2023/4010797] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/15/2022] [Accepted: 12/26/2022] [Indexed: 01/22/2023]
Abstract
As a common malignant tumor, the morbidity and mortality of lung cancer have been rising in recent years. The concept of "premetastatic niche" may lead to a revolutionary change in antitumor metastasis therapeutic strategies. Traditional Chinese medicine with multitargets and lower poisonous agents may be a potentially effective means to intervene in the "premetastatic niche (PMN)" to prevent and treat tumor metastasis. Astragalus polysaccharide (APS) is a substance with strong immune activity in Astragalus membranaceus that has excellent biological activities such as immunomodulation, anti-inflammatory, and antitumor. In this study, we constructed a tumor lung metastasis animal model to explore the intervention mechanism of APS on the premetastatic niche. We found that APS inhibited the formation of the lung premetastatic niche and inhibited the recruitment of myeloid-derived suppressor cells (MDSCs) in the lung. Mechanistically, we showed that the proteins and gene expression of S1PR1, STAT3, and p-STAT3 in the S1PR1/STAT3 signaling pathway were suppressed by APS. In line with the above findings, our results confirmed that APS may inhibit the accumulation of MDSCs in the premetastatic niche through the intervention of the S1PR1-STAT3 signaling pathway to achieve the antitumor effect.
Collapse
|
14
|
Liu Z, Wu X, Wang Q, Li Z, Liu X, Sheng X, Zhu H, Zhang M, Xu J, Feng X, Wu B, Lv X. CD73-Adenosine A 1R Axis Regulates the Activation and Apoptosis of Hepatic Stellate Cells Through the PLC-IP 3-Ca 2+/DAG-PKC Signaling Pathway. Front Pharmacol 2022; 13:922885. [PMID: 35784730 PMCID: PMC9245432 DOI: 10.3389/fphar.2022.922885] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022] Open
Abstract
Alcohol-related liver fibrosis (ALF) is a form of alcohol-related liver disease (ALD) that generally occurs in response to heavy long-term drinking. Ecto-5'-nucleotidase (NT5E), also known as CD73, is a cytomembrane protein linked to the cell membrane via a GPI anchor that regulates the conversion of extracellular ATP to adenosine. Adenosine and its receptors are important regulators of the cellular response. Previous studies showed that CD73 and adenosine A1 receptor (A1R) were important in alcohol-related liver disease, however the exact mechanism is unclear. The aim of this study was to elucidate the role and mechanism of the CD73-A1R axis in both a murine model of alcohol and carbon tetrachloride (CCl4) induced ALF and in an in vitro model of fibrosis induced by acetaldehyde. The degree of liver injury was determined by measuring serum AST and ALT levels, H & E staining, and Masson's trichrome staining. The expression levels of fibrosis indicators and PLC-IP3-Ca2+/DAG-PKC signaling pathway were detected by quantitative real-time PCR, western blotting, ELISA, and calcium assay. Hepatic stellate cell (HSC) apoptosis was detected using the Annexin V-FITC/PI cell apoptosis detection kit. Knockdown of CD73 significantly attenuated the accumulation of α-SMA and COL1a1 damaged the histological architecture of the mouse liver induced by alcohol and CCl4. In vitro, CD73 inhibition attenuated acetaldehyde-induced fibrosis and downregulated A1R expression in HSC-T6 cells. Inhibition of CD73/A1R downregulated the expression of the PLC-IP3-Ca2+/DAG-PKC signaling pathway. In addition, silencing of CD73/A1R promoted apoptosis in HSC-T6 cells. In conclusion, the CD73-A1R axis can regulate the activation and apoptosis of HSCs through the PLC-IP3-Ca2+/DAG-PKC signaling pathway.
Collapse
Affiliation(s)
- Zhenni Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xue Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Qi Wang
- Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Zixuan Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xueqi Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiaodong Sheng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Hong Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Mengda Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Junrui Xu
- General Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaowen Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Baoming Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiongwen Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| |
Collapse
|
15
|
Pharmacological Targeting of Sphingosine Kinases Impedes HIV-1 Infection of CD4 T Cells through SAMHD1 Modulation. J Virol 2022; 96:e0009622. [PMID: 35412343 PMCID: PMC9093127 DOI: 10.1128/jvi.00096-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a sphingolipid modulator of a myriad of cellular processes, and therapeutic targeting of S1P signaling is utilized clinically to treat multiple sclerosis. We have previously shown that functional antagonism of S1P receptors reduces cell-free, cell-to-cell, and latent HIV-1 infection in primary CD4 T cells. In this work, we examined whether targeting sphingosine kinase 1 or 2 (SPHK1/2) to inhibit S1P production would prevent infection using multiple HIV-1 primary isolates and infectious molecular clones. SPHK inhibition reduced HIV transmission between primary CD4 T cells in both cell-to-cell transmission and pretreatment coculture models. Mechanistically, pharmacological inhibition of SPHK reduced susceptibility to infection primarily by downregulating phosphorylated SAMHD1 (pSAMHD1), enhancing the activity of this innate HIV-1 restriction factor. Furthermore, genetic disruption of either SPHK1 or SPHK2 by CRISPR/Cas9 reduced phosphorylation of SAMHD1, demonstrating the role of these kinases in modulation of SAMHD1 activity. The effect of SPHK inhibition on limiting HIV-1 infection in CD4 T cells was observed irrespective of the biological sex or age of the donor, with neither variable significantly influencing the effectiveness of SPHK inhibition. Our results demonstrate that targeting SPHK inhibits transmission of HIV-1 via modulation of SAMHD1 phosphorylation to decrease permissiveness to infection in CD4 T cells and suggests that therapeutic targeting of this pathway early in infection enables development of strategies to prevent establishment of infection and hinder cell-to-cell transmission of HIV-1. IMPORTANCE HIV-1 infection, once established, requires lifelong treatment due to the ability of the virus to maintain latent infection in its host and become reactivated during an interruption in antiretroviral treatment (ART). Although preventing transmission and acquisition of HIV is an important goal, no ART thus far have exploited harnessing a component of the host immune system to combat transmission of the virus. We have previously shown that inhibition of sphingosine-1-phosphate (S1P) receptors, a component of S1P signaling, reduces HIV-1 infection in human CD4 T cells. We therefore investigated inhibition of sphingosine kinases, another element of this signaling system, in this work. We found that inhibition of sphingosine kinases 1 and 2 (SPHK1/2) could reduce HIV-1 transmission, both among CD4 T cells and between macrophages and CD4 T cells. Our research therefore suggests that therapeutic targeting of SPHK or S1P receptors may aid in the development of strategies to prevent establishment and transmission of HIV-1 infection among immune cells.
