1
|
Limbu KR, Chhetri RB, Kim S, Shrestha J, Oh YS, Baek DJ, Park EY. Targeting sphingosine 1-phosphate and sphingosine kinases in pancreatic cancer: mechanisms and therapeutic potential. Cancer Cell Int 2024; 24:353. [PMID: 39462385 PMCID: PMC11514880 DOI: 10.1186/s12935-024-03535-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Pancreatic cancer is known to be the most lethal cancer. Fewer new treatments are being developed for pancreatic cancer as compared to other cancers. The bioactive lipid S1P, which is mainly regulated by sphingosine kinase 1 (SK1) and sphingosine kinase 2 (SK2) enzymes, plays significant roles in pancreatic cancer initiation and exacerbation. S1P controls many signaling pathways to modulate the progression of pancreatic cancer through the G-coupled receptor S1PR1-5. Several papers reporting amelioration of pancreatic cancer via modulation of S1P levels or downstream signaling pathways have previously been published. In this paper, for the first time, we have reviewed the results of previous studies to understand how S1P and its receptors contribute to the development of pancreatic cancer, and whether S1P can be a therapeutic target. In addition, we have also reviewed papers dealing with the effects of SK1 and SK2, which are kinases that regulate the level of S1P, on the pathogenesis of pancreatic cancer. We have also listed available drugs that particularly focus on S1P, S1PRs, SK1, and SK2 for the treatment of pancreatic cancer. Through this review, we would like to suggest that the SK/S1P/S1PR signaling system can be an important target for treating pancreatic cancer, where a new treatment target is desperately warranted.
Collapse
Affiliation(s)
- Khem Raj Limbu
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | | | - Subin Kim
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | - Jitendra Shrestha
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, Seongnam, 13135, South Korea
| | - Dong Jae Baek
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| | - Eun-Young Park
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Mohammed S, Bindu A, Viswanathan A, Harikumar KB. Sphingosine 1-phosphate signaling during infection and immunity. Prog Lipid Res 2023; 92:101251. [PMID: 37633365 DOI: 10.1016/j.plipres.2023.101251] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Sphingolipids are essential components of all eukaryotic membranes. The bioactive sphingolipid molecule, Sphingosine 1-Phosphate (S1P), regulates various important biological functions. This review aims to provide a comprehensive overview of the role of S1P signaling pathway in various immune cell functions under different pathophysiological conditions including bacterial and viral infections, autoimmune disorders, inflammation, and cancer. We covered the aspects of S1P pathways in NOD/TLR pathways, bacterial and viral infections, autoimmune disorders, and tumor immunology. This implies that targeting S1P signaling can be used as a strategy to block these pathologies. Our current understanding of targeting various components of S1P signaling for therapeutic purposes and the present status of S1P pathway inhibitors or modulators in disease conditions where the host immune system plays a pivotal role is the primary focus of this review.
Collapse
Affiliation(s)
- Sabira Mohammed
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India
| | - Anu Bindu
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India
| | - Arun Viswanathan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India; Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Kuzhuvelil B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India.
| |
Collapse
|
5
|
Kim KM, Shin EJ, Yang JH, Ki SH. Integrative roles of sphingosine kinase in liver pathophysiology. Toxicol Res 2023; 39:549-564. [PMID: 37779595 PMCID: PMC10541397 DOI: 10.1007/s43188-023-00193-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 10/03/2023] Open
Abstract
Bioactive sphingolipids and enzymes that metabolize sphingolipid-related substances have been considered as critical messengers in various signaling pathways. One such enzyme is the crucial lipid kinase, sphingosine kinase (SphK), which mediates the conversion of sphingosine to the potent signaling substance, sphingosine-1-phosphate. Several studies have demonstrated that SphK metabolism is strictly regulated to maintain the homeostatic balance of cells. Here, we summarize the role of SphK in the course of liver disease and illustrate its effects on both physiological and pathological conditions of the liver. SphK has been implicated in a variety of liver diseases, such as steatosis, liver fibrosis, hepatocellular carcinoma, and hepatic failure. This study may advance the understanding of the cellular and molecular foundations of liver disease and establish therapeutic approaches via SphK modulation.
Collapse
Affiliation(s)
- Kyu Min Kim
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju, 61452 Republic of Korea
| | - Eun Jin Shin
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju, 61452 Republic of Korea
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-Do 58245 Republic of Korea
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, 309 Pilmun-Daero, Dong-Gu, Gwangju, 61452 Republic of Korea
| |
Collapse
|
6
|
Kim SB, Lee T, Oh YS, Park E, Baek DJ. Synthesis and biological investigation of protein phosphatase
2A
‐activating compounds with dimeric tail as non‐small cell lung cancer cell death agents. B KOREAN CHEM SOC 2022. [DOI: 10.1002/bkcs.12661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Su Bin Kim
- College of Pharmacy Mokpo National University Jeonnam Korea
| | - Taeho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences Kyungpook National University Daegu Korea
| | - Yoon Sin Oh
- Department of Food and Nutrition Eulji University Seongnam Korea
| | - Eun‐Young Park
- College of Pharmacy Mokpo National University Jeonnam Korea
| | - Dong Jae Baek
- College of Pharmacy Mokpo National University Jeonnam Korea
| |
Collapse
|
7
|
Targeting sphingosine kinase 1/2 by a novel dual inhibitor SKI-349 suppresses non-small cell lung cancer cell growth. Cell Death Dis 2022; 13:602. [PMID: 35831279 PMCID: PMC9279331 DOI: 10.1038/s41419-022-05049-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/16/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Sphingosine kinase 1 (SphK1) and sphingosine kinase (SphK2) are both important therapeutic targets of non-small cell lung cancer (NSCLC). SKI-349 is a novel, highly efficient and small molecular SphK1/2 dual inhibitor. Here in primary human NSCLC cells and immortalized cell lines, SKI-349 potently inhibited cell proliferation, cell cycle progression, migration and viability. The dual inhibitor induced mitochondrial depolarization and apoptosis activation in NSCLC cells, but it was non-cytotoxic to human lung epithelial cells. SKI-349 inhibited SphK activity and induced ceramide accumulation in primary NSCLC cells, without affecting SphK1/2 expression. SKI-349-induced NSCLC cell death was attenuated by sphingosine-1-phosphate and by the SphK activator K6PC-5, but was potentiated by the short-chain ceramide C6. Moreover, SKI-349 induced Akt-mTOR inactivation, JNK activation, and oxidative injury in primary NSCLC cells. In addition, SKI-349 decreased bromodomain-containing protein 4 (BRD4) expression and downregulated BRD4-dependent genes (Myc, cyclin D1 and Klf4) in primary NSCLC cells. At last, SKI-349 (10 mg/kg) administration inhibited NSCLC xenograft growth in nude mice. Akt-mTOR inhibition, JNK activation, oxidative injury and BRD4 downregulation were detected in SKI-349-treated NSCLC xenograft tissues. Taken together, targeting SphK1/2 by SKI-349 potently inhibits NSCLC cell growth in vitro and in vivo.
