1
|
Clark AJ, Singh R, Leonis RL, Stahlberg EA, Clark ZS, Lillard JW. Gene Co-Expression Network Analysis Associated with Endometrial Cancer Tumorigenesis and Survival Outcomes. Int J Mol Sci 2024; 25:12356. [PMID: 39596419 PMCID: PMC11594472 DOI: 10.3390/ijms252212356] [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: 10/14/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Endometrial cancer (EC) presents a substantial health challenge, with increasing incidence and mortality rates. Despite advances in diagnosis and treatment, understanding the molecular underpinnings of EC progression remains unknown. In this study, we conducted a comprehensive investigation utilizing The Cancer Genome Atlas (TCGA-UCEC n = 588) data to analyze gene co-expression patterns, elucidate biological process pathways, and identify potential prognostic and diagnostic biomarkers for EC, using weighted gene co-expression network analysis (WGCNA), differential gene expression, survival analysis, and functional analysis, respectively. We determined that the Green module (M5) was significantly correlated with patient survival. Functional analysis of the genes in module M5 indicates involvement in cell cycle regulation, mitotic spindle assembly, and intercellular signaling. TPX2, BUB1, and ESPL1 were among the top differentially expressed genes in the Green module, suggesting their involvement in critical pathways that contribute to disease progression and patient survival outcomes. The biological and clinical assessments of our findings provide an understanding of the molecular landscape of EC and identified several potential prognostic markers for patient risk stratification and treatment selection.
Collapse
Affiliation(s)
- Alexis J. Clark
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (A.J.C.); (R.S.)
| | - Rajesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (A.J.C.); (R.S.)
| | - Regina L. Leonis
- Department of Obstetrics & Gynecology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Eric A. Stahlberg
- Cancer Data Science Initiatives, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA;
| | - Zachary S. Clark
- Department of Information Technology, Clayton State University, Morrow, GA 30260, USA;
| | - James W. Lillard
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (A.J.C.); (R.S.)
| |
Collapse
|
2
|
Böcker M, Chatziioannou E, Niessner H, Hirn C, Busch C, Ikenberg K, Kalbacher H, Handgretinger R, Sinnberg T. Ecto-NOX Disulfide-Thiol Exchanger 2 (ENOX2/tNOX) Is a Potential Prognostic Marker in Primary Malignant Melanoma and May Serve as a Therapeutic Target. Int J Mol Sci 2024; 25:11853. [PMID: 39519404 PMCID: PMC11545956 DOI: 10.3390/ijms252111853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/10/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
With an increasing incidence of malignant melanoma, new prognostic biomarkers for clinical decision making have become more important. In this study, we evaluated the role of ecto-NOX disulfide-thiol exchanger 2 (ENOX2/tNOX), a cancer- and growth-associated protein, in the prognosis and therapy of primary malignant melanoma. We conducted a tissue microarray analysis of immunohistochemical ENOX2 protein expression and The Cancer Genome Atlas (TCGA) ENOX2 RNA expression analysis, as well as viability assays and Western blots of melanoma cell lines treated with the ENOX2 inhibitor phenoxodiol (PXD) and BRAF inhibitor (BRAFi) vemurafenib. We discovered that high ENOX2 expression is associated with decreased overall (OS), disease-specific (DSS) and metastasis-free survival (MFS) in primary melanoma (PM) and a reduction in electronic tumor-infiltrating lymphocytes (eTILs). A gradual rise in ENOX2 expression was found with an increase in malignant potential from benign nevi (BNs) via PMs to melanoma metastases (MMs), as well as with an increasing tumor thickness and stage. These results highlight the important role of ENOX2 in cancer growth, progression and metastasis. The ENOX2 expression was not limited to malignant cell lines but could also be found in keratinocytes, fibroblasts and melanocytes. The viability of melanoma cell lines could be inhibited by PXD. A reduced induction of phospho-AKT under PXD could prevent the development of acquired BRAFi resistance. In conclusion, ENOX2 may serve as a potential prognostic marker and therapeutic target in malignant melanoma.
Collapse
Affiliation(s)
- Matti Böcker
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany (E.C.); (H.N.); (C.H.)
- Department of Urology and Pediatric Urology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Eftychia Chatziioannou
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany (E.C.); (H.N.); (C.H.)
| | - Heike Niessner
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany (E.C.); (H.N.); (C.H.)
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany
| | - Constanze Hirn
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany (E.C.); (H.N.); (C.H.)
| | - Christian Busch
- Dermatologie zum Delfin, Stadthausstraße 12, 8400 Winterthur, Switzerland;
| | - Kristian Ikenberg
- Institute of Clinical Pathology, University Hospital Zuerich, Schmelzbergstraße 12, 8091 Zuerich, Switzerland;
| | - Hubert Kalbacher
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Elfriede-Aulhorn-Straße 8, 72076 Tuebingen, Germany;
| | - Rupert Handgretinger
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tuebingen, Hoppe-Seyler-Straße 1, 72076 Tuebingen, Germany;
| | - Tobias Sinnberg
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany (E.C.); (H.N.); (C.H.)
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany
- Department of Dermatology, Venereology and Allergology, Charité-Universitaetsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
3
|
Wang D, Xu R, Wang Z. Protective Role of Sphingosine-1-Phosphate During Radiation-Induced Testicular Injury. Antioxidants (Basel) 2024; 13:1322. [PMID: 39594464 PMCID: PMC11591009 DOI: 10.3390/antiox13111322] [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: 10/07/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
The impact of ionizing radiation on the male reproductive system is gaining increasing attention, particularly when it comes to testicular damage, which may result in decreased sperm quality and hormonal imbalances. Finding effective protective measures to mitigate testicular damage caused by radiation has become a focal point in the biomedical field. S1P, an essential biological signaling molecule, has garnered significant interest due to its multiple roles in regulating cellular functions and its protective effects against radiation-induced testicular injury. S1P not only effectively reduces the generation of ROS induced by radiation but also alleviates oxidative stress by enhancing the activity of antioxidant enzymes. Furthermore, S1P inhibits radiation-induced cell apoptosis by regulating the expression of anti-apoptotic and pro-apoptotic proteins. Additionally, S1P alleviates radiation-induced inflammation by inhibiting the production of inflammatory factors, thereby further protecting testicular tissue. In summary, S1P effectively reduces radiation-induced testicular damage through multiple mechanisms, offering a promising therapeutic approach to safeguard male reproductive health. Future research should explore the specific mechanisms of action and clinical application potential of S1P, aiming to contribute significantly to the prevention and treatment of radiation damage.
Collapse
Affiliation(s)
- Defan Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen 361102, China;
| | - Renfeng Xu
- Fujian Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China;
| | - Zhengchao Wang
- Fujian Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China;
| |
Collapse
|
4
|
Vettorazzi M, Díaz I, Angelina E, Salido S, Gutierrez L, Alvarez SE, Cobo J, Enriz RD. Second generation of pyrimidin-quinolone hybrids obtained from virtual screening acting as sphingosine kinase 1 inhibitors and potential anticancer agents. Bioorg Chem 2024; 144:107112. [PMID: 38237390 DOI: 10.1016/j.bioorg.2024.107112] [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: 10/27/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 02/17/2024]
Abstract
We report here the virtual screening design, synthesis and activity of eight new inhibitors of SphK1. For this study we used a pre-trained Graph Convolutional Network (GCN) combined with docking calculations. This exploratory analysis proposed nine compounds from which eight displayed significant inhibitory effect against sphingosine kinase 1 (SphK1) demonstrating a high level of efficacy for this approach. Four of these compounds also displayed anticancer activity against different tumor cell lines, and three of them (5), (6) and (7) have shown a wide inhibitory action against many of the cancer cell line tested, with GI50 below 5 µM, being (5) the most promising with TGI below 10 µM for the half of cell lines. Our results suggest that the three most promising compounds reported here are the pyrimidine-quinolone hybrids (1) and (6) linked by p-aminophenylsulfanyl and o-aminophenol fragments respectively, and (8) without such aryl linker. We also performed an exhaustive study about the molecular interactions that stabilize the different ligands at the binding site of SphK1. This molecular modeling analysis was carried out by using combined techniques: docking calculations, MD simulations and QTAIM analysis. In this study we also included PF543, as reference compound, in order to better understand the molecular behavior of these ligands at the binding site of SphK1.These results provide useful information for the design of new inhibitors of SphK1 possessing these structural scaffolds.
Collapse
Affiliation(s)
- Marcela Vettorazzi
- Universidad Nacional de San Luis, Facultad de Química, Bioquímica y Farmacia, Ejercito de los Andes 950, (5700) San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejercito de los Andes 950, (5700) San Luis, Argentina
| | - Iván Díaz
- Universidad de Jaén, Departamento de Química Inorgánica y Orgánica, Campus Las Lagunillas s/n, 23071 Jaén, Spain
| | - Emilio Angelina
- Universidad Nacional del Nordeste, Facultad de Ciencias Exactas y Naturales y Agrimensura, Departamento de Química, Área de Química Física, Laboratorio de Estructura Molecular y Propiedades, Avda. Libertad 5460, 3400 Corrientes, Argentina
| | - Sofía Salido
- Universidad de Jaén, Departamento de Química Inorgánica y Orgánica, Campus Las Lagunillas s/n, 23071 Jaén, Spain
| | - Lucas Gutierrez
- Universidad Nacional de San Luis, Facultad de Química, Bioquímica y Farmacia, Ejercito de los Andes 950, (5700) San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejercito de los Andes 950, (5700) San Luis, Argentina
| | - Sergio E Alvarez
- Universidad Nacional de San Luis, Facultad de Química, Bioquímica y Farmacia, Ejercito de los Andes 950, (5700) San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejercito de los Andes 950, (5700) San Luis, Argentina
| | - Justo Cobo
- Universidad de Jaén, Departamento de Química Inorgánica y Orgánica, Campus Las Lagunillas s/n, 23071 Jaén, Spain.
| | - Ricardo D Enriz
- Universidad Nacional de San Luis, Facultad de Química, Bioquímica y Farmacia, Ejercito de los Andes 950, (5700) San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejercito de los Andes 950, (5700) San Luis, Argentina.
| |
Collapse
|
5
|
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
|
6
|
Mehta V, Dwivedi AR, Ludhiadch A, Rana V, Goel KK, Uniyal P, Joshi G, Kumar A, Kumar B. A decade of USFDA-approved small molecules as anti-inflammatory agents: Recent trends and Commentaries on the "industrial" perspective. Eur J Med Chem 2024; 263:115942. [PMID: 38000212 DOI: 10.1016/j.ejmech.2023.115942] [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: 10/08/2023] [Revised: 11/05/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023]
Abstract
Inflammation is the human body's defence process against various pathogens, toxic substances, irradiation, and physically injured cells that have been damaged. Inflammation is characterized by swelling, pain, redness, heat, as well as diminished tissue function. Multiple important inflammatory markers determine the prognosis of inflammatory processes, which include likes of pro-inflammatory cytokines which are controlled by nuclear factor kappa-B (NF-kB), mitogen-activated protein kinase (MAPK), Janus kinase signal transducer and activator of transcription (JAK-STAT) pathway, all of which are activated in response to the stimulation of specific receptors. Besides these, the cyclooxygenase (COX) enzyme family also plays a significant role in inflammation. The current review is kept forth to compile a summary of small molecules-based drugs approved by the USFDA during the study period of 2013-2023. A thorough discussion has also been made to focus on biologics, macromolecules, and small chemical entities approved during this study period and their greener synthetic routes with a brief discussion on the chemical spacing parameters of anti-inflammatory drugs. The compilation is expected to assist the medicinal chemist and the scientist actively engaged in drug discovery and development of anti-inflammatory agents from newer perspectives during the current years.
Collapse
Affiliation(s)
- Vikrant Mehta
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, Texas, 78229, USA
| | | | - Abhilash Ludhiadch
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, 10032, USA
| | - Vikas Rana
- School of Pharmacy, Graphic Era Hill University, Clement town, Dehradun, 248002, Uttarakhand, India
| | - Kapil Kumar Goel
- Department of Pharmaceutical Sciences, Gurukul Kangri (Deemed to Be University), Haridwar, 249404, Uttarakhand, India
| | - Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Clement town, Dehradun, 248002, Uttarakhand, India
| | - Gaurav Joshi
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Garhwal, Srinagar, Uttarakhand, 246174, India; Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, 248002, Uttarakhand, India.
| | - Asim Kumar
- Amity Institute of Pharmacy (AIP), Amity University Haryana, Panchgaon, Manesar, 122413, India.