Collapse
|
16
|
Wang G, Hu JQ, Liu JY, Zhang XM. Angiogenesis-Related Gene Signature-Derived Risk Score for Glioblastoma: Prospects for Predicting Prognosis and Immune Heterogeneity in Glioblastoma. Front Cell Dev Biol 2022; 10:778286. [PMID: 35372355 PMCID: PMC8971933 DOI: 10.3389/fcell.2022.778286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/15/2022] [Indexed: 12/16/2022] Open
Abstract
Background: Glioblastoma multiforme (GBM) is the most common malignant tumor in the central nervous system with poor prognosis and unsatisfactory therapeutic efficacy. Considering the high correlation between tumors and angiogenesis, we attempted to construct a more effective model with angiogenesis-related genes (ARGs) to better predict therapeutic response and prognosis. Methods: The ARG datasets were downloaded from the NCBI-Gene and Molecular Signatures Database. The gene expression data and clinical information were obtained from TCGA and CGGA databases. The differentially expressed angiogenesis-related genes (DE-ARGs) were screened with the R package “DESeq2”. Univariate Cox proportional hazards regression analysis was used to screen for ARGs related to overall survival. The redundant ARGs were removed by least absolute shrinkage and selection operator (LASSO) regression analysis. Based on the gene signature of DE-ARGs, a risk score model was established, and its effectiveness was estimated through Kaplan–Meier analysis, ROC analysis, etc. Results: A total of 626 DE-ARGs were explored between GBM and normal samples; 31 genes were identified as key DE-ARGs. Then, the risk score of ARG signature was established. Patients with high-risk score had poor survival outcomes. It was proved that the risk score could predict some medical treatments’ response, such as temozolomide chemotherapy, radiotherapy, and immunotherapy. Besides, the risk score could serve as a promising prognostic predictor. Three key prognostic genes (PLAUR, ITGA5, and FMOD) were selected and further discussed. Conclusion: The angiogenesis-related gene signature-derived risk score is a promising predictor of prognosis and treatment response in GBM and will help in making appropriate therapeutic strategies.
Collapse
Affiliation(s)
- Gang Wang
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jin-Qu Hu
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ji-Yuan Liu
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiao-Mei Zhang
- Department of Rheumatology and Immunology, ShengJing Hospital of China Medical University, Shenyang, China
- *Correspondence: Xiao-Mei Zhang,
| |
Collapse
|
17
|
Ajoolabady A, Aslkhodapasandhokmabad H, Zhou Y, Ren J. Epigenetic modification in alcohol‐related liver diseases. Med Res Rev 2022; 42:1463-1491. [DOI: 10.1002/med.21881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/21/2022] [Accepted: 01/30/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Amir Ajoolabady
- School of Pharmacy University of Wyoming College of Health Sciences Laramie Wyoming USA
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | | | - Yuan Zhou
- Department of Biomedical Informatics, School of Basic Medical Sciences Peking University Beijing China
| | - Jun Ren
- School of Pharmacy University of Wyoming College of Health Sciences Laramie Wyoming USA
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
- Department of Laboratory Medicine and Pathology University of Washington Seattle Washington USA
| |
Collapse
|
18
|
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
|
19
|
Yang LQ, Hu HY, Han Y, Tang ZY, Gao J, Zhou QY, Liu YX, Chen HS, Xu TN, Ao L, Xu Y, Che X, Jiang YB, Xu CW, Zhang XC, Jiang YX, Heger M, Wang XM, Cheng SQ, Pan WW. CpG-binding protein CFP1 promotes ovarian cancer cell proliferation by regulating BST2 transcription. Cancer Gene Ther 2022; 29:1895-1907. [PMID: 35864225 PMCID: PMC9750859 DOI: 10.1038/s41417-022-00503-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/28/2022] [Accepted: 06/28/2022] [Indexed: 02/07/2023]
Abstract
Epigenetic alterations have been functionally linked to ovarian cancer development and occurrence. The CXXC zinc finger protein 1 (CFP1) is an epigenetic regulator involved in DNA methylation and histone modification in mammalian cells. However, its role in ovarian cancer cells is unknown. Here, we show that CFP1 protein is highly expressed in human ovarian cancer tissues. Loss of CFP1 inhibited the growth of human ovarian cancer cells, promoted apoptosis, and increased senescence. CFP1 knockdown resulted in reduced levels of SETD1 (a CFP1 partner) and histone H3 trimethylation at the fourth lysine residue (H3K4me3). RNA-sequencing revealed that deletion of CFP1 resulted in mRNA reduction of bone marrow stromal cell antigen 2 (BST2). Bioinformatics analysis and chromatin immunoprecipitation showed that CFP1 binds to the promoter of BST2 and regulates its transcription directly. Overexpression of BST2 rescued the growth inhibitory effect of CFP1 loss. Furthermore, depletion of cullin-RING ubiquitin ligases 4 (CRL4) components ROC1 or CUL4A had significantly inhibited the expression of CFP1 and BST2 similar to MLN4924 treatment that blocked cullin neddylation and inactivated CRL4s. In conclusion, CFP1 promotes ovarian cancer cell proliferation and apoptosis by regulating the transcription of BST2, and the expression of CFP1 was affected by CRL4 ubiquitin ligase complex.