Collapse
|
8
|
Kim SB, Kim KJ, Shrestha J, Oh YS, Lee JY, Lee S, Park EY, Baek DJ. Synthesis and Cytotoxic Activity of Fingolimod (FTY720) Analogs with Various Amide Head Groups. Pharm Chem J 2022. [DOI: 10.1007/s11094-022-02659-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
9
|
Pashikanti S, Foster DJ, Kharel Y, Brown AM, Bevan DR, Lynch KR, Santos WL. Sphingosine Kinase 2 Inhibitors: Rigid Aliphatic Tail Derivatives Deliver Potent and Selective Analogues. ACS BIO & MED CHEM AU 2022; 2:469-489. [PMID: 36281302 PMCID: PMC9585524 DOI: 10.1021/acsbiomedchemau.2c00017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Sphingosine 1-phosphate
(S1P) is a pleiotropic signaling molecule
that interacts with five native G-protein coupled receptors (S1P1–5)
to regulate cell growth, survival, and proliferation. S1P has been
implicated in a variety of pathologies including cancer, kidney fibrosis,
and multiple sclerosis. As key mediators in the synthesis of S1P,
sphingosine kinase (SphK) isoforms 1 and 2 have attracted attention
as viable targets for pharmacologic intervention. In this report,
we describe the design, synthesis, and biological evaluation of sphingosine
kinase 2 (SphK2) inhibitors with a focus on systematically introducing
rigid structures in the aliphatic lipid tail present in existing SphK2
inhibitors. Experimental as well as molecular modeling studies suggest
that conformationally restricted “lipophilic tail” analogues
bearing a bulky terminal moiety or an internal phenyl ring are useful
to complement the “J”-shaped sphingosine binding pocket
of SphK2. We identified 14c (SLP9101555) as a potent
SphK2 inhibitor (Ki = 90 nM) with 200-fold
selectivity over SphK1. Molecular docking studies indicated key interactions:
the cyclohexyl ring binding in the cleft deep in the pocket, a trifluoromethyl
group fitting in a small side cavity, and a hydrogen bond between
the guanidino group and Asp308 (amino acid numbering refers to human
SphK2 (isoform c) orthologue). In vitro studies using
U937 human histiocytic lymphoma cells showed marked decreases in extracellular
S1P levels in response to our SphK2 inhibitors. Administration of 14c (dose: 5 mg/kg) to mice resulted in a sustained increase
of circulating S1P levels, suggesting target engagement.
Collapse
Affiliation(s)
- Srinath Pashikanti
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24060, United States
- Department of Biomedical and Pharmaceutical Sciences, Idaho State University, Pocatello, Idaho 83209, United States
| | - Daniel J. Foster
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24060, United States
| | - Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Anne M. Brown
- Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24060, United States
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia 24060, United States
| | - David R. Bevan
- Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24060, United States
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia 24060, United States
| | - Kevin R. Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Webster L. Santos
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24060, United States
- Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24060, United States
| |
Collapse
|
10
|
Determining the Anticancer Activity of Sphingosine Kinase Inhibitors Containing Heteroatoms in Their Tail Structure. Pharmaceutics 2022; 14:pharmaceutics14010157. [PMID: 35057052 PMCID: PMC8779255 DOI: 10.3390/pharmaceutics14010157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 11/17/2022] Open
Abstract
Sphingosine kinase (SK) enzyme, a central player of sphingolipid rheostat, catalyzes the phosphorylation of sphingosine to the bioactive lipid mediator sphingosine 1 phosphate (S1P), which regulates cancer cell proliferation, migration, differentiation, and angiogenesis through its extracellular five G protein-coupled S1P receptors (S1PR1–5). Recently, several research studies on SK inhibitors have taken place in order use them for the development of novel anticancer-targeted therapy. In this study, we designed and synthesized analog derivatives of known SK1 inhibitors, namely RB005 and PF-543, by introducing heteroatoms at their tail structure, as well as investigated their anticancer activities and pharmacokinetic parameters in vitro. Compounds 1–20 of RB005 and PF-543 derivatives containing an aliphatic chain or a tail structure of benzenesulfonyl were synthesized. All compounds of set 1 (1–10) effectively reduced cell viability in both HT29 and HCT116 cells, whereas set 2 derivatives (11–20) showed poor anticancer effect. Compound 10, having the highest cytotoxic effect (48 h, HT29 IC50 = 6.223 µM, HCT116 IC50 = 8.694 µM), induced HT29 and HCT116 cell death in a concentration-dependent manner through the mitochondrial apoptotic pathway, which was demonstrated by increased annexin V-FITC level, and increased apoptotic marker cleaved caspase-3 and cleaved PARP. Compound 10 inhibited SK1 by 20%, and, thus, the S1P level decreased by 42%. Unlike the apoptosis efficacy, the SK1 inhibitory effect and selectivity of the PF-543 derivative were superior to that of the RB005 analog. As a result, compounds with an aliphatic chain tail exhibited stronger apoptotic effects. However, this ability was not proportional to the degree of SK inhibition. Compound 10 increased the protein phosphatase 2A (PP2A) activity (1.73 fold) similar to FTY720 (1.65 fold) and RB005 (1.59 fold), whereas compounds 11 and 13 had no effect on PP2A activation. Since the PP2A activity increased in compounds with an aliphatic chain tail, it can be suggested that PP2A activation has an important effect on anticancer and SK inhibitory activities.