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Garhwal, Srinagar, Uttarakhand, 246174, India.
| |
Collapse
|
7
|
Nenkov M, Shi Y, Ma Y, Gaßler N, Chen Y. Targeting Farnesoid X Receptor in Tumor and the Tumor Microenvironment: Implication for Therapy. Int J Mol Sci 2023; 25:6. [PMID: 38203175 PMCID: PMC10778939 DOI: 10.3390/ijms25010006] [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: 11/08/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
The farnesoid-X receptor (FXR), a member of the nuclear hormone receptor superfamily, can be activated by bile acids (BAs). BAs binding to FXR activates BA signaling which is important for maintaining BA homeostasis. FXR is differentially expressed in human organs and exists in immune cells. The dysregulation of FXR is associated with a wide range of diseases including metabolic disorders, inflammatory diseases, immune disorders, and malignant neoplasm. Recent studies have demonstrated that FXR influences tumor cell progression and development through regulating oncogenic and tumor-suppressive pathways, and, moreover, it affects the tumor microenvironment (TME) by modulating TME components. These characteristics provide a new perspective on the FXR-targeted therapeutic strategy in cancer. In this review, we have summarized the recent research data on the functions of FXR in solid tumors and its influence on the TME, and discussed the mechanisms underlying the distinct function of FXR in various types of tumors. Additionally, the impacts on the TME by other BA receptors such as takeda G protein-coupled receptor 5 (TGR5), sphingosine-1-phosphate receptor 2 (S1PR2), and muscarinic receptors (CHRM2 and CHRM3), have been depicted. Finally, the effects of FXR agonists/antagonists in a combination therapy with PD1/PD-L1 immune checkpoint inhibitors and other anti-cancer drugs have been addressed.
Collapse
Affiliation(s)
- Miljana Nenkov
- Section Pathology of the Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (M.N.); (Y.M.); (N.G.)
| | - Yihui Shi
- California Pacific Medical Center Research Institute, Sutter Bay Hospitals, San Francisco, CA 94107, USA;
| | - Yunxia Ma
- Section Pathology of the Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (M.N.); (Y.M.); (N.G.)
| | - Nikolaus Gaßler
- Section Pathology of the Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (M.N.); (Y.M.); (N.G.)
| | - Yuan Chen
- Section Pathology of the Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (M.N.); (Y.M.); (N.G.)
| |
Collapse
|
8
|
Gao G, Liao W, Shu P, Ma Q, He X, Zhang B, Qin D, Wang Y. Targeting sphingosine 1-phosphate receptor 3 inhibits T-cell exhaustion and regulates recruitment of proinflammatory macrophages to improve antitumor efficacy of CAR-T cells against solid tumor. J Immunother Cancer 2023; 11:e006343. [PMID: 37591632 PMCID: PMC10441059 DOI: 10.1136/jitc-2022-006343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUNDS Chimeric antigen receptor (CAR)-modified T cells (CAR-T) are limited in solid tumors due to the hostile tumor microenvironment (TME). Combination therapy could be a promising approach to overcome this obstacle. Recent studies have shown that sphingosine 1-phosphate receptor (S1PR)3 has tremendous potential in regulating the immune environment. However, the functional significance of S1PR3 in T-cell-based immunotherapies and the molecular mechanisms have not been fully understood. METHODS Here, we studied the combination of EpCAM-specific CAR T-cell therapy with pharmacological blockade of S1PR3 against solid tumor. We have applied RNA sequencing, flow cytometry, ELISA, cellular/molecular immunological technology, and mouse models of solid cancers. RESULTS Our study provided evidence that S1PR3 high expression is positively associated with resistance to programmed cell death protein-1 (PD-1)-based immunotherapy and increased T-cell exhaustion. In addition, pharmacological inhibition of S1PR3 improves the efficacy of anti-PD-1 therapy. Next, we explored the possible combination of S1PR3 antagonist with murine EpCAM-targeted CAR-T cells in immunocompetent mouse models of breast cancer and colon cancer. The results indicated that the S1PR3 antagonist could significantly enhance the efficacy of murine EpCAM CAR-T cells in vitro and in vivo. Mechanistically, the S1PR3 antagonist improved CAR-T cell activation, regulated the central memory phenotype, and reduced CAR-T cell exhaustion in vitro. Targeting S1PR3 was shown to remodel the TME through the recruitment of proinflammatory macrophages by promoting macrophage activation and proinflammatory phenotype polarization, resulting in improved CAR-T cell infiltration and amplified recruitment of CD8+T cells. CONCLUSIONS This work demonstrated targeting S1PR3 could increase the antitumor activities of CAR-T cell therapy at least partially by inhibiting T-cell exhaustion and remodeling the TME through the recruitment of proinflammatory macrophages. These findings provided additional rationale for combining S1PR3 inhibitor with CAR-T cells for the treatment of solid tumor.
Collapse
Affiliation(s)
- Ge Gao
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Weiting Liao
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Pei Shu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Qizhi Ma
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Xia He
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, Sichuan University West China Hospital, Chengdu, Sichuan, China
- Department of Clinical Research Management, Sichuan University West China Hospital, Chengdu,Sichuan, China
| | - Benxia Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Diyuan Qin
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Pérez CN, Falcón CR, Mons JD, Orlandi FC, Sangiacomo M, Fernandez-Muñoz JM, Guerrero M, Benito PG, Colombo MI, Zoppino FCM, Alvarez SE. Melanoma cells with acquired resistance to vemurafenib have decreased autophagic flux and display enhanced ability to transfer resistance. Biochim Biophys Acta Mol Basis Dis 2023:166801. [PMID: 37419396 DOI: 10.1016/j.bbadis.2023.166801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/31/2023] [Accepted: 06/28/2023] [Indexed: 07/09/2023]
Abstract
Over the last years, the incidence of melanoma, the deadliest form of skin cancer, has risen significantly. Nearly half of the melanoma patients exhibit the BRAFV600E mutation. Although the use of BRAF and MEK inhibitors (BRAFi and MEKi) showed an impressive success rate in melanoma patients, durability of response remains an issue because tumor quickly becomes resistant. Here, we generated and characterized Lu1205 and A375 melanoma cells resistant to vemurafenib (BRAFi). Resistant cells (Lu1205R and A375R) exhibit higher IC50 (5-6 fold increase) and phospho-ERK levels and 2-3 times reduced apoptosis than their sensitive parents (Lu1205S and A375S). Moreover, resistant cells are 2-3 times bigger, display a more elongated morphology and have a modulation the migration capacity. Interestingly, pharmacological inhibition of sphingosine kinases, that prevents sphingosine-1-phosphate production, reduces migration of Lu1205R cells by 50 %. In addition, although Lu1205R cells showed increased basal levels of the autophagy markers LC3II and p62, they have decreased autophagosome degradation and autophagy flux. Remarkably, expression of Rab27A and Rab27B, which are involved in the release of extracellular vesicles are dramatically augmented in resistant cells (i.e. 5-7 fold increase). Indeed, conditioned media obtained from Lu1205R cells increased the resistance to vemurafenib of sensitive cells. Hence, these results support that resistance to vemurafenib modulates migration and the autophagic flux and may be transferred to nearby sensitive melanoma cells by factors that are released to the extracellular milieu by resistant cells.
Collapse
Affiliation(s)
- Celia N Pérez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina
| | - Cristian R Falcón
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina
| | - Johinna Delgado Mons
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina
| | - Federico Cuello Orlandi
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina
| | - Mercedes Sangiacomo
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina
| | | | - Martín Guerrero
- Instituto de Biología y Medicina Experimental de Cuyo (IMBECU), CONICET, Argentina
| | - Paula G Benito
- Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo-CONICET, Argentina
| | - María I Colombo
- Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo-CONICET, Argentina
| | - Felipe C M Zoppino
- Instituto de Biología y Medicina Experimental de Cuyo (IMBECU), CONICET, Argentina
| | - Sergio E Alvarez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina.
| |
Collapse
|
10
|
Jang H, Ojha U, Jeong JH, Park KG, Lee SY, Lee YM. Myriocin suppresses tumor growth by modulating macrophage polarization and function through the PI3K/Akt/mTOR pathway. Arch Pharm Res 2023; 46:629-645. [PMID: 37468765 DOI: 10.1007/s12272-023-01454-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/02/2023] [Indexed: 07/21/2023]
Abstract
Macrophages within the tumor microenvironment (TME), referred to as tumor-associated macrophages (TAMs), are involved in various aspects of tumor progression including initiation, angiogenesis, metastasis, immunosuppression, and resistance to therapy. Myriocin, a natural compound isolated from Mycelia sterilia, is an immunosuppressant that inhibits tumor growth and angiogenesis. However, the mechanisms underlying the effects of myriocin on TAMs and TAM-mediated tumor growth have not yet been elucidated. In this study, we determined the effects of myriocin on TAMs and the underlying mechanism in vitro and in vivo. Myriocin significantly suppressed monocyte-macrophage differentiation and M2 polarization of macrophages but not M1 polarization. In addition, myriocin inhibited the expression of anti-inflammatory cytokines and secretion of proangiogenic factors, such as vascular endothelial growth factor, in M2 macrophages as well as M2-induced endothelial cell permeability. Myriocin also inhibited the PI3K/Akt/mTOR signaling pathway in M2 macrophages. Myriocin reduced the population of M2-like TAMs within the tumor tissue of a mouse allograft tumor model but not that of M1-like TAMs. Moreover, combined treatment with myriocin and cisplatin synergistically suppressed tumor growth and enhanced survival rate in mice by reducing the population of M2-like TAMs. Overall, these results suggest that myriocin inhibits tumor growth by remodeling the TME through suppression of differentiation and polarization of M2-like TAMs via the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hyeonha Jang
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Uttam Ojha
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ji-Hak Jeong
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Keun-Gyu Park
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Shin Yup Lee
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - You Mie Lee
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
11
|
Abdelraheem KM, Younis NN, Shaheen MA, Elswefy SE, Ali SI. Raspberry ketone improves non-alcoholic fatty liver disease induced in rats by modulating sphingosine kinase/sphingosine-1-phosphate and toll-like receptor 4 pathways. J Pharm Pharmacol 2023:7160323. [PMID: 37167472 DOI: 10.1093/jpp/rgad044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 04/19/2023] [Indexed: 05/13/2023]
Abstract
OBJECTIVES To investigate the therapeutic role of calorie-restricted diet (CR) and raspberry ketone (RK) in non-alcoholic fatty liver disease (NAFLD) and the implication of sphingosine kinase-1 (SphK1)/sphingosine-1-phosphate (S1P) and toll-like receptor 4 (TLR4) signalling. METHODS NAFLD was induced by feeding rats high-fat-fructose-diet (HFFD) for 6 weeks. Rats were then randomly assigned to three groups (n = 6 each); NAFLD group continued on HFFD for another 8 weeks. CR group was switched to CR diet (25% calorie restriction) for 8 weeks and RK group was switched to normal diet and received RK (55 mg/kg/day; orally) for 8 weeks. Another six rats were used as normal control. KEY FINDINGS HFFD induced a state of NAFLD indicated by increased fat deposition in liver tissue along with dyslipidemia, elevated liver enzymes, oxidative stress and inflammation. Either CR diet or RK reversed these changes and decreased HFFD-induced elevation of hepatic SphK1, S1P, S1PR1 and TLR4. Of notice, RK along with a normal calorie diet was even better than CR alone in most studied parameters. CONCLUSIONS SphK1/S1P and TLR4 are interconnected and related to the establishment of HFFD-induced NAFLD and can be modulated by RK. Supplementation of RK without calorie restriction to patients with NAFLD unable to follow CR diet to achieve their treatment goals would be a promising therapeutic modality.