Collapse
Affiliation(s)
- Liu-Qing Yang
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Han-Yin Hu
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Yao Han
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Ze-Yi Tang
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Jie Gao
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Qi-Yin Zhou
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Yi-Xuan Liu
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Hao-Sa Chen
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Tu-Nan Xu
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Lei Ao
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Ying Xu
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Xuan Che
- grid.411870.b0000 0001 0063 8301Department of Anesthesiology, Jiaxing Maternity and Child Health Care Hospital, Affiliated Women and Children Hospital, Jiaxing University, Jiaxing, 314001 Zhejiang Province China
| | - Ya-Bo Jiang
- grid.73113.370000 0004 0369 1660Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200438 China
| | - Chun-Wei Xu
- grid.256112.30000 0004 1797 9307Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, 350014 Fuzhou, Fujian China
| | - Xian-Chao Zhang
- grid.411870.b0000 0001 0063 8301Institute of Information Network and Artificial Intelligence, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Yu-Xin Jiang
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Michal Heger
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China ,grid.5477.10000000120346234Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands ,grid.5645.2000000040459992XLaboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, the Netherlands
| | - Xiao-Min Wang
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Shu-Qun Cheng
- grid.73113.370000 0004 0369 1660Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200438 China ,grid.411870.b0000 0001 0063 8301G60 STI Valley Industry & Innovation Institute, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Wei-Wei Pan
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China ,grid.411870.b0000 0001 0063 8301G60 STI Valley Industry & Innovation Institute, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| |
Collapse
|
20
|
Parsons RB, Facey PD. Nicotinamide N-Methyltransferase: An Emerging Protagonist in Cancer Macro(r)evolution. Biomolecules 2021; 11:1418. [PMID: 34680055 PMCID: PMC8533529 DOI: 10.3390/biom11101418] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 12/15/2022] Open
Abstract
Nicotinamide N-methyltransferase (NNMT) has progressed from being considered merely a Phase II metabolic enzyme to one with a central role in cell function and energy metabolism. Over the last three decades, a significant body of evidence has accumulated which clearly demonstrates a central role for NNMT in cancer survival, metastasis, and drug resistance. In this review, we discuss the evidence supporting a role for NNMT in the progression of the cancer phenotype and how it achieves this by driving the activity of pro-oncogenic NAD+-consuming enzymes. We also describe how increased NNMT activity supports the Warburg effect and how it promotes oncogenic changes in gene expression. We discuss the regulation of NNMT activity in cancer cells by both post-translational modification of the enzyme and transcription factor binding to the NNMT gene, and describe for the first time three long non-coding RNAs which may play a role in the regulation of NNMT transcription. We complete the review by discussing the development of novel anti-cancer therapeutics which target NNMT and provide insight into how NNMT-based therapies may be best employed clinically.
Collapse
Affiliation(s)
- Richard B. Parsons
- Institute of Pharmaceutical Science, King’s College London, 150 Stamford Street, London SE1 9NH, UK
| | - Paul D. Facey
- Singleton Park Campus, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK;
| |
Collapse
|
21
|
Ajoolabady A, Wang S, Kroemer G, Penninger JM, Uversky VN, Pratico D, Henninger N, Reiter RJ, Bruno A, Joshipura K, Aslkhodapasandhokmabad H, Klionsky DJ, Ren J. Targeting autophagy in ischemic stroke: From molecular mechanisms to clinical therapeutics. Pharmacol Ther 2021; 225:107848. [PMID: 33823204 PMCID: PMC8263472 DOI: 10.1016/j.pharmthera.2021.107848] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/23/2021] [Accepted: 04/01/2021] [Indexed: 01/18/2023]
Abstract
Stroke constitutes the second leading cause of death and a major cause of disability worldwide. Stroke is normally classified as either ischemic or hemorrhagic stroke (HS) although 87% of cases belong to ischemic nature. Approximately 700,000 individuals suffer an ischemic stroke (IS) in the US each year. Recent evidence has denoted a rather pivotal role for defective macroautophagy/autophagy in the pathogenesis of IS. Cellular response to stroke includes autophagy as an adaptive mechanism that alleviates cellular stresses by removing long-lived or damaged organelles, protein aggregates, and surplus cellular components via the autophagosome-lysosomal degradation process. In this context, autophagy functions as an essential cellular process to maintain cellular homeostasis and organismal survival. However, unchecked or excessive induction of autophagy has been perceived to be detrimental and its contribution to neuronal cell death remains largely unknown. In this review, we will summarize the role of autophagy in IS, and discuss potential strategies, particularly, employment of natural compounds for IS treatment through manipulation of autophagy.
Collapse
Affiliation(s)
- Amir Ajoolabady
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Shuyi Wang
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; School of Medicine Shanghai University, Shanghai 200444, China
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria; Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow region 142290, Russia
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Nils Henninger
- Department of Neurology, University of Massachusetts, Worcester, Massachusetts, USA; Department of Psychiatry, University of Massachusetts, Worcester, Massachusetts, USA
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Askiel Bruno
- Department of Neurology, Medical College of Georgia, Augusta University, GA 30912, USA
| | - Kaumudi Joshipura
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Center for Clinical Research and Health Promotion, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00936-5067, Puerto Rico
| | | | - Daniel J Klionsky
- Life Sciences Institute and Departments of Molecular, Cellular and Developmental Biology and Biological Chemistry, University of Michigan, Ann Arbor 48109, USA.
| | - Jun Ren
- Department of Laboratory Medicine and Pathology, University of Washington Seattle, Seattle, WA 98195, USA; Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
22
|
Ulasov IV, Borovjagin A, Laevskaya A, Kamynina M, Timashev P, Cerchia L, Rozhkova EA. The IL13α 2R paves the way for anti-glioma nanotherapy. Genes Dis 2021; 10:89-100. [PMID: 37013057 PMCID: PMC10066331 DOI: 10.1016/j.gendis.2021.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/05/2021] [Accepted: 08/17/2021] [Indexed: 11/20/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive (grade IV) gliomas characterized by a high rate of recurrence, resistance to therapy and a grim survival prognosis. The long-awaited improvement in GBM patients' survival rates essentially depends on advances in the development of new therapeutic approaches. Recent preclinical studies show that nanoscale materials could greatly contribute to the improvement of diagnosis and management of brain cancers. In the current review, we will discuss how specific features of glioma pathobiology can be employed for designing efficient targeting approaches. Moreover, we will summarize the main evidence for the potential of the IL-13R alpha 2 receptor (IL13α2R) targeting in GBM early diagnosis and experimental therapy.