Collapse
|
11
|
Bu Y, Wu H, Deng R, Wang Y. Therapeutic Potential of SphK1 Inhibitors Based on Abnormal Expression of SphK1 in Inflammatory Immune Related-Diseases. Front Pharmacol 2021; 12:733387. [PMID: 34737701 PMCID: PMC8560647 DOI: 10.3389/fphar.2021.733387] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/04/2021] [Indexed: 01/12/2023] Open
Abstract
Sphingosine kinase 1(SphK1) a key enzyme that catalyzes the conversion of sphingosine (Sph) to sphingosine 1-phosphate (S1P), so as to maintain the dynamic balance of sphingolipid-rheostat in cells and participate in cell growth and death, proliferation and migration, vasoconstriction and remodeling, inflammation and metabolism. The normal expression of SphK1 maintains the balance of physiological and pathological states, which is reflected in the regulation of inflammatory factor secretion, immune response in traditional immune cells and non-traditional immune cells, and complex signal transduction. However, abnormal SphK1 expression and activity are found in various inflammatory and immune related-diseases, such as hypertension, atherosclerosis, Alzheimer’s disease, inflammatory bowel disease and rheumatoid arthritis. In view of the therapeutic potential of regulating SphK1 and its signal, the current research is aimed at SphK1 inhibitors, such as SphK1 selective inhibitors and dual SphK1/2 inhibitor, and other compounds with inhibitory potency. This review explores the regulatory role of over-expressed SphK1 in inflammatory and immune related-diseases, and investigate the latest progress of SphK1 inhibitors and the improvement of disease or pathological state.
Collapse
Affiliation(s)
- Yanhong Bu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Hong Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Ran Deng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Yan Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| |
Collapse
|
12
|
Gupta P, Taiyab A, Hussain A, Alajmi MF, Islam A, Hassan MI. Targeting the Sphingosine Kinase/Sphingosine-1-Phosphate Signaling Axis in Drug Discovery for Cancer Therapy. Cancers (Basel) 2021; 13:1898. [PMID: 33920887 PMCID: PMC8071327 DOI: 10.3390/cancers13081898] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/11/2021] [Accepted: 04/03/2021] [Indexed: 02/07/2023] Open
Abstract
Sphingolipid metabolites have emerged as critical players in the regulation of various physiological processes. Ceramide and sphingosine induce cell growth arrest and apoptosis, whereas sphingosine-1-phosphate (S1P) promotes cell proliferation and survival. Here, we present an overview of sphingolipid metabolism and the compartmentalization of various sphingolipid metabolites. In addition, the sphingolipid rheostat, a fine metabolic balance between ceramide and S1P, is discussed. Sphingosine kinase (SphK) catalyzes the synthesis of S1P from sphingosine and modulates several cellular processes and is found to be essentially involved in various pathophysiological conditions. The regulation and biological functions of SphK isoforms are discussed. The functions of S1P, along with its receptors, are further highlighted. The up-regulation of SphK is observed in various cancer types and is also linked to radio- and chemoresistance and poor prognosis in cancer patients. Implications of the SphK/S1P signaling axis in human pathologies and its inhibition are discussed in detail. Overall, this review highlights current findings on the SphK/S1P signaling axis from multiple angles, including their functional role, mechanism of activation, involvement in various human malignancies, and inhibitor molecules that may be used in cancer therapy.
Collapse
Affiliation(s)
- Preeti Gupta
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| | - Aaliya Taiyab
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.H.); (M.F.A.)
| | - Mohamed F. Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.H.); (M.F.A.)
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| |
Collapse
|
13
|
Erythrocyte sphingosine kinase regulates intraerythrocytic development of Plasmodium falciparum. Sci Rep 2021; 11:1257. [PMID: 33441957 PMCID: PMC7806667 DOI: 10.1038/s41598-020-80658-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/26/2020] [Indexed: 11/18/2022] Open
Abstract
The sphingolipid pool is key regulator of vital cellular functions in Plasmodium falciparum a causative agent for deadly malaria. Erythrocytes, the host for asexual stage of Plasmodium, are major reservoir for Sphingosine-1-phosphate (S1P). Erythrocyte possesses Sphingosine kinase (SphK) that catalyzed its biosynthesis from sphingosine (Sph). Since, Plasmodium lacks SphK homologous protein it can be envisaged that it co-opts sphingolipids from both intraerythrocytic as well as extracellular pools for its growth and development. Herein, by sphingosine-NBD probing, we report that infected erythrocytes imports Sph from extracellular pool, which is converted to S1P and thereby taken by P. falciparum. Next, by targeting of the SphK through specific inhibitor N,N-Dimethylsphingosine DMS, we show a reduction in erythrocyte endogenous S1P pool and SphK-phosphorylation that led to inhibition in growth and development of ring stage P. falciparum. Owing to the role of S1P in erythrocyte glycolysis we analyzed uptake of NBD-Glucose and production of lactate in DMS treated and untreated plasmodium. DMS treatment led to decreased glycolysis in Plasmodium. Interestingly the host free Plasmodium did not show any effect on glycolysis with DMS treatment indicating its host-mediated effect. Further to understand the in-vivo anti-plasmodial effects of exogenous and endogenous erythrocyte S1P level, Sphingosine-1-phosphate lyase (S1PL) inhibitor (THI), S1P and SphK-1 inhibitor (DMS), were used in Plasmodium berghei ANKA (PbA) mice model. DMS treatment led to reduction of endogenous S1P conferred significant decrease in parasite load, whereas the plasma level S1P modulated by (THI) and exogenous S1P have no effect on growth of Plasmodium. This suggested erythrocyte endogenous S1P pool is important for Plasmodium growth whereas the plasma level S1P has no effect. Altogether, this study provides insight on cellular processes regulated by S1P in P. falciparum and highlights the novel mechanistically distinct molecular target i.e. SphK-1.
Collapse
|
14
|
Ren R, Pang B, Han Y, Li Y. A Glimpse of the Structural Biology of the Metabolism of Sphingosine-1-Phosphate. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2021; 4:2515256421995601. [PMID: 37366379 PMCID: PMC10243590 DOI: 10.1177/2515256421995601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 06/28/2023]
Abstract
As a key sphingolipid metabolite, sphingosine-1-phosphate (S1P) plays crucial roles in vascular and immune systems. It regulates angiogenesis, vascular integrity and homeostasis, allergic responses, and lymphocyte trafficking. S1P is interconverted with sphingosine, which is also derived from the deacylation of ceramide. S1P levels and the ratio to ceramide in cells are tightly regulated by its metabolic pathways. Abnormal S1P production causes the occurrence and progression of numerous severe diseases, such as metabolic syndrome, cancers, autoimmune disorders such as multiple sclerosis, and kidney and cardiovascular diseases. In recent years, huge advances on the structure of S1P metabolic pathways have been accomplished. In this review, we have got a glimpse of S1P metabolism through structural and biochemical studies of: sphingosine kinases, S1P transporters and S1P receptors, and the development of therapeutics targeting S1P signaling. The progress we summarize here could provide fresh perspectives to further the exploration of S1P functions and facilitate the development of therapeutic molecules targeting S1P signaling with improved specificity and therapeutic effects.