Collapse
Affiliation(s)
- Kareem M Abdelraheem
- Biochemistry Department, Faculty of Pharmacy, Sinai University - Qantara Branch, Ismailia, Egypt
| | - Nahla N Younis
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed A Shaheen
- Histology and Cell Biology Department, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| | - Sahar E Elswefy
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
- Biochemistry Department, Faculty of Pharmacy, Delta University for Sciences and Technology, Gamasa, Egypt
| | - Sousou I Ali
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
12
|
Wang X, Guo W, Shi X, Chen Y, Yu Y, Du B, Tan M, Tong L, Wang A, Yin X, Guo J, Martin RC, Bai O, Li Y. S1PR1/S1PR3-YAP signaling and S1P-ALOX15 signaling contribute to an aggressive behavior in obesity-lymphoma. J Exp Clin Cancer Res 2023; 42:3. [PMID: 36600310 PMCID: PMC9814427 DOI: 10.1186/s13046-022-02589-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/28/2022] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Excess body weight has been found to associate with an increased risk of lymphomas and some metabolic pathways are currently recognized in lymphomagenesis. Bioactive lipid metabolites such as sphingosine-1-phosphate (S1P) have been proposed to play an important role linking obesity and lymphomas. However, the underlying mechanism(s) of S1P signaling in obesity-lymphomagenesis have not been well addressed. METHODS The gene expression of sphingosine kinase (SPHK), lymphoma prognosis, and S1P production were analyzed using Gene Expression Omnibus (GEO) and human lymphoma tissue array. Obesity-lymphoma mouse models and lymphoma cell lines were used to investigate the S1P/SPHK-YAP axis contributing to obesity-lymphomagenesis. By using the mouse models and a monocyte cell line, S1P-mediated polarization of macrophages in the tumor microenvironment were investigated. RESULTS In human study, up-regulated S1P/SPHK1 was found in human lymphomas, while obesity negatively impacted progression-free survival and overall survival in lymphoma patients. In animal study, obesity-lymphoma mice showed an aggressive tumor growth pattern. Both in vivo and in vitro data suggested the existence of S1P-YAP axis in lymphoma cells, while the S1P-ALOX15 signaling mediated macrophage polarization towards TAMs exacerbated the lymphomagenesis. In addition, treatment with resveratrol in obesity-lymphoma mice showed profound effects of anti-lymphomagenesis, via down-regulating S1P-YAP axis and modulating polarization of macrophages. CONCLUSION S1P/S1PR initiated the feedback loops, whereby S1P-S1PR1/S1PR3-YAP signaling mediated lymphomagenesis contributing to tumor aggressive growth, while S1P-ALOX15 signaling mediated TAMs contributing to immunosuppressive microenvironment in obesity-lymphoma. S1P-targeted therapy could be potentially effective and immune-enhancive against obesity-lymphomagenesis.
Collapse
Affiliation(s)
- Xingtong Wang
- Department of Surgery, School of Medicine, University of Louisville, 511 S Floyd ST MDR Bldg Rm326A, Louisville, KY, 40202, USA
- Department of Hematology, Cancer Center, The First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, Jilin, China
| | - Wei Guo
- Department of Surgery, School of Medicine, University of Louisville, 511 S Floyd ST MDR Bldg Rm326A, Louisville, KY, 40202, USA
- Department of Hematology, Cancer Center, The First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, Jilin, China
| | - Xiaoju Shi
- Department of Surgery, School of Medicine, University of Louisville, 511 S Floyd ST MDR Bldg Rm326A, Louisville, KY, 40202, USA
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yujia Chen
- Department of Surgery, School of Medicine, University of Louisville, 511 S Floyd ST MDR Bldg Rm326A, Louisville, KY, 40202, USA
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Youxi Yu
- Department of Surgery, School of Medicine, University of Louisville, 511 S Floyd ST MDR Bldg Rm326A, Louisville, KY, 40202, USA
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Beibei Du
- Department of Cardiology, China-Japan Union hospital of Jilin University, Changchun, 130033, China
| | - Min Tan
- Department of Surgery, School of Medicine, University of Louisville, 511 S Floyd ST MDR Bldg Rm326A, Louisville, KY, 40202, USA
| | - Li Tong
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Anna Wang
- Department of Hematology, Cancer Center, The First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, Jilin, China
| | - Xianying Yin
- Department of Hematology, Cancer Center, The First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, Jilin, China
| | - Jing Guo
- Department of Hematology, Cancer Center, The First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, Jilin, China
| | - Robert C Martin
- Department of Surgery, School of Medicine, University of Louisville, 511 S Floyd ST MDR Bldg Rm326A, Louisville, KY, 40202, USA
| | - Ou Bai
- Department of Hematology, Cancer Center, The First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, Jilin, China.
| | - Yan Li
- Department of Surgery, School of Medicine, University of Louisville, 511 S Floyd ST MDR Bldg Rm326A, Louisville, KY, 40202, USA.
| |
Collapse
|
13
|
Sarapura Martinez VJ, Buonincontro B, Cassarino C, Bernatowiez J, Colado A, Cordini G, Custidiano MDR, Mahuad C, Pavlovsky MA, Bezares RF, Favale NO, Vermeulen M, Borge M, Giordano M, Gamberale R. Venetoclax resistance induced by activated T cells can be counteracted by sphingosine kinase inhibitors in chronic lymphocytic leukemia. Front Oncol 2023; 13:1143881. [PMID: 37020867 PMCID: PMC10067719 DOI: 10.3389/fonc.2023.1143881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
The treatment of chronic lymphocytic leukemia (CLL) patients with venetoclax-based regimens has demonstrated efficacy and a safety profile, but the emergence of resistant cells and disease progression is a current complication. Therapeutic target of sphingosine kinases (SPHK) 1 and 2 has opened new opportunities in the treatment combinations of cancer patients. We previously reported that the dual SPHK1/2 inhibitor, SKI-II enhanced the in vitro cell death triggered by fludarabine, bendamustine or ibrutinib and reduced the activation and proliferation of chronic lymphocytic leukemia (CLL) cells. Since we previously showed that autologous activated T cells from CLL patients favor the activation of CLL cells and the generation of venetoclax resistance due to the upregulation of BCL-XL and MCL-1, we here aim to determine whether SPHK inhibitors affect this process. To this aim we employed the dual SPHK1/2 inhibitor SKI-II and opaganib, a SPHK2 inhibitor that is being studied in clinical trials. We found that SPHK inhibitors reduce the activation of CLL cells and the generation of venetoclax resistance induced by activated T cells mainly due to a reduced upregulation of BCL-XL. We also found that SPHK2 expression was enhanced in CLL cells by activated T cells of the same patient and the presence of venetoclax selects resistant cells with high levels of SPHK2. Of note, SPHK inhibitors were able to re-sensitize already resistant CLL cells to a second venetoclax treatment. Our results highlight the therapeutic potential of SPHK inhibitors in combination with venetoclax as a promising treatment option for the patients.
Collapse
Affiliation(s)
- Valeria J. Sarapura Martinez
- Laboratorio de Inmunología Oncológica, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)- Academia Nacional de Medicina (ANM), Buenos Aires, Argentina
| | - Brenda Buonincontro
- Laboratorio de Inmunología Oncológica, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)- Academia Nacional de Medicina (ANM), Buenos Aires, Argentina
| | - Chiara Cassarino
- Laboratorio de Inmunología Oncológica, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)- Academia Nacional de Medicina (ANM), Buenos Aires, Argentina
| | - Juliana Bernatowiez
- Laboratorio de Inmunología Oncológica, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)- Academia Nacional de Medicina (ANM), Buenos Aires, Argentina
| | - Ana Colado
- Laboratorio de Inmunología Oncológica, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)- Academia Nacional de Medicina (ANM), Buenos Aires, Argentina
| | - Gregorio Cordini
- Laboratorio de Inmunología Oncológica, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)- Academia Nacional de Medicina (ANM), Buenos Aires, Argentina
- Servicio de Hematología, Hospital de Clínicas, José de San Martín, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Maria del Rosario Custidiano
- Departamento de Hematología y Unidad de Trasplante Hematopoyético, Instituto Alexander Fleming, Buenos Aires, Argentina
| | - Carolina Mahuad
- Servicio de Hematología, Hospital Alemán, Buenos Aires, Argentina
| | | | | | - Nicolás O. Favale
- Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Química y Fisicoquímica Biológicas “Profesor Dr. Alejandro C. Paladini” (IQUIFIB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mónica Vermeulen
- Laboratorio de Células Presentadoras de Antígeno y Respuesta Inflamatoria, IMEX-CONICET-ANM, Buenos Aires, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA, Buenos Aires, Argentina
| | - Mercedes Borge
- Laboratorio de Inmunología Oncológica, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)- Academia Nacional de Medicina (ANM), Buenos Aires, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA, Buenos Aires, Argentina
| | - Mirta Giordano
- Laboratorio de Inmunología Oncológica, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)- Academia Nacional de Medicina (ANM), Buenos Aires, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA, Buenos Aires, Argentina
| | - Romina Gamberale
- Laboratorio de Inmunología Oncológica, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)- Academia Nacional de Medicina (ANM), Buenos Aires, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA, Buenos Aires, Argentina
- *Correspondence: Romina Gamberale,
| |
Collapse
|
14
|
S1P-Induced TNF-α and IL-6 Release from PBMCs Exacerbates Lung Cancer-Associated Inflammation. Cells 2022; 11:cells11162524. [PMID: 36010601 PMCID: PMC9406848 DOI: 10.3390/cells11162524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/22/2022] [Accepted: 08/13/2022] [Indexed: 12/03/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is involved in inflammatory signaling/s associated with the development of respiratory disorders, including cancer. However, the underlying mechanism/s are still elusive. The aim of this study was to investigate the role of S1P on circulating blood cells obtained from healthy volunteers and non-small cell lung cancer (NSCLC) patients. To pursue our goal, peripheral blood mononuclear cells (PBMCs) were isolated and stimulated with S1P. We found that the administration of S1P did not induce healthy PBMCs to release pro-inflammatory cytokines. In sharp contrast, S1P significantly increased the levels of TNF-α and IL-6 from lung cancer-derived PBMCs. This effect was S1P receptor 3 (S1PR3)-dependent. The pharmacological blockade of ceramidase and sphingosine kinases (SPHKs), key enzymes for S1P synthesis, completely reduced the release of both TNF-α and IL-6 after S1P addition on lung cancer-derived PBMCs. Interestingly, S1P-induced IL-6, but not TNF-α, release from lung cancer-derived PBMCs was mTOR- and K-Ras-dependent, while NF-κB was not involved. These data identify S1P as a bioactive lipid mediator in a chronic inflammation-driven diseases such as NSCLC. In particular, the higher presence of S1P could orchestrate the cytokine milieu in NSCLC, highlighting S1P as a pro-tumor driver.
Collapse
|
15
|
Li RZ, Wang XR, Wang J, Xie C, Wang XX, Pan HD, Meng WY, Liang TL, Li JX, Yan PY, Wu QB, Liu L, Yao XJ, Leung ELH. The key role of sphingolipid metabolism in cancer: New therapeutic targets, diagnostic and prognostic values, and anti-tumor immunotherapy resistance. Front Oncol 2022; 12:941643. [PMID: 35965565 PMCID: PMC9364366 DOI: 10.3389/fonc.2022.941643] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/24/2022] [Indexed: 12/13/2022] Open
Abstract
Biologically active sphingolipids are closely related to the growth, differentiation, aging, and apoptosis of cancer cells. Some sphingolipids, such as ceramides, are favorable metabolites in the sphingolipid metabolic pathway, usually mediating antiproliferative responses, through inhibiting cancer cell growth and migration, as well as inducing autophagy and apoptosis. However, other sphingolipids, such as S1P, play the opposite role, which induces cancer cell transformation, migration and growth and promotes drug resistance. There are also other sphingolipids, as well as enzymes, played potentially critical roles in cancer physiology and therapeutics. This review aimed to explore the important roles of sphingolipid metabolism in cancer. In this article, we summarized the role and value of sphingolipid metabolism in cancer, including the distribution of sphingolipids, the functions, and their relevance to cancer diagnosis and prognosis. We also summarized the known and potential antitumor targets present in sphingolipid metabolism, analyzed the correlation between sphingolipid metabolism and tumor immunity, and summarize the antitumor effects of natural compounds based on sphingolipids. Through the analysis and summary of sphingolipid antitumor therapeutic targets and immune correlation, we aim to provide ideas for the development of new antitumor drugs, exploration of new therapeutic means for tumors, and study of immunotherapy resistance mechanisms.
Collapse
Affiliation(s)
- Run-Ze Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Macao, Macao SAR, China
| | - Xuan-Run Wang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jian Wang
- Department of Oncology, Luzhou People’s Hospital, Luzhou, Sichuan, China
| | - Chun Xie
- Cancer Center, Faculty of Health Science, University of Macau, Macao, Macao SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao, Macao SAR, China
| | - Xing-Xia Wang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Hu-Dan Pan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Macao, Macao SAR, China
| | - Wei-Yu Meng
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Tu-Liang Liang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jia-Xin Li
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Pei-Yu Yan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Qi-Biao Wu
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Liang Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Macao, Macao SAR, China
- *Correspondence: Xiao-Jun Yao, ; Liang Liu, ; Elaine Lai-Han Leung,
| | - Xiao-Jun Yao
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
- *Correspondence: Xiao-Jun Yao, ; Liang Liu, ; Elaine Lai-Han Leung,
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Science, University of Macau, Macao, Macao SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao, Macao SAR, China
- Breast Surgery, Zhuhai Hospital of Traditional Chinese and Western Medicine, Zhuhai, China
- *Correspondence: Xiao-Jun Yao, ; Liang Liu, ; Elaine Lai-Han Leung,
| |
Collapse
|
16
|
Sphingosine 1-phosphate modulation and immune cell trafficking in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2022; 19:351-366. [PMID: 35165437 DOI: 10.1038/s41575-021-00574-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 12/12/2022]
Abstract
Immune cell trafficking is a critical element of the intestinal immune response, both in homeostasis and in pathological conditions associated with inflammatory bowel disease (IBD). This process involves adhesion molecules, chemoattractants and receptors expressed on immune cell surfaces, blood vessels and stromal intestinal tissue as well as signalling pathways, including those modulated by sphingosine 1-phosphate (S1P). The complex biological processes of leukocyte recruitment, activation, adhesion and migration have been targeted by various monoclonal antibodies (vedolizumab, etrolizumab, ontamalimab). Promising preclinical and clinical data with several oral S1P modulators suggest that inhibition of lymphocyte egress from the lymph nodes to the bloodstream might be a safe and efficacious alternative mechanism for reducing inflammation in immune-mediated disorders, including Crohn's disease and ulcerative colitis. Although various questions remain, including the potential positioning of S1P modulators in treatment algorithms and their long-term safety, this novel class of compounds holds great promise. This Review summarizes the critical mediators and mechanisms involved in immune cell trafficking in IBD and the available evidence for efficacy, safety and pharmacokinetics of S1P receptor modulators in IBD and other immune-mediated disorders. Further, it discusses potential future approaches to incorporate S1P modulators into the treatment of IBD.