Collapse
Affiliation(s)
- Ilya V. Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
- Corresponding author.
| | - Anton Borovjagin
- Department of BioMedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Anastasia Laevskaya
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
| | - Margarita Kamynina
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
- Department of Polymers and Composites, N.N. Semenov Institute of Chemical Physics, 4 Kosygin St, Moscow 119991, Russia
- Chemistry Department, Lomonosov Moscow State University, Leninskiye Gory 1-3, Moscow 119991, Russia
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), Naples 80131, Italy
| | - Elena A. Rozhkova
- Center for Nanoscale Materials, Argonne National Laboratory, Argonne, IL 60439, USA
| |
Collapse
|
23
|
Qin Y, Wang Y, Zhao H, Yang Z, Kang Y. Aberrant miRNA-mRNA regulatory network in polycystic ovary syndrome is associated with markers of insulin sensitivity and inflammation. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1405. [PMID: 34733957 PMCID: PMC8506717 DOI: 10.21037/atm-21-1288] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common multifactorial metabolic and endocrine disorder in women of reproductive age. Increasingly, evidence indicates that the microRNA (miRNA)-mRNA axis contributes to the development of PCOS. METHODS To investigate the molecular mechanisms through which miRNA-mRNA expression affects hyperandrogenism, ovarian tissue samples from prenatally androgenized (PNA) mice were subjected to miRNA-seq and RNA-seq analysis. Analyses were performed to identify differentially expressed microRNAs (DEmiRs) and differentially expressed genes (DEGs). In combination with our previous data obtained from clinical samples, we have performed an integrated miRNA-mRNA analysis of PNA mice and granulosa cells (GCs) from patients with PCOS. The changes in expression were validated by RT-qPCR in more mouse models and clinical samples. RESULTS In total, 3,432 genes and 16 miRNAs were differentially expressed in PNA mice compared with the control mice. We investigated the regulation pattern of miRNAs-mRNAs and observed a total of 12 miRNA-mRNA pairs involved in negative regulation. Functional analysis concentrated on insulin resistance, the T cell receptor signaling pathway, and other inflammation-related pathways. Verification of these results by RT-qPCR showed that the expression of miR-106-5p and miR-155-5p in clinical GCs was consistent with that in PNA mice. After predicting the target genes of miR-106-5p and miR-155-5p and performing negative regulation analysis, six target genes were obtained in GCs. The integration analysis showed that the network of miR-106-5p/miR-155-5p targets was mostly concentrated in pathway related to insulin resistance and inflammation. In addition, the upregulation of the inflammatory genes Il18/IL18 and Socs3/SOCS3 was validated in the PNA mice and GCs from patients compared with the appropriate control sample. The in vitro experiments showed that the regulatory relationship observed may be related to the direct stimulation of DHT. CONCLUSIONS Our results showed that the miRNA-mRNA regulatory network in PCOS was associated with markers of insulin sensitivity and inflammation. Our study provides a new genetic basis and novel insight regarding the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Yulan Qin
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, China
| | - Yuhuan Wang
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Zhao
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaoxia Yang
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, China
| | | |
Collapse
|
24
|
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
|
25
|
Sattar RSA, Sumi MP, Nimisha, Apurva, Kumar A, Sharma AK, Ahmad E, Ali A, Mahajan B, Saluja SS. S1P signaling, its interactions and cross-talks with other partners and therapeutic importance in colorectal cancer. Cell Signal 2021; 86:110080. [PMID: 34245863 DOI: 10.1016/j.cellsig.2021.110080] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/25/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
Sphingosine-1-Phosphate (S1P) plays an important role in normal physiology, inflammation, initiation and progression of cancer. Deregulation of S1P signaling causes aberrant proliferation, affects survival, leads to angiogenesis and metastasis. Sphingolipid rheostat is crucial for cellular homeostasis. Discrepancy in sphingolipid metabolism is linked to cancer and drug insensitivity. Owing to these diverse functions and being a potent mediator of tumor growth, S1P signaling might be a suitable candidate for anti-tumor therapy or combination therapy. In this review, with a focus on colorectal cancer we have summarized the interacting partners of S1P signaling pathway, its therapeutic approaches along with the contribution of S1P signaling to various cancer hallmarks.
Collapse
Affiliation(s)
- Real Sumayya Abdul Sattar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Mamta P Sumi
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Apurva
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Arun Kumar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Abhay Kumar Sharma
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Ejaj Ahmad
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Asgar Ali
- Department of Biochemistry, All India Institute of Medical Science (AIIMS), Patna, Bihar, India
| | - Bhawna Mahajan
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India; Department of Biochemistry, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India; Department of GI Surgery, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India.
| |
Collapse
|
26
|
Ajoolabady A, Aslkhodapasandhokmabad H, Henninger N, Demillard LJ, Nikanfar M, Nourazarian A, Ren J. Targeting autophagy in neurodegenerative diseases: From molecular mechanisms to clinical therapeutics. Clin Exp Pharmacol Physiol 2021; 48:943-953. [PMID: 33752254 PMCID: PMC8204470 DOI: 10.1111/1440-1681.13500] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023]
Abstract
Many neurodegenerative diseases are associated with pathological aggregation of proteins in neurons. Autophagy is a natural self-cannibalization process that can act as a powerful mechanism to remove aged and damaged organelles as well as protein aggregates. It has been shown that promoting autophagy can attenuate or delay neurodegeneration by removing protein aggregates. In this paper, we will review the role of autophagy in Alzheimer's disease (AD), Parkinson's Disease (PD), and Huntington's Disease (HD) and discuss opportunities and challenges of targeting autophagy as a potential therapeutic avenue for treatment of these common neurodegenerative diseases.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Nils Henninger
- Department of Neurology, University of Massachusetts, Worcester, MA 01655, USA
- Department of Psychiatry, University of Massachusetts, Worcester, MA 01655, USA
| | - Laurie J. Demillard
- School of Pharmacy, University of Wyoming College of Health Sciences, Laramie, WY 82071 USA
| | - Masoud Nikanfar
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jun Ren
- School of Pharmacy, University of Wyoming College of Health Sciences, Laramie, WY 82071 USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195 USA
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
27
|
Saito K, Ito M, Chiba T, Jia H, Kato H. A Comparison of Gene Expression Profiles of Rat Tissues after Mild and Short-Term Calorie Restrictions. Nutrients 2021; 13:2277. [PMID: 34209243 PMCID: PMC8308279 DOI: 10.3390/nu13072277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/14/2022] Open
Abstract
Many studies have shown the beneficial effects of calorie restriction (CR) on rodents' aging; however, the molecular mechanism explaining these beneficial effects is still not fully understood. Previously, we conducted transcriptomic analysis on rat liver with short-term and mild-to-moderate CR to elucidate its early response to such diet. Here, we expanded transcriptome analysis to muscle, adipose tissue, intestine, and brain and compared the gene expression profiles of these multiple organs and of our previous dataset. Several altered gene expressions were found, some of which known to be related to CR. Notably, the commonly regulated genes by CR include nicotinamide phosphoribosyltransferase and heat shock protein 90, which are involved in declining the aging process and thus potential therapeutic targets for aging-related diseases. The data obtained here provide information on early response markers and key mediators of the CR-induced delay in aging as well as on age-associated pathological changes in mammals.