Collapse
Affiliation(s)
- Ruobing Ren
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| | - Bin Pang
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| | - Yufei Han
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| | - Yihao Li
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| |
Collapse
|
15
|
McGowan EM, Haddadi N, Nassif NT, Lin Y. Targeting the SphK-S1P-SIPR Pathway as a Potential Therapeutic Approach for COVID-19. Int J Mol Sci 2020; 21:ijms21197189. [PMID: 33003377 PMCID: PMC7583882 DOI: 10.3390/ijms21197189] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
The world is currently experiencing the worst health pandemic since the Spanish flu in 1918-the COVID-19 pandemic-caused by the coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This pandemic is the world's third wake-up call this century. In 2003 and 2012, the world experienced two major coronavirus outbreaks, SARS-CoV-1 and Middle East Respiratory syndrome coronavirus (MERS-CoV), causing major respiratory tract infections. At present, there is neither a vaccine nor a cure for COVID-19. The severe COVID-19 symptoms of hyperinflammation, catastrophic damage to the vascular endothelium, thrombotic complications, septic shock, brain damage, acute disseminated encephalomyelitis (ADEM), and acute neurological and psychiatric complications are unprecedented. Many COVID-19 deaths result from the aftermath of hyperinflammatory complications, also referred to as the "cytokine storm syndrome", endotheliitus and blood clotting, all with the potential to cause multiorgan dysfunction. The sphingolipid rheostat plays integral roles in viral replication, activation/modulation of the immune response, and importantly in maintaining vasculature integrity, with sphingosine 1 phosphate (S1P) and its cognate receptors (SIPRs: G-protein-coupled receptors) being key factors in vascular protection against endotheliitus. Hence, modulation of sphingosine kinase (SphK), S1P, and the S1P receptor pathway may provide significant beneficial effects towards counteracting the life-threatening, acute, and chronic complications associated with SARS-CoV-2 infection. This review provides a comprehensive overview of SARS-CoV-2 infection and disease, prospective vaccines, and current treatments. We then discuss the evidence supporting the targeting of SphK/S1P and S1P receptors in the repertoire of COVID-19 therapies to control viral replication and alleviate the known and emerging acute and chronic symptoms of COVID-19. Three clinical trials using FDA-approved sphingolipid-based drugs being repurposed and evaluated to help in alleviating COVID-19 symptoms are discussed.
Collapse
Affiliation(s)
- Eileen M McGowan
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precise Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China;
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
- Correspondence: ; Tel.: +61-405814048
| | - Nahal Haddadi
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
| | - Najah T. Nassif
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
| | - Yiguang Lin
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precise Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China;
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
| |
Collapse
|
16
|
Roy S, Mahapatra AD, Mohammad T, Gupta P, Alajmi MF, Hussain A, Rehman MT, Datta B, Hassan MI. Design and Development of Novel Urea, Sulfonyltriurea, and Sulfonamide Derivatives as Potential Inhibitors of Sphingosine Kinase 1. Pharmaceuticals (Basel) 2020; 13:E118. [PMID: 32526899 PMCID: PMC7346089 DOI: 10.3390/ph13060118] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/17/2022] Open
Abstract
Sphingosine kinase 1 (SphK1) is one of the well-studied drug targets for cancer and inflammatory diseases. Recently discovered small-molecule inhibitors of SphK1 have been recommended in cancer therapeutics; however, selectivity and potency of first-generation inhibitors are great challenge. In search of effective SphK1 inhibitors, a set of small molecules have been designed and synthesized bearing urea, sulfonylurea, sulfonamide, and sulfonyltriurea groups. The binding affinity of these inhibitors was measured by fluorescence-binding assay and isothermal titration calorimetry. Compounds 1, 5, 6, and 7 showed an admirable binding affinity to the SphK1 in the sub-micromolar range and significantly inhibited SphK1 activity with admirable IC50 values. Molecular docking studies revealed that these compounds fit well into the sphingosine binding pocket of SphK1 and formed significant number of hydrogen bonds and van der Waals interactions. These molecules may be exploited as potent and selective inhibitors of SphK1 that could be implicated in cancer therapeutics after the required in vivo validation.
Collapse
Affiliation(s)
- Sonam Roy
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (S.R.); (T.M.); (P.G.)
| | - Amarjyoti Das Mahapatra
- Department of Chemistry, Indian Institute of Technology, Palaj, Gandhinagar, Gujarat 382355, India;
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (S.R.); (T.M.); (P.G.)
| | - Preeti Gupta
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (S.R.); (T.M.); (P.G.)
| | - Mohamed F. Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.); (M.T.R.)
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.); (M.T.R.)
| | - Md. Tabish Rehman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.); (M.T.R.)
| | - Bhaskar Datta
- Department of Chemistry, Indian Institute of Technology, Palaj, Gandhinagar, Gujarat 382355, India;
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (S.R.); (T.M.); (P.G.)
| |
Collapse
|
17
|
Liu X, Simon JM, Xie H, Hu L, Wang J, Zurlo G, Fan C, Ptacek TS, Herring L, Tan X, Li M, Baldwin AS, Kim WY, Wu T, Kirschner MW, Gong K, Zhang Q. Genome-wide Screening Identifies SFMBT1 as an Oncogenic Driver in Cancer with VHL Loss. Mol Cell 2020; 77:1294-1306.e5. [PMID: 32023483 DOI: 10.1016/j.molcel.2020.01.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/09/2019] [Accepted: 01/01/2020] [Indexed: 11/15/2022]
Abstract
von Hippel-Lindau (VHL) is a critical tumor suppressor in clear cell renal cell carcinomas (ccRCCs). It is important to identify additional therapeutic targets in ccRCC downstream of VHL loss besides hypoxia-inducible factor 2α (HIF2α). By performing a genome-wide screen, we identified Scm-like with four malignant brain tumor domains 1 (SFMBT1) as a candidate pVHL target. SFMBT1 was considered to be a transcriptional repressor but its role in cancer remains unclear. ccRCC patients with VHL loss-of-function mutations displayed elevated SFMBT1 protein levels. SFMBT1 hydroxylation on Proline residue 651 by EglN1 mediated its ubiquitination and degradation governed by pVHL. Depletion of SFMBT1 abolished ccRCC cell proliferation in vitro and inhibited orthotopic tumor growth in vivo. Integrated analyses of ChIP-seq, RNA-seq, and patient prognosis identified sphingosine kinase 1 (SPHK1) as a key SFMBT1 target gene contributing to its oncogenic phenotype. Therefore, the pVHL-SFMBT1-SPHK1 axis serves as a potential therapeutic avenue for ccRCC.