Collapse
|
17
|
Gupta P, Kadamberi IP, Mittal S, Tsaih S, George J, Kumar S, Vijayan DK, Geethadevi A, Parashar D, Topchyan P, McAlarnen L, Volkman BF, Cui W, Zhang KYJ, Di Vizio D, Chaluvally‐Raghavan P, Pradeep S. Tumor Derived Extracellular Vesicles Drive T Cell Exhaustion in Tumor Microenvironment through Sphingosine Mediated Signaling and Impacting Immunotherapy Outcomes in Ovarian Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104452. [PMID: 35289120 PMCID: PMC9108620 DOI: 10.1002/advs.202104452] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/10/2022] [Indexed: 05/13/2023]
Abstract
SPHK1 (sphingosine kinase-1) catalyzes the phosphorylation of sphingosine to sphingosine-1-phosphate (S1P), is found to be highly expressed in solid tumors. Here, extracellular vesicles (EVs) are identified as the key transporters of SPHK1 to the tumor microenvironment. Consequently, SPHK1-packaged EVs elevate S1P levels in the tumor microenvironment, where S1P appears as an immunosuppressive agent. However, the exact mechanism of how S1P mediates its immunosuppressive effects in cancer is not understood. It is investigated that S1P can induce T cell exhaustion. S1P can also upregulate programmed death ligand-1 (PDL-1) expression through E2F1-mediated transcription. Notably, an SPHK1 inhibitor PF543 improves T cell-mediated cytotoxicity. Furthermore, combining PF543 with an anti-PD-1 antibody reduces tumor burden and metastasis more effectively than PF543 alone in vivo. These data demonstrate a previously unrecognized mechanism of how SPHK1-packaged EVs contribute to the progression of ovarian cancer and thus present the potential clinical application of inhibiting SPHK1/S1P signaling to improve immune checkpoint blockage (anti-PD-1 antibody) therapy in ovarian cancer.
Collapse
Affiliation(s)
- Prachi Gupta
- Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsin53226USA
| | | | - Sonam Mittal
- Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsin53226USA
| | - Shirng‐Wern Tsaih
- Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsin53226USA
| | - Jasmine George
- Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsin53226USA
| | - Sudhir Kumar
- Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsin53226USA
| | - Dileep K. Vijayan
- Laboratory for computational and structural biologyJubilee Center for Medical ResearchThrissurKerala680006India
- Laboratory for Structural BioinformaticsCenter for Biosystems Dynamics ResearchRiken230‐0045Japan
| | - Anjali Geethadevi
- Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsin53226USA
| | - Deepak Parashar
- Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsin53226USA
| | - Paytsar Topchyan
- Department of Microbiology and ImmunologyMCW and Versiti Blood Research InstituteMilwaukeeWisconsin53226USA
| | - Lindsey McAlarnen
- Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsin53226USA
| | - Brian F Volkman
- Department of BiochemistryMedical College of WisconsinMilwaukee53226USA
| | - Weiguo Cui
- Department of Microbiology and ImmunologyMCW and Versiti Blood Research InstituteMilwaukeeWisconsin53226USA
| | - Kam Y. J. Zhang
- Laboratory for Structural BioinformaticsCenter for Biosystems Dynamics ResearchRiken230‐0045Japan
| | - Dolores Di Vizio
- Department of SurgeryPathology and Laboratory MedicineSamuel Oschin Comprehensive Cancer InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Pradeep Chaluvally‐Raghavan
- Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsin53226USA
- Department of PhysiologyMedical College of WisconsinMilwaukeeWisconsin53226USA
- Medical College of Wisconsin Cancer CenterMedical College of WisconsinMilwaukeeWisconsin53226USA
| | - Sunila Pradeep
- Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsin53226USA
- Department of PhysiologyMedical College of WisconsinMilwaukeeWisconsin53226USA
- Medical College of Wisconsin Cancer CenterMedical College of WisconsinMilwaukeeWisconsin53226USA
| |
Collapse
|
18
|
Pherez-Farah A, López-Sánchez RDC, Villela-Martínez LM, Ortiz-López R, Beltrán BE, Hernández-Hernández JA. Sphingolipids and Lymphomas: A Double-Edged Sword. Cancers (Basel) 2022; 14:2051. [PMID: 35565181 PMCID: PMC9104519 DOI: 10.3390/cancers14092051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/14/2022] [Indexed: 11/24/2022] Open
Abstract
Lymphomas are a highly heterogeneous group of hematological neoplasms. Given their ethiopathogenic complexity, their classification and management can become difficult tasks; therefore, new approaches are continuously being sought. Metabolic reprogramming at the lipid level is a hot topic in cancer research, and sphingolipidomics has gained particular focus in this area due to the bioactive nature of molecules such as sphingoid bases, sphingosine-1-phosphate, ceramides, sphingomyelin, cerebrosides, globosides, and gangliosides. Sphingolipid metabolism has become especially exciting because they are involved in virtually every cellular process through an extremely intricate metabolic web; in fact, no two sphingolipids share the same fate. Unsurprisingly, a disruption at this level is a recurrent mechanism in lymphomagenesis, dissemination, and chemoresistance, which means potential biomarkers and therapeutical targets might be hiding within these pathways. Many comprehensive reviews describing their role in cancer exist, but because most research has been conducted in solid malignancies, evidence in lymphomagenesis is somewhat limited. In this review, we summarize key aspects of sphingolipid biochemistry and discuss their known impact in cancer biology, with a particular focus on lymphomas and possible therapeutical strategies against them.
Collapse
Affiliation(s)
- Alfredo Pherez-Farah
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Nuevo Leon, Mexico
| | | | - Luis Mario Villela-Martínez
- Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán Rosales 80030, Sinaloa, Mexico
- Hospital Fernando Ocaranza, ISSSTE, Hermosillo 83190, Sonora, Mexico
- Centro Médico Dr. Ignacio Chávez, ISSSTESON, Hermosillo 83000, Sonora, Mexico
| | - Rocío Ortiz-López
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Nuevo Leon, Mexico
| | - Brady E Beltrán
- Hospital Edgardo Rebagliati Martins, Lima 15072, Peru
- Instituto de Investigaciones en Ciencias Biomédicas, Universidad Ricardo Palma, Lima 1801, Peru
| | | |
Collapse
|
19
|
Yan Y, Bao G, Pei J, Cao Y, Zhang C, Zhao P, Zhang Y, Damirin A. NF-κB and EGFR participate in S1PR3-mediated human renal cell carcinomas progression. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166401. [PMID: 35346818 DOI: 10.1016/j.bbadis.2022.166401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 11/24/2022]
Abstract
The bioactive lipid sphingosine 1-phosphate (S1P) is implicated in many pivotal processes for the physiological and pathological actions via activating five types of G-protein-coupled S1P receptors (S1PR1-5). The role of S1P in renal cell carcinoma (RCC) and its receptor subtype specific mediating mechanism are poorly studied. So we focus on the regulatory role of S1P in RCC progression and the receptor subtypes involved in S1P-induced actions, intending to further clarify a novel therapeutic target for RCC. Analysis of The Cancer Genome Atlas (TCGA) databases showed that the patients with high expression of S1PR3 had significantly worse overall than with low expression. We further demonstrated that S1P could promote proliferation, migration, and epithelial-mesenchymal transition (EMT) of renal cancer cells in vitro, and the actions were enhanced with the increase of S1PR3 expression. Meanwhile, the results in animal experiments also showed that S1PR3 could accelerate tumorigenesis and metastasis of RCC. Our study also clarified the mechanism for S1P induced cell proliferation is mediated by S1PR3/Gi/p38/Akt/p65/cyclin D1-CDK4 pathway and the main pathway for migration is S1PR3/Gi/q/ERK/p38/p65. In addition, S1PR3 was involved in epidermal growth factor (EGF)-induced actions by enhancing protein expression, not by transactivation of epidermal growth factor receptor (EGFR). These results also further supported our conclusion that the carcinogenic role of S1P/S1PR3 axis. Thus, our findings provide that S1PR3 may be a promising small molecular therapeutic target for S1PR3 expressed cancers.
Collapse
Affiliation(s)
- Yali Yan
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Gegentuya Bao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Jingyuan Pei
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Ying Cao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Chenyu Zhang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Pengfei Zhao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Yantao Zhang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Alatangaole Damirin
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
20
|
Ma Y, Chang N, Liu Y, Liu F, Dong C, Hou L, Qi C, Yang L, Li L. Silencing IQGAP1 alleviates hepatic fibrogenesis via blocking bone marrow mesenchymal stromal cell recruitment to fibrotic liver. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:471-483. [PMID: 35036058 PMCID: PMC8728523 DOI: 10.1016/j.omtn.2021.12.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 12/15/2021] [Indexed: 02/06/2023]
Abstract
IQ motif-containing guanosine triphosphatase (GTPase)-activating protein 1 (IQGAP1) is a cytosolic scaffolding protein involved in cell migration. Our previous studies suggest sphingosine 1-phosphate (S1P) triggers bone marrow (BM) mesenchymal stromal cells (BMSCs) to damaged liver, thereby promoting liver fibrosis. However, the role of IQGAP1 in S1P-induced BMSC migration and liver fibrogenesis remains unclear. Chimeric mice of BM cell labeled by EGFP were used to build methionine-choline-deficient and high-fat (MCDHF)-diet-induced mouse liver fibrosis. IQGAP1 small interfering RNA (siRNA) was utilized to silence IQGAP1 in vivo. IQGAP1 expression is significantly elevated in MCDHF-diet-induced mouse fibrotic livers. Positive correlations are presented between IQGAP1 and fibrosis hallmarks expressions in human and mouse fibrotic livers. In vitro, depressing IQGAP1 expression blocks S1P-induced motility and cytoskeleton remodeling of BMSCs. S1P facilitates IQGAP1 aggregating to plasma membrane via S1P receptor 3 (S1PR3) and Cdc42/Rac1. In addition, IQGAP1 binds to Cdc42/Rac1, regulating S1P-induced activation of Cdc42/Rac1 and mediating BMSC migration in concert. In vivo, silencing IQGAP1 reduces the recruitment of BMSCs to impaired liver and effectively alleviates liver fibrosis induced by MCDHF diet. Together, silencing IQGAP1 relieves liver fibrosis by blocking BMSC migration, providing an effective therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Yuehan Ma
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Na Chang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Yuran Liu
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Fuquan Liu
- Department of Interventional Therapy, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Chengbin Dong
- Department of Interventional Therapy, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Lei Hou
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Changbo Qi
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Lin Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Liying Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| |
Collapse
|
21
|
Olesch C, Brüne B, Weigert A. Keep a Little Fire Burning-The Delicate Balance of Targeting Sphingosine-1-Phosphate in Cancer Immunity. Int J Mol Sci 2022; 23:ijms23031289. [PMID: 35163211 PMCID: PMC8836181 DOI: 10.3390/ijms23031289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 11/16/2022] Open
Abstract
The sphingolipid sphingosine-1-phosphate (S1P) promotes tumor development through a variety of mechanisms including promoting proliferation, survival, and migration of cancer cells. Moreover, S1P emerged as an important regulator of tumor microenvironmental cell function by modulating, among other mechanisms, tumor angiogenesis. Therefore, S1P was proposed as a target for anti-tumor therapy. The clinical success of current cancer immunotherapy suggests that future anti-tumor therapy needs to consider its impact on the tumor-associated immune system. Hereby, S1P may have divergent effects. On the one hand, S1P gradients control leukocyte trafficking throughout the body, which is clinically exploited to suppress auto-immune reactions. On the other hand, S1P promotes pro-tumor activation of a diverse range of immune cells. In this review, we summarize the current literature describing the role of S1P in tumor-associated immunity, and we discuss strategies for how to target S1P for anti-tumor therapy without causing immune paralysis.