Collapse
Affiliation(s)
- Kenji Saito
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| | - Maiko Ito
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
- Biomedical Gerontology Laboratory, Faculty of Human Sciences, Waseda University, 2-579-1 Mikajima, Tokorozawa, Saitama 359-1164, Japan;
| | - Takuya Chiba
- Biomedical Gerontology Laboratory, Faculty of Human Sciences, Waseda University, 2-579-1 Mikajima, Tokorozawa, Saitama 359-1164, Japan;
| | - Huijuan Jia
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| | - Hisanori Kato
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| |
Collapse
|
28
|
Kolar EA, Shi X, Clay EM, Moser AB, Lal B, Nirujogi RS, Pandey A, Bandaru VVR, Laterra J, Pei Z, Watkins PA. Very long-chain acyl-CoA synthetase 3 mediates onco-sphingolipid metabolism in malignant glioma. ACTA ACUST UNITED AC 2021; 9. [PMID: 34395855 DOI: 10.18103/mra.v9i5.2433] [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] [Indexed: 01/11/2023]
Abstract
Gliomas are the largest category of primary malignant brain tumors in adults, and glioblastomas account for nearly half of malignant gliomas. Glioblastomas are notoriously aggressive and drug-resistant, with a very poor 5 year survival rate of about 5%. New approaches to treatment are thus urgently needed. We previously identified an enzyme of fatty acid metabolism, very long-chain acyl-CoA synthetase 3 (ACSVL3), as a potential therapeutic target in glioblastoma. Using the glioblastoma cell line U87MG, we created a cell line with genomic deletion of ACSVL3 (U87-KO) and investigated potential mechanisms to explain how this enzyme supports the malignant properties of glioblastoma cells. Compared to U87MG cells, U87-KO cells grew slower and assumed a more normal morphology. They produced fewer, and far smaller, subcutaneous xenografts in nude mice. Acyl-CoA synthetases, including ACSVL3, convert fatty acids to their acyl-CoA derivatives, allowing participation in diverse downstream lipid pathways. We examined the effect of ACSVL3 depletion on several such pathways. Fatty acid degradation for energy production was not affected in U87-KO cells. Fatty acid synthesis, and incorporation of de novo synthesized fatty acids into membrane phospholipids needed for rapid tumor cell growth, was not significantly affected by lack of ACSVL3. In contrast, U87-KO cells exhibited evidence of altered sphingolipid metabolism. Levels of ceramides containing 18-22 carbon fatty acids were significantly lower in U87-KO cells. This paralleled the fatty acid substrate specificity profile of ACSVL3. The rate of incorporation of stearate, an 18-carbon saturated fatty acid, into ceramides was reduced in U87-KO cells, and proteomics revealed lower abundance of ceramide synthesis pathway enzymes. Sphingolipids, including gangliosides, are functional constituents of lipid rafts, membrane microdomains thought to be organizing centers for receptor-mediated signaling. Both raft morphology and ganglioside composition were altered by deficiency of ACSVL3. Finally, levels of sphingosine-1-phosphate, a sphingolipid signaling molecule, were reduced in U87-KO cells. We conclude that ACSVL3 supports the malignant behavior of U87MG cells, at least in part, by altering cellular sphingolipid metabolism.
Collapse
Affiliation(s)
- Elizabeth A Kolar
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205
| | - Xiaohai Shi
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205
| | - Emily M Clay
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205
| | - Ann B Moser
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Bachchu Lal
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Raja Sekhar Nirujogi
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | | | - John Laterra
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Zhengtong Pei
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Paul A Watkins
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
29
|
Wang Y, Qiu L, Chen Y, Zhang X, Yang P, Xu F. Screening and Identification of Four Prognostic Genes Related to Immune Infiltration and G-Protein Coupled Receptors Pathway in Lung Adenocarcinoma. Front Oncol 2021; 10:622251. [PMID: 33628734 PMCID: PMC7897677 DOI: 10.3389/fonc.2020.622251] [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: 10/28/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a common malignant tumor with the highest morbidity and mortality worldwide. The degree of tumor immune infiltration and clinical prognosis depend on immune-related genes, but their interaction with the tumor immune microenvironment, the specific mechanism driving immune infiltration and their prognostic value are still not very clear. Therefore, the aim of this work was focused on the elucidation of these unclear aspects. Methods TCGA LUAD samples were divided into three immune infiltration subtypes according to the single sample gene set enrichment analysis (ssGSEA), in which the associated gene modules and hub genes were screened by weighted correlation network analysis (WGCNA). Four key genes related to immune infiltration were found and screened by differential expression analysis, univariate prognostic analysis, and Lasso-COX regression, and their PPI network was constructed. Finally, a Nomogram model based on the four genes and tumor stages was constructed and confirmed in two GEO data sets. Results Among the three subtypes—high, medium, and low immune infiltration subtype—the survival rate of the patients in the high one was higher than the rate in the other two subtypes. The four key genes related to LUAD immune infiltration subtypes were CD69, KLRB1, PLCB2, and P2RY13. The PPI network revealed that the downstream genes of the G-protein coupled receptors (GPCRs) pathway were activated by these four genes through the S1PR1. The risk score signature based on these four genes could distinguish high and low-risk LUAD patients with different prognosis. The Nomogram constructed by risk score and clinical tumor stage showed a good ability to predict the survival rate of LUAD patients. The universality and robustness of the Nomogram was confirmed by two GEO datasets. Conclusions The prognosis of LUAD patients could be predicted by the constructed risk score signature based on the four genes, making this score a potential independent biomarker. The screening, identification, and analysis of these four genes could contribute to the understanding of GPCRs and LUAD immune infiltration, thus guiding the formulation of more effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Yan Wang
- Department of Emergency Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Liwei Qiu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yu Chen
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xia Zhang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Peng Yang
- Department of Emergency Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng Xu
- Department of Emergency Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
30
|
Matsumoto Y, Kudelka MR, Hanes MS, Lehoux S, Dutta S, Jones MB, Stackhouse KA, Cervoni GE, Heimburg-Molinaro J, Smith DF, Ju T, Chaikof EL, Cummings RD. Identification of Tn antigen O-GalNAc-expressing glycoproteins in human carcinomas using novel anti-Tn recombinant antibodies. Glycobiology 2020; 30:282-300. [PMID: 31742337 DOI: 10.1093/glycob/cwz095] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/08/2019] [Accepted: 11/02/2019] [Indexed: 12/14/2022] Open
Abstract
The Tn antigen is a neoantigen abnormally expressed in many human carcinomas and expression correlates with metastasis and poor survival. To explore its biomarker potential, new antibodies are needed that specifically recognize this antigen in tumors. Here we generated two recombinant antibodies to the Tn antigen, Remab6 as a chimeric human IgG1 antibody and ReBaGs6 as a murine IgM antibody and characterized their specificities using multiple biochemical and biological approaches. Both Remab6 and ReBaGs6 recognize clustered Tn structures, but most importantly do not recognize glycoforms of human IgA1 that contain potential cross-reactive Tn antigen structures. In flow cytometry and immunofluorescence analyses, Remab6 recognizes human cancer cell lines expressing the Tn antigen, but not their Tn-negative counterparts. In immunohistochemistry (IHC), Remab6 stains many human cancers in tissue array format but rarely stains normal tissues and then mostly intracellularly. We used these antibodies to identify several unique Tn-containing glycoproteins in Tn-positive Colo205 cells, indicating their utility for glycoproteomics in future biomarker studies. Thus, recombinant Remab6 and ReBaGs6 are useful for biochemical characterization of cancer cells and IHC of tumors and represent promising tools for Tn biomarker discovery independently of recognition of IgA1.