Collapse
Affiliation(s)
- Xijuan Liu
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jeremy M Simon
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Haibiao Xie
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Lianxin Hu
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jun Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Giada Zurlo
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Travis S Ptacek
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Laura Herring
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA; UNC Proteomics Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xianming Tan
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Mingjie Li
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - William Y Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Tao Wu
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| | - Marc W Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Qing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
18
|
Abstract
There is substantial evidence that the enzymes, sphingosine kinase 1 and 2, which catalyse the formation of the bioactive lipid sphingosine 1-phosphate, are involved in pathophysiological processes. In this chapter, we appraise the evidence that both enzymes are druggable and describe how isoform-specific inhibitors can be developed based on the plasticity of the sphingosine-binding site. This is contextualised with the effect of sphingosine kinase inhibitors in cancer, pulmonary hypertension, neurodegeneration, inflammation and sickling.
Collapse
Affiliation(s)
- Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde , Glasgow, Scotland, UK
| | - David R Adams
- Institute of Chemical Sciences, Heriot-Watt University, Edinburgh, Scotland, UK
| | - Nigel J Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde , Glasgow, Scotland, UK.
| |
Collapse
|
19
|
Targeting sphingosine kinase 1 for the treatment of pulmonary arterial hypertension. Future Med Chem 2019; 11:2939-2953. [DOI: 10.4155/fmc-2019-0130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH), characterized by high morbidity and mortality, is a serious hazard to human life. Until now, the long-term survival of the PAH patients is still suboptimal. Recently, sphingosine kinase 1 (SPHK1) has drawn more and more attention due to its essential role in the pulmonary vasoconstriction, remodeling of pulmonary blood vessels and right cardiac lesions in PAH patients, and this enzyme is regarded as a new target for the treatment of PAH. Here, we discussed the multifarious functions of SPHK1 in PAH physiology and pathogenesis. Moreover, the structural features of SPHK1 and binding modes with different inhibitors were summarized. Finally, recent advances in the medicinal chemistry research of SPHK1 inhibitors are presented.
Collapse
|
20
|
Kim SW, Lee T, Oh YS, Shin SM, Lee JY, Kim S, Baek DJ, Park EY. Synthesis and Biological Evaluation of PF-543 Derivative Containing Aliphatic Side Chain. Chem Pharm Bull (Tokyo) 2019; 67:599-603. [PMID: 31155566 DOI: 10.1248/cpb.c18-00724] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The PF-543 is known as a potent and selective inhibitor of sphingosine kinase (SK) 1 amongst all the SK inhibitors known to date. In a recently reported study by Pfizer on the synthesis of PF-543 derivatives and the SK inhibitory effects, the introduction of propyl moiety into sulfonyl group of PF-543 in the case of 26b revealed an excellent result of 1.7 nM of IC50 of SK1, suggesting the potential substitution of chain structure for benzenesulfonyl structure. In the present work, we aimed for identification of antitumor activity and inhibitory effects of PF-543 derivative containing aliphatic long chain (similar to known SK inhibitors) on SK1. The synthesized compound 2 exhibited an inhibitory effect on SK1 in a manner similar to that of PF-543; the PF-543 derivative manifested similar antitumor activity on HT29, HCT116 (colorectal cancer cell line), and AGS (gastric cancer cell line) cells. Also, from the docking study conducted with PF-543 and compound 2, it was apparent that the aliphatic chain in compound 2 could probably replace benzenesulfonyl structure of PF-543.
Collapse
Affiliation(s)
- Seon Woong Kim
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University
| | - Taeho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University
| | | | - Joo-Youn Lee
- College of Pharmacy, Seoul National University.,Korea Chemical Bank, Korea Research Institute of Chemical Technology
| | - Sanghee Kim
- College of Pharmacy, Seoul National University
| | - Dong Jae Baek
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University
| | - Eun-Young Park
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University
| |
Collapse
|
21
|
Kim SW, Lee T, Lee JY, Kim S, Jun HS, Park EY, Baek DJ. Synthesis and Biological Evaluation of PF-543 Derivative. LETT ORG CHEM 2018. [DOI: 10.2174/1570178615666181009121430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
PF-543 has been known as a substance that strongly inhibits SK1. However, it also exhibits antineoplastic activity that is lower than other inhibitors of SK1. In this study, we compared PF-543 and synthesized a newly designed derivative of PF-543 (compound 2) in which two aromatic structures were connected in para-form. The synthesized derivative showed inhibitory effect on SK1, similar to that of PF-543. However, it was more cytotoxic to HT29, AGS, and PC3 cells than PF-543. We also carried out a docking study for SK1 and demonstrated that the synthesized derivative showed interaction with SK1 similar to PF-543. Results obtained from this study suggest that the structure of compound 2 may be well substituted for the structure of PF-543 in terms of biological activity, providing us important structural information for the design of new derivatives of PF-543.
Collapse
Affiliation(s)
- Seon Woong Kim
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, South Korea
| | - Taeho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, South Korea
| | - Joo-Youn Lee
- College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Sanghee Kim
- College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Hee-Sook Jun
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea
| | - Eun-Young Park
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, South Korea
| | - Dong Jae Baek
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, South Korea
| |
Collapse
|
22
|
Shrestha J, Ki SH, Shin SM, Kim SW, Lee JY, Jun HS, Lee T, Kim S, Baek DJ, Park EY. Synthesis of Novel FTY720 Analogs with Anticancer Activity through PP2A Activation. Molecules 2018; 23:molecules23112750. [PMID: 30355990 PMCID: PMC6278267 DOI: 10.3390/molecules23112750] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 12/16/2022] Open
Abstract
FTY720 inhibits various cancers through PP2A activation. The structure of FTY720 is also used as a basic structure for the design of sphingosine kinase (SK) inhibitors. We have synthesized derivatives using an amide chain in FTY720 with a phenyl backbone, and then compounds were screened by an MTT cell viability assay. The PP2A activity of compound 7 was examined. The phosphorylation levels of AKT and ERK, downstream targets of PP2A, in the presence of compound 7, were determined. Compound 7 may exhibit anticancer effects through PP2A activation rather than the mechanism by inhibition of SK1 in cancer cells. In the docking study of compound 7 and PP2A, the amide chain of compound 7 showed an interaction with Asn61 that was different from FTY720, which is expected to affect the activity of the compound.
Collapse
Affiliation(s)
- Jitendra Shrestha
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Korea.