Collapse
Affiliation(s)
- Catherine Olesch
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (C.O.); (B.B.)
- Bayer Joint Immunotherapeutics Laboratory, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (C.O.); (B.B.)
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60596 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (C.O.); (B.B.)
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60596 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
- Correspondence:
| |
Collapse
|
22
|
Ye M, Gai Y, Ji H, Jiang Y, Qiao P, Wang W, Zhang Y, Xia X, Lan X. A Novel Radioimmune 99mTc-Labeled Tracer for Imaging Sphingosine 1-Phosphate Receptor 1 in Tumor Xenografts: An In Vitro and In Vivo Study. Front Immunol 2021; 12:660842. [PMID: 34484174 PMCID: PMC8416251 DOI: 10.3389/fimmu.2021.660842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a phospholipid that regulates pleiotropic biological activities and exerts extracellular functions by binding to five specific G-protein-coupled receptors, S1P receptors (S1PR) 1-5. When activated by S1P, S1PR promote the proliferation and invasion of tumor cells by inducing the formation of new blood vessels. We developed and assessed a new monoclonal antibody imaging probe 99mTc-HYNIC-S1PR1mAb, to explore the feasibility of targeting the S1PR1 in vitro and in vivo. S1PR1mAb was prepared and followed by technetium-99m labeling with succinimidyl 6-hydraziniumnicotinate hydrochloride. Cell uptake and blocking studies were performed to investigate the binding specificity of 99mTc-HYNIC-S1PR1mAb in vitro. 99mTc-HYNIC-S1P1mAb was also tested in vivo in mice xenografted with SK-HEP-1 (high-expression of S1PR1) and MCF-7 (low-expression of S1PR1) using single-photon emission-computed tomography (SPECT). Ex vivo gamma counting of tissues from tumor-bearing mice was used to evaluate 99mTc-HYNIC-S1PR1mAb biodistribution. The biodistribution study results showed significantly higher uptake in SK-HEP-1 tumors than in MCF-7 tumors (P < 0.001). Reduced uptake of 99mTc-HYNIC-S1PR1mAb in SK-HEP-1 was observed in tumor-bearing nude mice pretreated with fingolimod, which binds competitively to the receptors, especially S1PR1. 99mTc-HYNIC-S1PR1mAb can be synthesized and specifically targeted to S1PR1 in vitro and in vivo, allowing S1PR1 expression assessment with SPECT imaging.
Collapse
Affiliation(s)
- Min Ye
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Hao Ji
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yaqun Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Pengxin Qiao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Wenxia Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
23
|
Mangiola S, McCoy P, Modrak M, Souza-Fonseca-Guimaraes F, Blashki D, Stuchbery R, Keam SP, Kerger M, Chow K, Nasa C, Le Page M, Lister N, Monard S, Peters J, Dundee P, Williams SG, Costello AJ, Neeson PJ, Pal B, Huntington ND, Corcoran NM, Papenfuss AT, Hovens CM. Transcriptome sequencing and multi-plex imaging of prostate cancer microenvironment reveals a dominant role for monocytic cells in progression. BMC Cancer 2021; 21:846. [PMID: 34294073 PMCID: PMC8296706 DOI: 10.1186/s12885-021-08529-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/23/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Prostate cancer is caused by genomic aberrations in normal epithelial cells, however clinical translation of findings from analyses of cancer cells alone has been very limited. A deeper understanding of the tumour microenvironment is needed to identify the key drivers of disease progression and reveal novel therapeutic opportunities. RESULTS In this study, the experimental enrichment of selected cell-types, the development of a Bayesian inference model for continuous differential transcript abundance, and multiplex immunohistochemistry permitted us to define the transcriptional landscape of the prostate cancer microenvironment along the disease progression axis. An important role of monocytes and macrophages in prostate cancer progression and disease recurrence was uncovered, supported by both transcriptional landscape findings and by differential tissue composition analyses. These findings were corroborated and validated by spatial analyses at the single-cell level using multiplex immunohistochemistry. CONCLUSIONS This study advances our knowledge concerning the role of monocyte-derived recruitment in primary prostate cancer, and supports their key role in disease progression, patient survival and prostate microenvironment immune modulation.
Collapse
Affiliation(s)
- Stefano Mangiola
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Patrick McCoy
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Martin Modrak
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Fernando Souza-Fonseca-Guimaraes
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Daniel Blashki
- The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Ryan Stuchbery
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Simon P Keam
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Michael Kerger
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Ken Chow
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Chayanica Nasa
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Melanie Le Page
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Natalie Lister
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Simon Monard
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Justin Peters
- Epworth Center of Cancer Research, Clayton, Victoria, Australia
| | - Phil Dundee
- Epworth Center of Cancer Research, Clayton, Victoria, Australia
| | - Scott G Williams
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Anthony J Costello
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Paul J Neeson
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Bhupinder Pal
- The Olivia Newton-John Cancer Research Institute, Heidelberg, Melbourne, Australia
| | - Nicholas D Huntington
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Niall M Corcoran
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Urology, Frankston Hospital, Frankston, Victoria, Australia
| | - Anthony T Papenfuss
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- School of Mathematics and Statistics, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Christopher M Hovens
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
24
|
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: 1.0] [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
|
25
|
Giussani P, Prinetti A, Tringali C. The Role of Sphingolipids in Cancer Immunotherapy. Int J Mol Sci 2021; 22:ijms22126492. [PMID: 34204326 PMCID: PMC8234743 DOI: 10.3390/ijms22126492] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 01/04/2023] Open
Abstract
Immunotherapy is now considered an innovative and strong strategy to beat metastatic, drug-resistant, or relapsing tumours. It is based on the manipulation of several mechanisms involved in the complex interplay between cancer cells and immune system that culminates in a form of immune-tolerance of tumour cells, favouring their expansion. Current immunotherapies are devoted enforcing the immune response against cancer cells and are represented by approaches employing vaccines, monoclonal antibodies, interleukins, checkpoint inhibitors, and chimeric antigen receptor (CAR)-T cells. Despite the undoubted potency of these treatments in some malignancies, many issues are being investigated to amplify the potential of application and to avoid side effects. In this review, we discuss how sphingolipids are involved in interactions between cancer cells and the immune system and how knowledge in this topic could be employed to enhance the efficacy of different immunotherapy approaches. In particular, we explore the following aspects: how sphingolipids are pivotal components of plasma membranes and could modulate the functionality of surface receptors expressed also by immune cells and thus their functionality; how sphingolipids are related to the release of bioactive mediators, sphingosine 1-phosphate, and ceramide that could significantly affect lymphocyte egress and migration toward the tumour milieu, in addition regulating key pathways needed to activate immune cells; given the renowned capability of altering sphingolipid expression and metabolism shown by cancer cells, how it is possible to employ sphingolipids as antigen targets.
Collapse
|
26
|
Lysophospholipids in Lung Inflammatory Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:373-391. [PMID: 33788203 DOI: 10.1007/978-3-030-63046-1_20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The lysophospholipids (LPLs) belong to a group of bioactive lipids that play pivotal roles in several physiological and pathological processes. LPLs are derivatives of phospholipids and consist of a single hydrophobic fatty acid chain, a hydrophilic head, and a phosphate group with or without a large molecule attached. Among the LPLs, lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are the simplest, and have been shown to be involved in lung inflammatory symptoms and diseases such as acute lung injury, asthma, and chronic obstructive pulmonary diseases. G protein-coupled receptors (GPCRs) mediate LPA and S1P signaling. In this chapter, we will discuss on the role of LPA, S1P, their metabolizing enzymes, inhibitors or agonists of their receptors, and their GPCR-mediated signaling in lung inflammatory symptoms and diseases, focusing specially on acute respiratory distress syndrome, asthma, and chronic obstructive pulmonary disease.
Collapse
|
27
|
LEDGF/p75-mediated chemoresistance of mixed-lineage leukemia involves cell survival pathways and super enhancer activators. Cancer Gene Ther 2021; 29:133-140. [PMID: 33795806 DOI: 10.1038/s41417-021-00319-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 02/12/2021] [Accepted: 02/26/2021] [Indexed: 11/08/2022]
Abstract
MLL is an aggressive subtype of leukemia with a poor prognosis that mostly affects pediatric patients. MLL-rearranged fusion proteins (MLLr) induce aberrant target gene expression resulting in leukemogenesis. MLL and its fusions are tethered to chromatin by LEDGF/p75, a transcriptional co-activator that specifically recognizes H3K36me2/3. LEDGF/p75 is ubiquitously expressed and associated with regulation of gene expression, autoimmune responses, and HIV replication. LEDGF/p75 was proven to be essential for leukemogenesis in MLL. Apart from MLL, LEDGF/p75 has been linked to lung, breast, and prostate cancer. Intriguingly, LEDGF/p75 interacts with Med-1, which co-localizes with BRD4. Both are known as co-activators of super-enhancers. Here, we describe LEDGF/p75-dependent chemoresistance of MLLr cell lines. Investigation of the underlying mechanism revealed a role of LEDGF/p75 in the cell cycle and in survival pathways and showed that LEDGF/p75 protects against apoptosis during chemotherapy. Remarkably, LEDGF/p75 levels also affected expression of BRD4 and Med1. Altogether, our data suggest a role of LEDGF/p75 in cancer survival, stem cell renewal, and activation of nuclear super enhancers.
Collapse
|
28
|
Kiknavelidze K, Shavdia M, Chikhladze N, Abshilava L, Messina M, Mautner G, Kelly G. NOX66 as Monotherapy, and in Combination With Carboplatin, in Patients With Refractory Solid Tumors: Phase Ia/b Study. Curr Ther Res Clin Exp 2021; 94:100631. [PMID: 34306271 PMCID: PMC8296080 DOI: 10.1016/j.curtheres.2021.100631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/22/2021] [Indexed: 11/23/2022] Open
Abstract
NOX66 contains idronoxil, formulated as a rectal suppository. CEP-1 is the first study to assess NOX66 in patients with refractory solid tumors. NOX66 was well tolerated at 400/800 mg as monotherapy and combined with carboplatin. The safety profile justifies continuation of the NOX66 clinical research program. Early results suggest most patients had stable disease by study end
Background Although oral and intravenous forms of idronoxil have been well tolerated, the safety of NOX66, with idronoxil formulated as a rectal suppository, is not known. This Phase Ia/b clinical study (protocol No. NOX66-001A), known as Chemotherapy Enhancement Program-1, is the first to assess NOX66 in patients with refractory solid tumors. Objective The study aimed to determine the safety profile of NOX66 both as a monotherapy and in combination with carboplatin, and to evaluate whether or not NOX66 has a meaningful anticancer effect when combined with carboplatin in this patient population. Methods Chemotherapy Enhancement Program-1 was a multicenter, open-label, nonrandomized, 2-dose cohort study of NOX66 as monotherapy (Phase Ia) and in combination with carboplatin (Phase Ib). Patients with refractory solid tumors who had stopped responding to standard treatments were eligible to participate. Twenty patients were screened and 19 enrolled in the study. They were divided into 2 groups: cohort 1 (n = 8) received 1 suppository daily (400 mg) and cohort 2 (n = 11) received 2 suppositories daily (800 mg) for 14 consecutive days followed by 7 days of rest. Patients who completed Phase Ia without significant toxicity continued to Phase Ib, where NOX66 was combined with carboplatin for up to 6x 28-day treatment cycles, with low-dose carboplatin (600 mg) for cycles 1B through 3B and standard dose carboplatin (900 mg) for cycles 4B through 6B. The main outcomes assessed were safety (n = 18) and efficacy signals (n = 14). Results NOX66 generally was well tolerated at 400 mg and 800 mg, both as monotherapy and in combination with carboplatin in patients with refractory solid tumors. The safety profile was consistent for oncology patients, with 77.8% experiencing at least 1 treatment-emergent adverse event. The most common adverse events were blood and lymphatic system disorders (44.4%), with only anemia considered as possibly related to NOX66. Although the study was primarily designed to assess safety and tolerability, the efficacy measurements demonstrated that most patients had stable disease or better by study end. Conclusions The favorable safety profile of NOX66 provides reassurance to justify continuation of clinical research. The efficacy findings are encouraging in terms of the chemosensitizing potential of NOX66 in refractory solid tumors. (Curr Ther Res Clin Exp. 2021; 82:XXX–XXX)
Collapse
Affiliation(s)
- Koba Kiknavelidze
- Oncology Unit, Z Tskhakaia West Georgia National Center of Interventional Medicine, Kutaisi, Georgia
| | | | - Nana Chikhladze
- First Clinic of Tbilisi State Medical University, Tbilisi, Georgia
| | - Lia Abshilava
- Medulla-Chemotherapy and Immunotherapy Center, Tbilisi, Georgia
| | | | | | - Graham Kelly
- Noxopharm Limited, Gordon, New South Wales, Australia
| |
Collapse
|
29
|
Ceramides and sphingosine-1-phosphate mediate the distinct effects of M1/M2-macrophage infusion on liver recovery after hepatectomy. Cell Death Dis 2021; 12:324. [PMID: 33771984 PMCID: PMC7998020 DOI: 10.1038/s41419-021-03616-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 02/01/2023]
Abstract
Post-hepatectomy liver dysfunction is a life-threatening morbidity that lacks efficient therapy. Bioactive lipids involved in macrophage polarization crucially regulate tissue injury and regeneration. Herein, we investigate the key bioactive lipids that mediate the cytotherapeutic potential of polarized-macrophage for post-hepatectomy liver dysfunction. Untargeted lipidomics identified elevation of ceramide (CER) metabolites as signature lipid species relevant to M1/M2 polarization in mouse bone-marrow-derived-macrophages (BMDMs). M1 BMDMs expressed a CER-generation-metabolic pattern, leading to elevation of CER; M2 BMDMs expressed a CER-breakdown-metabolic pattern, resulting in upregulation of sphingosine-1-phosphate (S1P). After infusing M1- or M2-polarized BMDMs into the mouse liver after hepatectomy, we found that M1-BMDM infusion increased M1 polarization and CER accumulation, resulting in exaggeration of hepatocyte apoptosis and liver dysfunction. Conversely, M2-BMDM infusion enhanced M2 polarization and S1P generation, leading to alleviation of liver dysfunction with improved hepatocyte proliferation. Treatment of exogenous CER and S1P or inhibition CER and S1P synthesis by siRNA targeting relevant enzymes further revealed that CER induced apoptosis while S1P promoted proliferation in post-hepatectomy primary hepatocytes. In conclusion, CER and S1P are uncovered as critical lipid mediators for M1- and M2-polarized BMDMs to promote injury and regeneration in the liver after hepatectomy, respectively. Notably, the upregulation of hepatic S1P induced by M2-BMDM infusion may have therapeutic potential for post-hepatectomy liver dysfunction.