Collapse
Affiliation(s)
- Yasuyuki Matsumoto
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11090, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Matthew R Kudelka
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11090, 3 Blackfan Circle, Boston, MA 02115, USA.,Department of Biochemistry, Emory University School of Medicine, 1518 Clifton Rd, Atlanta, GA 30322, USA
| | - Melinda S Hanes
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11090, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Sylvain Lehoux
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11090, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Sucharita Dutta
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11090, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Mark B Jones
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11090, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Kathryn A Stackhouse
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11090, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Gabrielle E Cervoni
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11090, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Jamie Heimburg-Molinaro
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11090, 3 Blackfan Circle, Boston, MA 02115, USA
| | - David F Smith
- Department of Biochemistry, Emory University School of Medicine, 1518 Clifton Rd, Atlanta, GA 30322, USA
| | - Tongzhong Ju
- Department of Biochemistry, Emory University School of Medicine, 1518 Clifton Rd, Atlanta, GA 30322, USA.,Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Bldg 52/72, Room 2120, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Elliot L Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11090, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11090, 3 Blackfan Circle, Boston, MA 02115, USA.,Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11087, 3 Blackfan Circle, Boston, MA 02115, USA
| |
Collapse
|
31
|
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: 16] [Impact Index Per Article: 4.0] [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
|
32
|
Wu ZL, Deng YJ, Zhang GZ, Ren EH, Yuan WH, Xie QQ. Development of a novel immune-related genes prognostic signature for osteosarcoma. Sci Rep 2020; 10:18402. [PMID: 33110201 PMCID: PMC7591524 DOI: 10.1038/s41598-020-75573-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
Immune-related genes (IRGs) are responsible for osteosarcoma (OS) initiation and development. We aimed to develop an optimal IRGs-based signature to assess of OS prognosis. Sample gene expression profiles and clinical information were downloaded from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) and Genotype-Tissue Expression (GTEx) databases. IRGs were obtained from the ImmPort database. R software was used to screen differentially expressed IRGs (DEIRGs) and functional correlation analysis. DEIRGs were analyzed by univariate Cox regression and iterative LASSO Cox regression analysis to develop an optimal prognostic signature, and the signature was further verified by independent cohort (GSE39055) and clinical correlation analysis. The analyses yielded 604 DEIRGs and 10 hub IRGs. A prognostic signature consisting of 13 IRGs was constructed, which strikingly correlated with OS overall survival and distant metastasis (p < 0.05, p < 0.01), and clinical subgroup showed that the signature's prognostic ability was independent of clinicopathological factors. Univariate and multivariate Cox regression analyses also supported its prognostic value. In conclusion, we developed an IRGs signature that is a prognostic indicator in OS patients, and the signature might serve as potential prognostic indicator to identify outcome of OS and facilitate personalized management of the high-risk patients.
Collapse
Affiliation(s)
- Zuo-Long Wu
- Guanghe Traditional Chinese and Western Medicine Hospital, Lanzhou, 730000, Gansu, China
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Ya-Jun Deng
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Guang-Zhi Zhang
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - En-Hui Ren
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, No.29 Tongren Road, Xining, 810000, Qinghai, China
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Wen-Hua Yuan
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Qi-Qi Xie
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, No.29 Tongren Road, Xining, 810000, Qinghai, China.
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
33
|
Fang T, Jiang YX, Chen L, Huang L, Tian XH, Zhou YD, Nagle DG, Zhang DD. Coix Seed Oil Exerts an Anti-Triple-Negative Breast Cancer Effect by Disrupting miR-205/S1PR1 Axis. Front Pharmacol 2020; 11:529962. [PMID: 33101013 PMCID: PMC7556270 DOI: 10.3389/fphar.2020.529962] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Coix Seed Oil (CSO) possesses a wide range of pharmacological activities. Kanglaite Injection, a commercial product of CSO, has been used clinically as an anticancer drug in China for decades. However, its molecular mechanisms on triple-negative breast cancer (TNBC) remains to be elucidated. In this study, the effect of CSO was evaluated on murine TNBC 4T1 cells and the orthotopic tumor-bearing mouse model and underlying mechanisms were explored. CSO suppressed cell proliferation, colony formation in vitro, and tumor growth in vivo. miR-205-5p was substantially altered in CSO treated tumor tissues compared to the control group by miRNA-sequencing analysis. Sphingomyelin metabolism (SM) decreased in serum in model group compared to the control group, while it increased by CSO administration by lipid metabolomics analysis. The expression of sphingosine 1 phosphate receptor 1 (S1PR1), the critical effector of SM, was downregulated upon CSO treatment. Mechanically, miRNA-205 directly targeted S1PR1 to regulate SM and cell proliferation. CSO reduced the expression of S1PR1, cyclinD1, and phosphorylation levels of STAT3, MAPK, and AKT while upregulated p27. These results revealed that CSO exerted an anti-TNBC effect via the miR-205/S1PR1 axis to regulate sphingomyelin metabolism, and the downstream STAT3/MAPK/AKT signal pathways were partly involved.