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, Gwangju, 61452, Korea.
| | - Sang Mi Shin
- College of Pharmacy, Chosun University, Gwangju, 61452, Korea.
| | - Seon Woong Kim
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Korea.
| | - Joo-Youn Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
- Korea Chemical Bank, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea.
| | - Hee-Sook Jun
- Lee Gil Ya Cancer and Diabetes Institute, Department of Molecular Medicine, Gachon University, Incheon 21999, Korea.
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Korea.
| | - Taeho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Sanghee Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Dong Jae Baek
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Korea.
| | - Eun-Young Park
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Korea.
| |
Collapse
|
23
|
Dellafiora L, Galaverna G, Dall’Asta C. Mechanisms of Fumonisin B1 Toxicity: A Computational Perspective beyond the Ceramide Synthases Inhibition. Chem Res Toxicol 2018; 31:1203-1212. [DOI: 10.1021/acs.chemrestox.8b00188] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Luca Dellafiora
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Gianni Galaverna
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Chiara Dall’Asta
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| |
Collapse
|
24
|
Zheng X, Li W, Ren L, Liu J, Pang X, Chen X, Kang D, Wang J, Du G. The sphingosine kinase-1/sphingosine-1-phosphate axis in cancer: Potential target for anticancer therapy. Pharmacol Ther 2018; 195:85-99. [PMID: 30347210 DOI: 10.1016/j.pharmthera.2018.10.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sphingolipid metabolites, such as ceramide, sphingosine and sphingosine-1-phosphate (S1P), play many important roles in cellular activities. Ceramide and sphingosine inhibit cell proliferation and induce cell apoptosis while S1P has the opposite effect. Maintaining a metabolic balance of sphingolipids is essential for growth and development of cells. Sphingosine kinase (SPHK) is an important regulator for keeping this balance. It controls the level of S1P and plays important roles in proliferation, migration, and invasion of cancer cells and tumor angiogenesis. There are two isoenzymes of sphingosine kinase, SPHK1 and SPHK2. SPHK1 is ubiquitously expressed in most cancers where it promotes survival and proliferation, while SPHK2 is restricted to only certain tissues and its functions are not well characterized. SPHK1 is currently considered as a novel target for the treatment of cancers. Targeting SPHK1 would provide new strategies for cancer treatment and improve the prognosis of cancer patients. Here we review and summarize the current research findings on the SPHK1-S1P axis in cancer from many aspects including structure, expression, regulation, mechanism, and potential inhibitors.
Collapse
Affiliation(s)
- Xiangjin Zheng
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Wan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Liwen Ren
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Jinyi Liu
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiaocong Pang
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - De Kang
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
25
|
Pulkoski-Gross MJ, Obeid LM. Molecular mechanisms of regulation of sphingosine kinase 1. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1413-1422. [PMID: 30591148 DOI: 10.1016/j.bbalip.2018.08.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 01/08/2023]
Abstract
Within the last 3 decades, there has been intense study of bioactive sphingolipids and the enzymes which metabolize those lipids. One enzyme is the critical lipid kinase sphingosine kinase 1 (SK1), which produces the potent and pleiotropic signaling lipid, sphingosine 1-phosphate (S1P). SK1 and S1P have been implicated in a host of different diseases including cancer, chronic inflammation, and metabolic diseases. However, while there is ample knowledge about the importance of these molecules in the development and progression of disease there is a dearth of knowledge of the molecular mechanisms which regulate SK1 function. In this review, we will cover some of the more recent and exciting findings about the different ways SK1 function can be regulated, from transcriptional regulation to protein stability. Finally, we will delve into recent structural insights into SK1 and how they might relate to function at cell membranes.
Collapse
Affiliation(s)
- Michael J Pulkoski-Gross
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11790, USA; Department of Medicine, The Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11790, USA.
| | - Lina M Obeid
- Department of Medicine, The Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11790, USA; Northport Veterans Affairs Medical Center, Northport, NY 11768, USA.
| |
Collapse
|
26
|
Park EY, Lee T, Oh YS, Lee JY, Shrestha J, Hong SW, Jin YJ, Jo G, Kim S, Hwang GT, Han DS, Baek DJ. Synthesis of dansyl labeled sphingosine kinase 1 inhibitor. Chem Phys Lipids 2018; 215:29-33. [PMID: 30044952 DOI: 10.1016/j.chemphyslip.2018.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 07/10/2018] [Accepted: 07/21/2018] [Indexed: 02/04/2023]
Abstract
PF-543 is a non-sphingosine analogue with inhibitory effect against SK1, based on a Ki of 4.3 nM and 130-fold selectivity for SK1 over SK2. Since the development of PF-543, animal studies demonstrated its valuable role in multiple sclerosis, myocardial infarction, and colorectal cancer. We synthesized labeled PF-543 for biochemical studies involving SK1. Overall, the 8-step synthetic route used 3,5-dimethylphenol as the starting material. A docking study of SK1 and SK1 inhibitory activity confirmed the structural similarity between the synthetic dansyl-PF-543 and PF-543. We also provide fluorescence spectra of dansyl-PF-543.
Collapse
Affiliation(s)
- Eun-Young Park
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, South Korea
| | - Taeho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, South Korea
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, Seongnam 13135, South Korea
| | - Joo-Youn Lee
- College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Korea Chemical Bank, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea
| | - Jitendra Shrestha
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, South Korea
| | - Seung Woo Hong
- Department of Chemistry, Kyungpook National University, Daegu 41566, South Korea
| | - Yun Ji Jin
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, South Korea
| | - GeunHyung Jo
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, South Korea
| | - Sanghee Kim
- College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Gil Tae Hwang
- Department of Chemistry, Kyungpook National University, Daegu 41566, South Korea
| | - Dong-Sul Han
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, South Korea.
| | - Dong Jae Baek
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, South Korea.