Collapse
|
30
|
Perciani CT, Liu LY, Wood L, MacParland SA. Enhancing Immunity with Nanomedicine: Employing Nanoparticles to Harness the Immune System. ACS NANO 2021; 15:7-20. [PMID: 33346646 DOI: 10.1021/acsnano.0c08913] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The failure of immune responses to vaccines and dysfunctional immune responses to viral infection, tumor development, or neoantigens lead to chronic viral infection, tumor progression, or incomplete immune protection after vaccination. Thus, strategies to boost host immunity are a topic of intense research and development. Engineered nanoparticles (NPs) possess immunological properties and can be modified to promote improved local immune responses. Nanoparticle-based approaches have been employed to enhance vaccine efficacy and host immune responses to viral and tumor antigens, with impressive results. In this Perspective, we present an overview of studies, such as the one reported by Alam et al. in this issue of ACS Nano, in which virus-like particles have been employed to enhance immunity. We review the cellular cornerstones of effective immunity and discuss how NPs can harness these interactions to overcome the current obstacles in vaccinology and oncology. We also discuss the barriers to effective NP-mediated immune priming including (1) NP delivery to the site of interest, (2) the quality of response elicited, and (3) the potential of the response to overcome immune escape. Through this Perspective, we aim to highlight the value of nanomedicine not only in delivering therapies but also in coordinating the enhancement of host immune responses. We provide a forward-looking outlook for future NP-based approaches and how they could be tailored to promote this outcome.
Collapse
Affiliation(s)
- Catia T Perciani
- Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
| | - Lewis Y Liu
- Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
- Department of Immunology, University of Toronto, Medical Sciences Building, Room 6271, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Lawrence Wood
- Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
- Department of Immunology, University of Toronto, Medical Sciences Building, Room 6271, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Sonya A MacParland
- Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
- Department of Immunology, University of Toronto, Medical Sciences Building, Room 6271, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, Room 6271, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
31
|
Alkaline ceramidase family: The first two decades. Cell Signal 2020; 78:109860. [PMID: 33271224 DOI: 10.1016/j.cellsig.2020.109860] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 11/21/2022]
Abstract
Ceramidases are a group of enzymes that catalyze the hydrolysis of ceramide, dihydroceramide, and phytoceramide into sphingosine (SPH), dihydrosphingosine (DHS), and phytosphingosine (PHS), respectively, along with a free fatty acid. Ceramidases are classified into the acid, neutral, and alkaline ceramidase subtypes according to the pH optima for their catalytic activity. YPC1 and YDC1 were the first alkaline ceramidase genes to be identified and cloned from the yeast Saccharomyces cerevisiae two decades ago. Subsequently, alkaline ceramidase genes were identified from other species, including one Drosophila melanogaster ACER gene (Dacer), one Arabidopsis thaliana ACER gene (AtACER), three Mus musculus ACER genes (Acer1, Acer2, and Acer3), and three Homo sapiens ACER genes (ACER1, ACER2, and ACER3). The protein products of these genes constitute a large protein family, termed the alkaline ceramidase (ACER) family. All the biochemically characterized members of the ACER family are integral membrane proteins with seven transmembrane segments in the Golgi complex or endoplasmic reticulum, and they each have unique substrate specificity. An increasing number of studies suggest that the ACER family has diverse roles in regulating sphingolipid metabolism and biological processes. Here we discuss the discovery of the ACER family, the biochemical properties, structures, and catalytic mechanisms of its members, and its role in regulating sphingolipid metabolism and biological processes in yeast, insects, plants, and mammals.
Collapse
|
32
|
Prakash H, Upadhyay D, Bandapalli OR, Jain A, Kleuser B. Host sphingolipids: Perspective immune adjuvant for controlling SARS-CoV-2 infection for managing COVID-19 disease. Prostaglandins Other Lipid Mediat 2020; 152:106504. [PMID: 33147503 PMCID: PMC7605809 DOI: 10.1016/j.prostaglandins.2020.106504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 01/12/2023]
Abstract
That Sphingolipid derivatives are promising drug candidates for the management of novel COVID-19 disease. C-1P based tailoring of Th1 effector immunity for the eradication of infection is a translationally viable approach and deserves immediate attention. That C-1P would promote the killing of infected cells and resolve infection in moderate to severely infected cases. Ceramide derivatives can be exploited as drug candidates for controlling SARS-CoV-2 against novel COVID-19 disease.
Sphingolipids are potent bioactive agents involved in the pathogenesis of various respiratory bacterial infections. To date, several sphingolipid derivatives are known, but S1P (Sphingosine-1-phosphate) and Ceramide are the best-studied sphingolipid derivatives in the context of human diseases. These are membrane-bound lipids that influence host-pathogen interactions. Based on these features, we believe that sphingolipids might control SARS-CoV-2 infection in the host. SARS-CoV-2 utilizes the ACE-II receptor (Angiotensin-converting enzyme II receptor) on epithelial cells for its entry and replication. Activation of the ACE-II receptor is indirectly associated with the activation of S1P Receptor 1 signaling which is associated with IL-6 driven fibrosis. This is expected to promote pathological responses during SARS-CoV-2 infection in COVID-19 cases. Given this, mitigating S1P signaling by application of either S1P Lyase (SPL) or S1P analog (Fingolimod / FTY720) seems to be potential approach for controlling these pathological outcomes. However, due to the immunosuppressive nature of FTY720, it can modulate hyper-inflammatory responses and only provide symptomatic relief, which may not be sufficient for controlling the novel COVID-19 infection. Since Th1 effector immune responses are essential for the clearance of infection, we believe that other sphingolipid derivatives like Cermaide-1 Phosphate with antiviral potential and adjuvant immune potential can potentially control SARS-CoV-2 infection in the host by its ability in enhancing autophagy and antigen presentation by DC to promote T cell response which can be helpful in controlling SARS-CoV-2 infection in novel COVID-19 patients.
Collapse
Affiliation(s)
- Hridayesh Prakash
- Amity Institute of Virology and Immunology, Amity University, Noida, India.
| | - Dilip Upadhyay
- Amity Institute of Virology and Immunology, Amity University, Noida, India
| | | | - Aklank Jain
- Department of Zoology, Central University of Punjab, Bathinda, India
| | - Burkhard Kleuser
- Institute of Nutritional Science, Department of Nutritional Toxicology, University of Potsdam Nuthetal, Germany
| |
Collapse
|
33
|
Shnaider PV, Ivanova OM, Malyants IK, Anufrieva KS, Semenov IA, Pavlyukov MS, Lagarkova MA, Govorun VM, Shender VO. New Insights into Therapy-Induced Progression of Cancer. Int J Mol Sci 2020; 21:E7872. [PMID: 33114182 PMCID: PMC7660620 DOI: 10.3390/ijms21217872] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
The malignant tumor is a complex heterogeneous set of cells functioning in a no less heterogeneous microenvironment. Like any dynamic system, cancerous tumors evolve and undergo changes in response to external influences, including therapy. Initially, most tumors are susceptible to treatment. However, remaining cancer cells may rapidly reestablish the tumor after a temporary remission. These new populations of malignant cells usually have increased resistance not only to the first-line agent, but also to the second- and third-line drugs, leading to a significant decrease in patient survival. Multiple studies describe the mechanism of acquired therapy resistance. In past decades, it became clear that, in addition to the simple selection of pre-existing resistant clones, therapy induces a highly complicated and tightly regulated molecular response that allows tumors to adapt to current and even subsequent therapeutic interventions. This review summarizes mechanisms of acquired resistance, such as secondary genetic alterations, impaired function of drug transporters, and autophagy. Moreover, we describe less obvious molecular aspects of therapy resistance in cancers, including epithelial-to-mesenchymal transition, cell cycle alterations, and the role of intercellular communication. Understanding these molecular mechanisms will be beneficial in finding novel therapeutic approaches for cancer therapy.
Collapse
Affiliation(s)
- Polina V. Shnaider
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (P.V.S.); (O.M.I.); (K.S.A.); (M.A.L.)
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
- Faculty of Biology, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Olga M. Ivanova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (P.V.S.); (O.M.I.); (K.S.A.); (M.A.L.)
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
| | - Irina K. Malyants
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
- Faculty of Chemical-Pharmaceutical Technologies and Biomedical Drugs, Mendeleev University of Chemical Technology of Russia, Moscow 125047, Russia
| | - Ksenia S. Anufrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (P.V.S.); (O.M.I.); (K.S.A.); (M.A.L.)
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
- Moscow Institute of Physics and Technology (State University), Dolgoprudny 141701, Russia
| | - Ilya A. Semenov
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
| | - Marat S. Pavlyukov
- Laboratory of Membrane Bioenergetics, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia;
| | - Maria A. Lagarkova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (P.V.S.); (O.M.I.); (K.S.A.); (M.A.L.)
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
| | - Vadim M. Govorun
- Laboratory of Simple Systems, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia;
| | - Victoria O. Shender
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (P.V.S.); (O.M.I.); (K.S.A.); (M.A.L.)
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
- Laboratory of Molecular Oncology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
34
|
Cancer stem cells and ceramide signaling: the cutting edges of immunotherapy. Mol Biol Rep 2020; 47:8101-8111. [PMID: 32885363 DOI: 10.1007/s11033-020-05790-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022]
Abstract
The multipotent, self renewing "cancer stem cells" (CSCs), a small population within tumor microenvironment facilitates transformed cells to grow and propagate within the body. The CSCs are discovered as resistant to the chemotherapeutic drug with distinct immunological characteristics. In recent years, immunologically targeting CSCs have emerged as an integral part of effective and successful cancer therapy. CSCs notably exhibit dysregulation in conventional sub-cellular sphingolipid metabolism. Recently, ceramide decaying enzymes have been shown to activate alternative ceramide signaling pathways leading to reduction in efficacy of the chemotherapeutic drugs. Therefore, a control over ceramide mediated modulations of CSCs offers an attractive dimension of effective cancer treatment strategy in future. In this review, we focused on the recent findings on broad spectrum of ceramide mediated signaling in CSCs within the tumor niche and their role in potential cancer immunotherapy.