Collapse
Affiliation(s)
- Ting Fang
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yi-Xin Jiang
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Long Chen
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Huang
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin-Hui Tian
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu-Dong Zhou
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Chemistry and Biochemistry, College of Liberal Arts, University of Mississippi, University, Misissippi, MS, United States
| | - Dale G Nagle
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences (RIPS), School of Pharmacy, University of Mississippi, University, Mississippi, MS, United States
| | - Dan-Dan Zhang
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
34
|
Hawkins CC, Ali T, Ramanadham S, Hjelmeland AB. Sphingolipid Metabolism in Glioblastoma and Metastatic Brain Tumors: A Review of Sphingomyelinases and Sphingosine-1-Phosphate. Biomolecules 2020; 10:E1357. [PMID: 32977496 PMCID: PMC7598277 DOI: 10.3390/biom10101357] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/18/2020] [Accepted: 09/20/2020] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma (GBM) is a primary malignant brain tumor with a dismal prognosis, partially due to our inability to completely remove and kill all GBM cells. Rapid tumor recurrence contributes to a median survival of only 15 months with the current standard of care which includes maximal surgical resection, radiation, and temozolomide (TMZ), a blood-brain barrier (BBB) penetrant chemotherapy. Radiation and TMZ cause sphingomyelinases (SMase) to hydrolyze sphingomyelins to generate ceramides, which induce apoptosis. However, cells can evade apoptosis by converting ceramides to sphingosine-1-phosphate (S1P). S1P has been implicated in a wide range of cancers including GBM. Upregulation of S1P has been linked to the proliferation and invasion of GBM and other cancers that display a propensity for brain metastasis. To mediate their biological effects, SMases and S1P modulate signaling via phospholipase C (PLC) and phospholipase D (PLD). In addition, both SMase and S1P may alter the integrity of the BBB leading to infiltration of tumor-promoting immune populations. SMase activity has been associated with tumor evasion of the immune system, while S1P creates a gradient for trafficking of innate and adaptive immune cells. This review will explore the role of sphingolipid metabolism and pharmacological interventions in GBM and metastatic brain tumors with a focus on SMase and S1P.
Collapse
Affiliation(s)
- Cyntanna C. Hawkins
- Department of Cell, Developmental, and Integrative Biology, University of Birmingham at Alabama, Birmingham, AL 35233, USA; (C.C.H.); (S.R.)
| | - Tomader Ali
- Research Department, Imperial College London Diabetes Centre, Abu Dhabi P.O. Box 48338, UAE;
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology, University of Birmingham at Alabama, Birmingham, AL 35233, USA; (C.C.H.); (S.R.)
- Comprehensive Diabetes Center, University of Birmingham at Alabama, Birmingham, AL 35294, USA
| | - Anita B. Hjelmeland
- Department of Cell, Developmental, and Integrative Biology, University of Birmingham at Alabama, Birmingham, AL 35233, USA; (C.C.H.); (S.R.)
| |
Collapse
|
35
|
Resop RS, Fromentin R, Newman D, Rigsby H, Dubrovsky L, Bukrinsky M, Chomont N, Bosque A. Fingolimod inhibits multiple stages of the HIV-1 life cycle. PLoS Pathog 2020; 16:e1008679. [PMID: 32790802 PMCID: PMC7425850 DOI: 10.1371/journal.ppat.1008679] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
Antiretroviral drugs that target various stages of the Human Immunodeficiency Virus (HIV) life cycle have been effective in curbing the AIDS epidemic. However, drug resistance, off-target effects of antiretroviral therapy (ART), and varying efficacy in prevention underscore the need to develop novel and alternative therapeutics. In this study, we investigated whether targeting the signaling molecule Sphingosine-1-phosphate (S1P) would inhibit HIV-1 infection and generation of the latent reservoir in primary CD4 T cells. We show that FTY720 (Fingolimod), an FDA-approved functional antagonist of S1P receptors, blocks cell-free and cell-to-cell transmission of HIV and consequently reduces detectable latent virus. Mechanistically, FTY720 impacts the HIV-1 life cycle at two levels. Firstly, FTY720 reduces the surface density of CD4, thereby inhibiting viral binding and fusion. Secondly, FTY720 decreases the phosphorylation of the innate HIV restriction factor SAMHD1 which is associated with reduced levels of total and integrated HIV, while reducing the expression of Cyclin D3. In conclusion, targeting the S1P pathway with FTY720 could be a novel strategy to inhibit HIV replication and reduce the seeding of the latent reservoir.
Collapse
Affiliation(s)
- Rachel S. Resop
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, D.C., United States of America
| | - Rémi Fromentin
- Centre de recherche du CHUM and Department of microbiology, infectiology and immunology, Université de Montréal, Montreal, Canada
| | - Daniel Newman
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, D.C., United States of America
| | - Hawley Rigsby
- Centre de recherche du CHUM and Department of microbiology, infectiology and immunology, Université de Montréal, Montreal, Canada
| | - Larisa Dubrovsky
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, D.C., United States of America
| | - Michael Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, D.C., United States of America
| | - Nicolas Chomont
- Centre de recherche du CHUM and Department of microbiology, infectiology and immunology, Université de Montréal, Montreal, Canada
| | - Alberto Bosque
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, D.C., United States of America
- * E-mail:
| |
Collapse
|
36
|
Sudhadevi T, Ha AW, Ebenezer DL, Fu P, Putherickal V, Natarajan V, Harijith A. Advancements in understanding the role of lysophospholipids and their receptors in lung disorders including bronchopulmonary dysplasia. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158685. [PMID: 32169655 PMCID: PMC7206974 DOI: 10.1016/j.bbalip.2020.158685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/25/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a devastating chronic neonatal lung disease leading to serious adverse consequences. Nearly 15 million babies are born preterm accounting for >1 in 10 births globally. The aetiology of BPD is multifactorial and the survivors suffer lifelong respiratory morbidity. Lysophospholipids (LPL), which include sphingosine-1-phosphate (S1P), and lysophosphatidic acid (LPA) are both naturally occurring bioactive lipids involved in a variety of physiological and pathological processes such as cell survival, death, proliferation, migration, immune responses and vascular development. Altered LPL levels have been observed in a number of lung diseases including BPD, which underscores the importance of these signalling lipids under normal and pathophysiological situations. Due to the paucity of information related to LPLs in BPD, most of the ideas related to BPD and LPL are speculative. This article is intended to promote discussion and generate hypotheses, in addition to the limited review of information related to BPD already established in the literature.