| |
Collapse
|
27
|
|
28
|
McNaughton M, Pitman M, Pitson SM, Pyne NJ, Pyne S. Proteasomal degradation of sphingosine kinase 1 and inhibition of dihydroceramide desaturase by the sphingosine kinase inhibitors, SKi or ABC294640, induces growth arrest in androgen-independent LNCaP-AI prostate cancer cells. Oncotarget 2017; 7:16663-75. [PMID: 26934645 PMCID: PMC4941342 DOI: 10.18632/oncotarget.7693] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 02/11/2016] [Indexed: 11/25/2022] Open
Abstract
Sphingosine kinases (two isoforms termed SK1 and SK2) catalyse the formation of the bioactive lipid sphingosine 1-phosphate. We demonstrate here that the SK2 inhibitor, ABC294640 (3-(4-chlorophenyl)-adamantane-1-carboxylic acid (pyridin-4-ylmethyl)amide) or the SK1/SK2 inhibitor, SKi (2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole)) induce the proteasomal degradation of SK1a (Mr = 42 kDa) and inhibit DNA synthesis in androgen-independent LNCaP-AI prostate cancer cells. These effects are recapitulated by the dihydroceramide desaturase (Des1) inhibitor, fenretinide. Moreover, SKi or ABC294640 reduce Des1 activity in Jurkat cells and ABC294640 induces the proteasomal degradation of Des1 (Mr = 38 kDa) in LNCaP-AI prostate cancer cells. Furthermore, SKi or ABC294640 or fenretinide increase the expression of the senescence markers, p53 and p21 in LNCaP-AI prostate cancer cells. The siRNA knockdown of SK1 or SK2 failed to increase p53 and p21 expression, but the former did reduce DNA synthesis in LNCaP-AI prostate cancer cells. Moreover, N-acetylcysteine (reactive oxygen species scavenger) blocked the SK inhibitor-induced increase in p21 and p53 expression but had no effect on the proteasomal degradation of SK1a. In addition, siRNA knockdown of Des1 increased p53 expression while a combination of Des1/SK1 siRNA increased the expression of p21. Therefore, Des1 and SK1 participate in regulating LNCaP-AI prostate cancer cell growth and this involves p53/p21-dependent and -independent pathways. Therefore, we propose targeting androgen-independent prostate cancer cells with compounds that affect Des1/SK1 to modulate both de novo and sphingolipid rheostat pathways in order to induce growth arrest.
Collapse
Affiliation(s)
- Melissa McNaughton
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, UK
| | - Melissa Pitman
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide SA 5000, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide SA 5000, Australia
| | - Nigel J Pyne
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, UK
| | - Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, UK
| |
Collapse
|
29
|
Sphingosine Kinase 1: A Potential Therapeutic Target in Pulmonary Arterial Hypertension? Trends Mol Med 2017; 23:786-798. [DOI: 10.1016/j.molmed.2017.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/04/2017] [Accepted: 07/10/2017] [Indexed: 12/22/2022]
|
30
|
Vettorazzi M, Angelina E, Lima S, Gonec T, Otevrel J, Marvanova P, Padrtova T, Mokry P, Bobal P, Acosta LM, Palma A, Cobo J, Bobalova J, Csollei J, Malik I, Alvarez S, Spiegel S, Jampilek J, Enriz RD. An integrative study to identify novel scaffolds for sphingosine kinase 1 inhibitors. Eur J Med Chem 2017; 139:461-481. [PMID: 28822281 DOI: 10.1016/j.ejmech.2017.08.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/02/2017] [Accepted: 08/05/2017] [Indexed: 10/19/2022]
Abstract
Sphingosine kinase 1 (SphK1), the enzyme that produces the bioactive sphingolipid metabolite, sphingosine-1-phosphate, is a promising new molecular target for therapeutic intervention in cancer and inflammatory diseases. In view of its importance, the main objective of this work was to find new and more potent inhibitors for this enzyme possessing different structural scaffolds than those of the known inhibitors. Our theoretical and experimental study has allowed us to identify two new structural scaffolds (three new compounds), which could be used as starting structures for the design and then the development of new inhibitors of SphK1. Our study was carried out in different steps: virtual screening, synthesis, bioassays and molecular modelling. From our results, we propose a new dihydrobenzo[b]pyrimido[5,4-f]azepine and two alkyl{3-/4-[1-hydroxy-2-(4-arylpiperazin-1-yl)ethyl]phenyl}carbamates as initial structures for the development of new inhibitors. In addition, our molecular modelling study using QTAIM calculations, allowed us to describe in detail the molecular interactions that stabilize the different Ligand-Receptor complexes. Such analyses indicate that the cationic head of the different compounds must be refined in order to obtain an increase in the binding affinity of these ligands.
Collapse
Affiliation(s)
- Marcela Vettorazzi
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina
| | - Emilio Angelina
- Laboratorio de Estructura Molecular y Propiedades, Área de Química Física, Departamento de Química, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Avda. Libertad 5460, 3400 Corrientes, Argentina
| | - Santiago Lima
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Tomas Gonec
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Jan Otevrel
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Pavlina Marvanova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Tereza Padrtova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Petr Mokry
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Pavel Bobal
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Lina M Acosta
- Laboratorio de Síntesis Orgánica, Escuela de Química, Universidad Industrial de Santander, Carrera 27, Calle 9, A.A 678, Bucaramanga, Colombia
| | - Alirio Palma
- Laboratorio de Síntesis Orgánica, Escuela de Química, Universidad Industrial de Santander, Carrera 27, Calle 9, A.A 678, Bucaramanga, Colombia
| | - Justo Cobo
- Inorganic and Organic Department, University of Jaén, Campus Las Lagunillas s/n, 23071, Jaén, Spain
| | - Janette Bobalova
- Institute of Analytical Chemistry of the Czech Academy of Sciences, v. v. i., Veveri 97, 602 00 Brno, Czech Republic
| | - Jozef Csollei
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Comenius University, Odbojarov 10, 83232 Bratislava, Slovakia
| | - Ivan Malik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Comenius University, Odbojarov 10, 83232 Bratislava, Slovakia
| | - Sergio Alvarez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Josef Jampilek
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Comenius University, Odbojarov 10, 83232 Bratislava, Slovakia
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina.
| |
Collapse
|
31
|
Hatoum D, Haddadi N, Lin Y, Nassif NT, McGowan EM. Mammalian sphingosine kinase (SphK) isoenzymes and isoform expression: challenges for SphK as an oncotarget. Oncotarget 2017; 8:36898-36929. [PMID: 28415564 PMCID: PMC5482707 DOI: 10.18632/oncotarget.16370] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/02/2017] [Indexed: 12/16/2022] Open
Abstract
The various sphingosine kinase (SphK) isoenzymes (isozymes) and isoforms, key players in normal cellular physiology, are strongly implicated in cancer and other diseases. Mutations in SphKs, that may justify abnormal physiological function, have not been recorded. Nonetheless, there is a large and growing body of evidence demonstrating the contribution of gain or loss of function and the imbalance in the SphK/S1P rheostat to a plethora of pathological conditions including cancer, diabetes and inflammatory diseases. SphK is expressed as two isozymes SphK1 and SphK2, transcribed from genes located on different chromosomes and both isozymes catalyze the phosphorylation of sphingosine to S1P. Expression of each SphK isozyme produces alternately spliced isoforms. In recent years the importance of the contribution of SpK1 expression to treatment resistance in cancer has been highlighted and, additionally, differences in treatment outcome appear to also be dependent upon SphK isoform expression. This review focuses on an exciting emerging area of research involving SphKs functions, expression and subcellular localization, highlighting the complexity of targeting SphK in cancer and also comorbid diseases. This review also covers the SphK isoenzymes and isoforms from a historical perspective, from their first discovery in murine species and then in humans, their role(s) in normal cellular function and in disease processes, to advancement of SphK as an oncotarget.