Collapse
|
35
|
Li H, Xing X, Zhang X, Li L, Jiang Z, Wang T, Huang X, Wang X, Zhang L, Sun L. Effects of triptolide on the sphingosine kinase - Sphingosine-1-phosphate signaling pathway in colitis-associated colon cancer. Int Immunopharmacol 2020; 88:106892. [PMID: 32810834 DOI: 10.1016/j.intimp.2020.106892] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUNDS Triptolide (TP) exhibits effective activity against colon cancer in multiple preclinical models, but the mechanisms underlying the observed effects are not fully understood. Sphingosine-1-phosphate (S1P) is a potent bioactive sphingolipid involved in the regulation of colon cancer progression. The aim of this study was to investigate the effect of TP on the sphingosine kinase (SPHK)-S1P signaling pathway in colitis-associated colon cancer. METHODS An azoxymethane (AOM)/dextran sulfate sodium (DSS) mouse model and the THP-1 cell line were used to evaluate the therapeutic effects and mechanisms of TP in colitis-associated colon cancer (CACC). Various molecular cell biology experiments, including Western blotting, real-time PCR and immunofluorescence, were used to obtain relevant experimental data. A liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was also established to detect the levels of S1P in tissue and plasma. RESULTS In the AOM/DSS mouse model, TP treatment induced a dose-dependent decrease in tumor incidence and inhibited macrophage recruitment and M2 polarization in the tumors. TP also efficiently decreased the S1P levels and SPHK1/S1PR1/S1PR2 expression and significantly inhibited activation of the S1P-mediated phosphorylation of ERK protein in macrophages. CONCLUSIONS The results indicated that TP might influence the recruitment and polarization of tumor-associated macrophages by suppressing the SPHK-S1P signaling pathway.
Collapse
Affiliation(s)
- Han Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Xin Xing
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Xi Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Liping Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Nanjing 210009, China
| | - Tao Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Nanjing 210009, China
| | - Xin Huang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Xinzhi Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Nanjing 210009, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Nanjing 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
36
|
Zhong L, Xie L, Yang Z, Li L, Song S, Cao D, Liu Y. Prognostic value of S1PR1 and its correlation with immune infiltrates in breast and lung cancers. BMC Cancer 2020; 20:766. [PMID: 32799825 PMCID: PMC7429796 DOI: 10.1186/s12885-020-07278-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/09/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Sphingosine-1-phosphate receptor (S1PR1) is involved in vascular development, a key process in tumorigenesis. This study aimed to evaluate its roles in tumor development and prognosis. METHODS S1PR1 expression levels were analyzed using TIMER and Oncomine database, and the prognostic significance of S1PR1 was assessed using PrognoScan and Kaplan-Meier plotter databases. The relationship between S1PR1 and tumor-infiltrated immune cells was analyzed using TIMER. RESULTS S1PR1 expression was remarkably lower in breast and lung cancer tissues than in the corresponding normal tissues. Lower expression was related to poor overall survival and disease-free survival in breast invasive carcinoma (BRCA), lung adenocarcinoma (LUAD), and lung squamous cell carcinoma (LUSC). A functional network analysis confirmed the function of S1PR1 in regulating vasculogenesis. In addition, S1PR1 levels were significantly negative with regard to the tumor purity of BRCA (r = - 0.508, P = 1.76e-66), LUAD (r = - 0.353, P = 6.05e-16), and LUSC (r = - 0.402, P = - 5.20e-20). Furthermore, S1PR1 levels were significantly positive with regard to infiltrating CD8+ (r = 0.38, P = 5.91e-35) and CD4+ T cells (r = 0.335, P = 1.03e-26), macrophages (r = 0.219, P = 3.67e-12), neutrophils (r = 0.168, P = 2.03e-7), and dendritic cells (DCs) (r = 0.208, P = 9.14e-11) in BRCA; S1PR1 levels were significantly positive with regard to CD8+ T cells (r = 0.308, P = 3.61e-12), macrophages (r = 0.376, P = 1.01e-17), neutrophils (r = 0.246, P = 4.15e-8), and DCs (r = 0.207, P = 4.16e-6) in LUAD; and positive with regard to B cells (r = 0.356, P = 1.57e-15), CD8+ (r = 0.459, P = 3.83e-26) and CD4+ T cells (r = 0.338, P = 3.98e-14), macrophages (r = 0.566, P = 2.61e-45), neutrophils (r = 0.453, P = 1.79e-25), and DCs (r = 0.56, P = 2.12e-40) in LUSC. CONCLUSIONS S1PR1 levels are positively correlated with multiple immune markers in breast and lung cancer. These observed correlations between S1PR1 and the prognosis and immune cell infiltration provide a foundation for further research on its immunomodulatory role in cancer.
Collapse
Affiliation(s)
- Limei Zhong
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, No. 466 Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong Province, China
| | - Linling Xie
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, No. 16 Airport Road, Baiyun District, Guangzhou, 510407, China
| | - Zhiyong Yang
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, No. 466 Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong Province, China
| | - Lijuan Li
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, No. 466 Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong Province, China
| | - Shaohua Song
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, No. 466 Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong Province, China
| | - Donglin Cao
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, No. 466 Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong Province, China.
| | - Yufeng Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, No. 16 Airport Road, Baiyun District, Guangzhou, 510407, China.
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China.
| |
Collapse
|
37
|
Abstract
Early mast cell (MC) infiltration has been reported in a wide range of human and animal tumors particularly malignant melanoma and breast and colorectal cancer. The consequences of their presence in the tumor microenvironment (TME) or at their margins still remain unclear as it is associated with a good or poor prognosis based on the type and anatomical site of the tumor. Within the tumor, MC interactions occur with infiltrated immune cells, tumor cells, and extracellular matrix (ECM) through direct cell-to-cell interactions or release of a broad range of mediators capable of remodeling the TME. MCs actively contribute to angiogenesis and induce neovascularization by releasing the classical proangiogenic factors including VEGF, FGF-2, PDGF, and IL-6, and nonclassical proangiogenic factors mainly proteases including tryptase and chymase. MCs support tumor invasiveness by releasing a broad range of matrix metalloproteinases (MMPs). MC presence within the tumor gained additional significance when it was assumed that controlling its activation by tyrosine kinase inhibitors (imatinib and masitinib) and tryptase inhibitors (gabexate and nafamostat mesylate) or controlling their interactions with other cell types may have therapeutic benefit.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Frank A Redegeld
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands.
| |
Collapse
|
38
|
Abstract
Sphingosine-1-phosphate (S1P) can regulate several physiological and pathological processes. S1P signaling via its cell surface receptor S1PR1 has been shown to enhance tumorigenesis and stimulate growth, expansion, angiogenesis, metastasis, and survival of cancer cells. S1PR1-mediated tumorigenesis is supported and amplified by activation of downstream effectors including STAT3, interleukin-6, and NF-κB networks. S1PR1 signaling can also trigger various other signaling pathways involved in carcinogenesis including activation of PI3K/AKT, MAPK/ERK1/2, Rac, and PKC/Ca, as well as suppression of cyclic adenosine monophosphate (cAMP). It also induces immunological tolerance in the tumor microenvironment, while the immunosuppressive function of S1PR1 can also lead to the generation of pre-metastatic niches. Some tumor cells upregulate S1PR1 signaling pathways, which leads to drug resistant cancer cells, mainly through activation of STAT3. This signaling pathway is also implicated in some inflammatory conditions leading to the instigation of inflammation-driven cancers. Furthermore, it can also increase survival via induction of anti-apoptotic pathways, for instance, in breast cancer cells. Therefore, S1PR1 and its signaling pathways can be considered as potential anti-tumor therapeutic targets, alone or in combination therapies. Given the oncogenic nature of S1PR1 and its distribution in a variety of cancer cell types along with its targeting advantages over other molecules of this family, S1PR1 should be considered a favorable target in therapeutic approaches to cancer. This review describes the role of S1PR1 in cancer development and progression, specifically addressing breast cancer, glioma, and hematopoietic malignancies. We also discuss the potential use of S1P signaling modulators as therapeutic targets in cancer therapy.
Collapse
|
39
|
Zhang L, Dong Y, Wang Y, Hu W, Dong S, Chen Y. Sphingosine-1-phosphate (S1P) receptors: Promising drug targets for treating bone-related diseases. J Cell Mol Med 2020; 24:4389-4401. [PMID: 32155312 PMCID: PMC7176849 DOI: 10.1111/jcmm.15155] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/22/2020] [Accepted: 02/01/2020] [Indexed: 12/20/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a natural bioactive lipid molecule and a common first or second messenger in the cardiovascular and immune systems. By binding with its receptors, S1P can serve as mediator of signalling during cell migration, differentiation, proliferation and apoptosis. Although the predominant role of S1P in bone regeneration has been noted in many studies, this role is not as well-known as its roles in the cardiovascular and immune systems. In this review, we summarize previous research on the role of S1P receptors (S1PRs) in osteoblasts and osteoclasts. In addition, S1P is regarded as a bridge between bone resorption and formation, which brings hope to patients with bone-related diseases. Finally, we discuss S1P and its receptors as therapeutic targets for treating osteoporosis, inflammatory osteolysis and bone metastasis based on the biological effects of S1P in osteoclastic/osteoblastic cells, immune cells and tumour cells.
Collapse
Affiliation(s)
- Lincheng Zhang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,Battalion One of Basic Medical Sciences, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yutong Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,Battalion One of Basic Medical Sciences, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yiran Wang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wenhui Hu
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yueqi Chen
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
40
|
Riboni L, Abdel Hadi L, Navone SE, Guarnaccia L, Campanella R, Marfia G. Sphingosine-1-Phosphate in the Tumor Microenvironment: A Signaling Hub Regulating Cancer Hallmarks. Cells 2020; 9:E337. [PMID: 32024090 PMCID: PMC7072483 DOI: 10.3390/cells9020337] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
As a key hub of malignant properties, the cancer microenvironment plays a crucial role intimately connected to tumor properties. Accumulating evidence supports that the lysophospholipid sphingosine-1-phosphate acts as a key signal in the cancer extracellular milieu. In this review, we have a particular focus on glioblastoma, representative of a highly aggressive and deleterious neoplasm in humans. First, we highlight recent advances and emerging concepts for how tumor cells and different recruited normal cells contribute to the sphingosine-1-phosphate enrichment in the cancer microenvironment. Then, we describe and discuss how sphingosine-1-phosphate signaling contributes to favor cancer hallmarks including enhancement of proliferation, stemness, invasion, death resistance, angiogenesis, immune evasion and, possibly, aberrant metabolism. We also discuss the potential of how sphingosine-1-phosphate control mechanisms are coordinated across distinct cancer microenvironments. Further progress in understanding the role of S1P signaling in cancer will depend crucially on increasing knowledge of its participation in the tumor microenvironment.
Collapse
Affiliation(s)
- Laura Riboni
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Loubna Abdel Hadi
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
- Department of Clinical Sciences and Community Health, University of Milan, 20100 Milan, Italy
| | - Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| |
Collapse
|
41
|
Nadella V, Sharma L, Kumar P, Gupta P, Gupta UD, Tripathi S, Pothani S, Qadri SSYH, Prakash H. Sphingosine-1-Phosphate (S-1P) Promotes Differentiation of Naive Macrophages and Enhances Protective Immunity Against Mycobacterium tuberculosis. Front Immunol 2020; 10:3085. [PMID: 32038629 PMCID: PMC6993045 DOI: 10.3389/fimmu.2019.03085] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022] Open
Abstract
Sphingosine-1-phosphate (S-1P) is a key sphingolipid involved in the pathobiology of various respiratory diseases. We have previously demonstrated the significance of S-1P in controlling non-pathogenic mycobacterial infection in macrophages, and here we demonstrate the therapeutic potential of S-1P against pathogenic Mycobacterium tuberculosis (H37Rv) in the mouse model of infection. Our study revealed that S-1P is involved in the expression of iNOS proteins in macrophages, their polarization toward M1 phenotype, and secretion of interferon (IFN)-γ during the course of infection. S-1P is also capable of enhancing infiltration of pulmonary CD11b+ macrophages and expression of S-1P receptor-3 (S-1PR3) in the lungs during the course of infection. We further revealed the influence of S-1P on major signaling components of inflammatory signaling pathways during M. tuberculosis infection, thus highlighting antimycobacterial potential of S-1P in animals. Our data suggest that enhancing S-1P levels by sphingolipid mimetic compounds/drugs can be used as an immunoadjuvant for boosting immunity against pathogenic mycobacteria.