Collapse
Affiliation(s)
- Tara Sudhadevi
- Department of Pediatrics, University of Illinois, Chicago, IL, United States of America
| | - Alison W Ha
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL, United States of America
| | - David L Ebenezer
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL, United States of America
| | - Panfeng Fu
- Department of Pharmacology, University of Illinois, Chicago, IL, United States of America
| | - Vijay Putherickal
- Department of Pharmacology, University of Illinois, Chicago, IL, United States of America
| | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois, Chicago, IL, United States of America; Department of Medicine, University of Illinois, Chicago, IL, United States of America
| | - Anantha Harijith
- Department of Pediatrics, University of Illinois, Chicago, IL, United States of America; Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL, United States of America; Department of Pharmacology, University of Illinois, Chicago, IL, United States of America.
| |
Collapse
|
37
|
Magaye RR, Savira F, Hua Y, Xiong X, Huang L, Reid C, Flynn B, Kaye D, Liew D, Wang BH. Exogenous dihydrosphingosine 1 phosphate mediates collagen synthesis in cardiac fibroblasts through JAK/STAT signalling and regulation of TIMP1. Cell Signal 2020; 72:109629. [PMID: 32278008 DOI: 10.1016/j.cellsig.2020.109629] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 02/03/2023]
Abstract
Cardiac fibrosis and myocyte hypertrophy are hallmarks of the cardiac remodelling process in cardiomyopathies such as heart failure (HF). Dyslipidemia or dysregulation of lipids contribute to HF. The dysregulation of high density lipoproteins (HDL) could lead to altered levels of other lipid metabolites that are bound to it such as sphingosine-1- phosphate (S1P). Recently, it has been shown that S1P and its analogue dihydrosphingosine-1-phosphate (dhS1P) are bound to HDL in plasma. The effects of dhS1P on cardiac cells have been obscure. In this study, we show that extracellular dhS1P is able to increase collagen synthesis in neonatal rat cardiac fibroblasts (NCFs) and cause hypertrophy of neonatal cardiac myocytes (NCMs). The janus kinase/signal transducer and activator (JAK/STAT) signalling pathway was involved in the increased collagen synthesis by dhS1P, through sustained increase of tissue inhibitor of matrix metalloproteinase 1 (TIMP1). Extracellular dhS1P increased phosphorylation levels of STAT1 and STAT3 proteins, also caused an early increase in gene expression of transforming growth factor-β (TGFβ), and sustained increase in TIMP1. Inhibition of JAKs led to inhibition of TIMP1 and TGFβ gene and protein expression. We also show that dhS1P is able to cause NCM hypertrophy through S1P-receptor-1 (S1PR1) signalling which is opposite to that of its analogue, S1P. Taken together, our results show that dhS1P increases collagen synthesis in cardiac fibroblasts causing fibrosis through dhS1P-JAK/STAT-TIMP1 signalling.
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 Flynn
- Australian Translational Medicinal Chemistry Facility, Medicinal Chemistry, 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
|
38
|
Zou Y, Hu J, Huang W, Ye S, Han F, Du J, Shao M, Guo R, Lin J, Zhao Y, Xiong Y, Wang X. Non-Mitogenic Fibroblast Growth Factor 1 Enhanced Angiogenesis Following Ischemic Stroke by Regulating the Sphingosine-1-Phosphate 1 Pathway. Front Pharmacol 2020; 11:59. [PMID: 32194396 PMCID: PMC7063943 DOI: 10.3389/fphar.2020.00059] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/22/2020] [Indexed: 12/11/2022] Open
Abstract
Ischemic strokes account for about 80% of all strokes and are associated with a high risk of mortality. Angiogenesis of brain microvascular endothelial cells may contribute to functional restoration following ischemia. Fibroblast growth factor 1 (FGF1), a member of FGF superfamily, involved in embryonic development, angiogenesis, wound healing, and neuron survival. However, the mitogenic activity of FGF1 is known to contribute to several human pathologies, thereby questioning the safety of its clinical applications. Here, we explored the effects and mechanism of action of non-mitogenic FGF1 (nmFGF1) on angiogenesis in mice after ischemia stroke and an oxygen-glucose deprivation (OGD)-induced human brain microvascular endothelial cells (HBMECs) injury model. We found that intranasal administration nmFGF1 significantly promoted angiogenesis in mice after stroke, and significantly increased the formation of matrigel tube and promoted scratch migration in a dose-dependent manner in OGD-induced HBMECs in vitro. However, the co-administration of an FGF receptor 1 (FGFR1)-specific inhibitor PD173074 significantly reversed the effects of nmFGF1 in vitro, suggesting that nmFGF1 functions via FGFR1 activation. Moreover, nmFGF1 activated sphingosine-1-phosphate receptor 1 (S1PR1, S1P1) in mice after stroke in vivo. S1P1 protein antagonist VPC23019 and agonist FTY720 were used to confirm that nmFGF1 promotes angiogenesis in vitro partially through the S1P1 pathway. OGD induced downregulation of S1P1 expression. The S1P1 antagonist VPC23019 blocked the stimulatory effects of nmFGF1, whereas the S1P1 agonist FTY720 exerted effects comparable with those of nmFGF1. Furthermore, PD173074 reversed the effect of nmFGF1 on upregulating S1P1 signaling. In conclusion, nmFGF1 enhanced angiogenesis in mice following stroke and OGD-induced HBMECs through S1P1 pathway regulation mediated via FGFR1 activation. This new discovery suggests the potential therapeutic role of nmFGF1 for the treatment of ischemic strokes.
Collapse
Affiliation(s)
- Yuchi Zou
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenting Huang
- School of the First Clinical Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Shasha Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fanyi Han
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingting Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Mingjie Shao
- School of the First Clinical Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Ruili Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingjing Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yeli Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou, China
| | - Ye Xiong
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xue Wang
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|