Collapse
Affiliation(s)
- Vinod Nadella
- Laboratory of Translational Medicine, School of Life Science, University of Hyderabad, Hyderabad, India
| | - Lalita Sharma
- Laboratory of Translational Medicine, School of Life Science, University of Hyderabad, Hyderabad, India
| | - Pankaj Kumar
- Laboratory of Translational Medicine, School of Life Science, University of Hyderabad, Hyderabad, India
| | - Pushpa Gupta
- Department of Experimental Animal Facility, National JALMA Institute for Leprosy and Other Mycobacterial Disease, Agra, India
| | - Umesh D Gupta
- Department of Experimental Animal Facility, National JALMA Institute for Leprosy and Other Mycobacterial Disease, Agra, India
| | - Srikant Tripathi
- Department of Bacteriology, National Institute of Research in Tuberculosis, Chennai, India
| | - Suresh Pothani
- National Animal Resource Facility for Biomedical Research, National Institute of Nutrition, Indian Council of Medical Research Hyderabad, Hyderabad, India
| | - S S Y H Qadri
- National Animal Resource Facility for Biomedical Research, National Institute of Nutrition, Indian Council of Medical Research Hyderabad, Hyderabad, India
| | - Hridayesh Prakash
- Laboratory of Translational Medicine, School of Life Science, University of Hyderabad, Hyderabad, India
| |
Collapse
|
42
|
Graber K, Khan F, Glück B, Weigel C, Marzo S, Doshi H, Ehrhardt C, Heller R, Gräler M, Henke A. The role of sphingosine-1-phosphate signaling in HSV-1-infected human umbilical vein endothelial cells. Virus Res 2020; 276:197835. [DOI: 10.1016/j.virusres.2019.197835] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/06/2019] [Accepted: 12/06/2019] [Indexed: 01/14/2023]
|
43
|
Leufven E, Bruserud Ø. Immunosuppression and Immunotargeted Therapy in Acute Myeloid Leukemia - The Potential Use of Checkpoint Inhibitors in Combination with Other Treatments. Curr Med Chem 2019; 26:5244-5261. [PMID: 30907305 DOI: 10.2174/0929867326666190325095853] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Immunotherapy by using checkpoint inhibitors is now tried in the treatment of several malignancies, including Acute Myeloid Leukemia (AML). The treatment is tried both as monotherapy and as a part of combined therapy. METHODS Relevant publications were identified through literature searches in the PubMed database. We searched for (i) original articles describing the results from clinical studies of checkpoint inhibition; (ii) published articles describing the immunocompromised status of AML patients; and (iii) published studies of antileukemic immune reactivity and immunotherapy in AML. RESULTS Studies of monotherapy suggest that checkpoint inhibition has a modest antileukemic effect and complete hematological remissions are uncommon, whereas combination with conventional chemotherapy increases the antileukemic efficiency with acceptable toxicity. The experience with a combination of different checkpoint inhibitors is limited. Thalidomide derivatives are referred to as immunomodulatory drugs and seem to reverse leukemia-induced immunosuppression, but in addition, they have direct inhibitory effects on the AML cells. The combination of checkpoint targeting and thalidomide derivatives thus represents a strategy for dual immunotargeting together with a direct antileukemic effect. CONCLUSION Checkpoint inhibitors are now tried in AML. Experimental studies suggest that these inhibitors should be combined with immunomodulatory agents (i.e. thalidomide derivatives) and/or new targeted or conventional antileukemic treatment. Such combinations would allow dual immunotargeting (checkpoint inhibitor, immunomodulatory agents) together with a double/triple direct targeting of the leukemic cells.
Collapse
Affiliation(s)
- Eva Leufven
- Department of Clinical Science, University of Bergen, Jonas Lies vei 87, N-5020 Bergen, Norway
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, Jonas Lies vei 87, N-5020 Bergen, Norway.,Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021, Bergen, Norway
| |
Collapse
|
44
|
Vijayan Y, Lankadasari MB, Harikumar KB. Acid Ceramidase: A Novel Therapeutic Target in Cancer. Curr Top Med Chem 2019; 19:1512-1520. [PMID: 30827244 DOI: 10.2174/1568026619666190227222930] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 01/22/2019] [Accepted: 02/18/2019] [Indexed: 12/15/2022]
Abstract
Sphingolipids are important constituents of the eukaryotic cell membrane which govern various signaling pathways related to different aspects of cell survival. Ceramide and Sphingosine are interconvertible sphingolipid metabolites, out of which Ceramide is pro-apoptotic and sphingosine is anti-apoptotic in nature. The conversion of ceramide to sphingosine is mediated by Acid Ceramidase (ASAH1) thus maintaining a rheostat between a tumor suppressor and a tumor promoter. This rheostat is completely altered in many tumors leading to uncontrolled proliferation. This intriguing property of ASAH1 can be used by cancer cells to their advantage, by increasing the expression of the tumor promoter, sphingosine inside cells, thus creating a favorable environment for cancer growth. The different possibilities through which this enzyme serves its role in formation, progression and resistance of different types of cancers will lead to the possibility of making Acid Ceramidase a promising drug target. This review discusses the current understanding of the role of acid ceramidase in cancer progression, metastasis and resistance, strategies to develop novel natural and synthetic inhibitors of ASAH1 and their usefulness in cancer therapy.
Collapse
Affiliation(s)
- Yadu Vijayan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India.,Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Manendra Babu Lankadasari
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India.,Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Kuzhuvelil B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India
| |
Collapse
|
45
|
Maceyka M, Rohrbach T, Milstien S, Spiegel S. Role of Sphingosine Kinase 1 and Sphingosine-1-Phosphate Axis in Hepatocellular Carcinoma. Handb Exp Pharmacol 2019; 259:3-17. [PMID: 31321542 DOI: 10.1007/164_2019_217] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is primarily diagnosed in the latter stages of disease progression and is the third leading cause of cancer deaths worldwide. Thus, there is a need to find biomarkers of early HCC as well as the development of more effective treatments for the disease. Sphingosine-1-phosphate (S1P) is a pleiotropic lipid signaling molecule produced by two isoforms of sphingosine kinase (SphK1 and SphK2) that is involved in regulation of many aspects of mammalian physiology and pathophysiology, including inflammation, epithelial and endothelial barrier function, cancer, and metastasis, among many others. Abundant evidence indicates that SphK1 and S1P promote cancer progression and metastasis in multiple types of cancers. However, the role of SphK/S1P in HCC is less well studied. Here, we review the current state of knowledge of SphKs and S1P in HCC, including evidence for the correlation of SphK1 expression and S1P levels with progression of HCC and negative outcomes, and discuss how this information could lead to the design of more effective diagnostic and treatment modalities for HCC.
Collapse
Affiliation(s)
- Michael Maceyka
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Timothy Rohrbach
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Sheldon Milstien
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
46
|
Hu C, Chen M, Jiang R, Guo Y, Wu M, Zhang X. Exosome-related tumor microenvironment. J Cancer 2018; 9:3084-3092. [PMID: 30210631 PMCID: PMC6134819 DOI: 10.7150/jca.26422] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 07/05/2018] [Indexed: 12/29/2022] Open
Abstract
The tumor microenvironment (tumor cells are located in the internal and external environment) is vital for the occurrence, growth and metastasis of tumors. An increasing number of studies have shown that exosomes are closely related to the tumor microenvironment. The mechanisms involved, however, are unclear. The focus of this review is on the exosome-related tumor microenvironment and other relevant factors, such as hypoxia, inflammation and angiogenesis. Many studies have suggested that exosomes are important mediators of metastasis, angiogenesis, and immune modulation in the tumor microenvironment. Additionally, exosomes can be isolated from bodily fluids of cancer patients, including urine, blood, saliva, milk, tumor effusion, cerebrospinal fluid, amniotic fluid and so on. Consequently, exosomes are potential biomarkers for clinical predictions and are also good drug carriers because they can cross the biofilm without triggering an immune response. Collectively, these findings illustrate that exosomes are crucial for developing potential targets for a new generation of pharmaceutical therapies that would improve the tumor microenvironment.
Collapse
Affiliation(s)
- Cheng Hu
- School of Medicine and Life Sciences , Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, P.R. China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, P.R. China
| | - Meijuan Chen
- School of Medicine and Life Sciences , Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, P.R. China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, P.R. China
| | - Rilei Jiang
- School of Medicine and Life Sciences , Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, P.R. China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, P.R. China
| | - Yuanyuan Guo
- School of Medicine and Life Sciences , Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, P.R. China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, P.R. China
| | - Mianhua Wu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, P.R. China
| | - Xu Zhang
- School of Medicine and Life Sciences , Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, P.R. China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, P.R. China
| |
Collapse
|
47
|
Lysophospholipid Signaling in the Epithelial Ovarian Cancer Tumor Microenvironment. Cancers (Basel) 2018; 10:cancers10070227. [PMID: 29987226 PMCID: PMC6071084 DOI: 10.3390/cancers10070227] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/03/2018] [Accepted: 07/05/2018] [Indexed: 12/12/2022] Open
Abstract
As one of the important cancer hallmarks, metabolism reprogramming, including lipid metabolism alterations, occurs in tumor cells and the tumor microenvironment (TME). It plays an important role in tumorigenesis, progression, and metastasis. Lipids, and several lysophospholipids in particular, are elevated in the blood, ascites, and/or epithelial ovarian cancer (EOC) tissues, making them not only useful biomarkers, but also potential therapeutic targets. While the roles and signaling of these lipids in tumor cells are extensively studied, there is a significant gap in our understanding of their regulations and functions in the context of the microenvironment. This review focuses on the recent study development in several oncolipids, including lysophosphatidic acid and sphingosine-1-phosphate, with emphasis on TME in ovarian cancer.
Collapse
|
48
|
Abstract
Sphingosine kinases (SK1 and SK2) are key, druggable targets within the sphingolipid metabolism pathway that promote tumor growth and pathologic inflammation. A variety of isozyme-selective and dual inhibitors of SK1 and SK2 have been described in the literature, and at least one compound has reached clinical testing in cancer patients. In this chapter, we will review the rationale for targeting SKs and summarize the preclinical and emerging clinical data for ABC294640 as the first-in-class selective inhibitor of SK2.
Collapse
|
49
|
Zilch A, Rien C, Weigel C, Huskobla S, Glück B, Spengler K, Sauerbrei A, Heller R, Gräler M, Henke A. Influence of sphingosine-1-phosphate signaling on HCMV replication in human embryonal lung fibroblasts. Med Microbiol Immunol 2018; 207:227-242. [DOI: 10.1007/s00430-018-0543-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 04/17/2018] [Indexed: 12/16/2022]
|
50
|
Wang S, Liang Y, Chang W, Hu B, Zhang Y. Triple Negative Breast Cancer Depends on Sphingosine Kinase 1 (SphK1)/Sphingosine-1-Phosphate (S1P)/Sphingosine 1-Phosphate Receptor 3 (S1PR3)/Notch Signaling for Metastasis. Med Sci Monit 2018; 24:1912-1923. [PMID: 29605826 PMCID: PMC5894569 DOI: 10.12659/msm.905833] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Triple negative breast cancer (TNBC) has a more aggressive recurrence. Previous reports have demonstrated that sphingosine kinase 1 (SphK1) is a crucial regulator of breast cancer progression. However, the correlation of SphK1 with clinical prognosis has been poorly investigated. Thus, we aimed to elaborate the role of SphK1 in TNBC metastasis. Material/Methods We first determined the level of SphK1 in breast cancer tissue samples and breast cancer cells. Furthermore, the expression of HER2 and phosphor-SphK1 (pSphK1) in human breast cancer tissue samples was determined by immunohistochemical analysis. Associations between SphK1 and clinical parameters of tumors were analyzed. The activity of SphK1 was measured by fluorescence analysis. Extracellular sphingosine-1-phosphate (S1P) was detected using an ELISA kit. Associations between SphK1 and metastasis potential were analyzed by Transwell assay. Results Levels of SphK1 in TNBC patients were significantly higher than levels in other patients with other breast tumors. The expression of SphK1 was positively correlated with poor overall survival (OS) and progression-free survival (PFS), as well as poor response to 5-FU and doxorubicin. The depression of SphK1 thus could repress the Notch signaling pathway, reduce migration, and invasion of TNBC cells in vivo and in vitro. Furthermore, silencing of SphK1 by Ad-SPHK1-siRNA or SphK1 inhibitor PF543 sensitized TNBCs to 5-FU and doxorubicin. Our results also indicated that SphK1 inhibition could effectively counteracts tumors metastasis via Notch signaling pathways, indicating a potentially anti-tumor strategy in TNBC. Conclusions We found that elevated levels of pSphK1 were positive correlation with high expression of S1P, which in turn promoted metastasis of TNBC through S1P/S1PR3/Notch signaling pathway.
Collapse
Affiliation(s)
- Shushu Wang
- Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China (mainland)
| | - Yueyang Liang
- Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China (mainland)
| | - Wenxiao Chang
- Outpatient Department of Stomatology, Shan Xi Da Yi Hospital, Taiyuan, Shanxi, China (mainland)
| | - Baoquan Hu
- Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China (mainland)
| | - Yi Zhang
- Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China (mainland)
| |
Collapse
|