Collapse
Affiliation(s)
- Diana Hatoum
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Nahal Haddadi
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Yiguang Lin
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Najah T. Nassif
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Eileen M. McGowan
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| |
Collapse
|
32
|
Barbour M, McNaughton M, Boomkamp SD, MacRitchie N, Jiang H, Pyne NJ, Pyne S. Effect of sphingosine kinase modulators on interleukin-1β release, sphingosine 1-phosphate receptor 1 expression and experimental autoimmune encephalomyelitis. Br J Pharmacol 2017; 174:210-222. [PMID: 27864936 PMCID: PMC5192795 DOI: 10.1111/bph.13670] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 11/04/2016] [Accepted: 11/13/2016] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE The sphingosine analogue, FTY720 (GilenyaR ), alleviates clinical disease progression in multiple sclerosis. Here, we variously assessed the effects of an azide analogue of (S)-FTY720 vinylphosphonate (compound 5; a sphingosine kinase 1 activator), (R)-FTY720 methyl ether (ROMe, a sphingosine kinase 2 inhibitor) and RB-020 (a sphingosine kinase 1 inhibitor and sphingosine kinase 2 substrate) on IL-1β formation, sphingosine 1-phosphate levels and expression of S1P1 receptors. We also assessed the effect of compound 5 and ROMe in an experimental autoimmune encephalomyelitis (EAE) model in mice. EXPERIMENTAL APPROACH We measured IL-1β formation by macrophages, sphingosine 1-phosphate levels and expression levels of S1P1 receptors in vitro and clinical score in mice with EAE and the extent of inflammatory cell infiltration into the spinal cord in vivo. KEY RESULTS Treatment of differentiated U937 macrophages with compound 5, RB-020 or sphingosine (but not ROMe) enhanced IL-1β release. These data suggest that these compounds might be pro-inflammatory in vitro. However, compound 5 or ROMe reduced disease progression and infiltration of inflammatory cells into the spinal cord in EAE, and ROMe induced a reduction in CD4+ and CD8+ T-cell levels in the blood (lymphopenia). Indeed, ROMe induced a marked decrease in expression of cell surface S1P1 receptors in vitro. CONCLUSION AND IMPLICATIONS This is the first demonstration that an activator of sphingosine kinase 1 (compound 5) and an inhibitor of sphingosine kinase 2 (ROMe, which also reduces cell surface S1P1 receptor expression) have an anti-inflammatory action in EAE.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/chemistry
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Cells, Cultured
- Cricetulus
- Dose-Response Relationship, Drug
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Humans
- Interleukin-1beta/metabolism
- Mice
- Mice, Inbred C57BL
- Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors
- Phosphotransferases (Alcohol Group Acceptor)/metabolism
- Piperidines/chemistry
- Piperidines/pharmacology
- Receptors, Lysosphingolipid/biosynthesis
- Sphingosine/chemistry
- Sphingosine/pharmacology
- Sphingosine-1-Phosphate Receptors
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Mark Barbour
- Strathclyde Institute of Pharmacy and Biomedical ScienceUniversity of StrathclydeGlasgowUK
| | - Melissa McNaughton
- Strathclyde Institute of Pharmacy and Biomedical ScienceUniversity of StrathclydeGlasgowUK
| | - Stephanie D Boomkamp
- Strathclyde Institute of Pharmacy and Biomedical ScienceUniversity of StrathclydeGlasgowUK
| | - Neil MacRitchie
- Strathclyde Institute of Pharmacy and Biomedical ScienceUniversity of StrathclydeGlasgowUK
| | - Hui‐Rong Jiang
- Strathclyde Institute of Pharmacy and Biomedical ScienceUniversity of StrathclydeGlasgowUK
| | - Nigel J Pyne
- Strathclyde Institute of Pharmacy and Biomedical ScienceUniversity of StrathclydeGlasgowUK
| | - Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical ScienceUniversity of StrathclydeGlasgowUK
| |
Collapse
|
33
|
Sphingosine 1-Phosphate Activation of EGFR As a Novel Target for Meningitic Escherichia coli Penetration of the Blood-Brain Barrier. PLoS Pathog 2016; 12:e1005926. [PMID: 27711202 PMCID: PMC5053521 DOI: 10.1371/journal.ppat.1005926] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 09/11/2016] [Indexed: 12/24/2022] Open
Abstract
Central nervous system (CNS) infection continues to be an important cause of mortality and morbidity, necessitating new approaches for investigating its pathogenesis, prevention and therapy. Escherichia coli is the most common Gram-negative bacillary organism causing meningitis, which develops following penetration of the blood-brain barrier (BBB). By chemical library screening, we identified epidermal growth factor receptor (EGFR) as a contributor to E. coli invasion of the BBB in vitro. Here, we obtained the direct evidence that CNS-infecting E. coli exploited sphingosine 1-phosphate (S1P) for EGFR activation in penetration of the BBB in vitro and in vivo. We found that S1P was upstream of EGFR and participated in EGFR activation through S1P receptor as well as through S1P-mediated up-regulation of EGFR-related ligand HB-EGF, and blockade of S1P function through targeting sphingosine kinase and S1P receptor inhibited EGFR activation, and also E. coli invasion of the BBB. We further found that both S1P and EGFR activations occurred in response to the same E. coli proteins (OmpA, FimH, NlpI), and that S1P and EGFR promoted E. coli invasion of the BBB by activating the downstream c-Src. These findings indicate that S1P and EGFR represent the novel host targets for meningitic E. coli penetration of the BBB, and counteracting such targets provide a novel approach for controlling E. coli meningitis in the era of increasing resistance to conventional antibiotics.
Collapse
|
34
|
Lynch KR, Thorpe SB, Santos WL. Sphingosine kinase inhibitors: a review of patent literature (2006-2015). Expert Opin Ther Pat 2016; 26:1409-1416. [PMID: 27539678 DOI: 10.1080/13543776.2016.1226282] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Sphingosine kinase (SphK1 & SphK2) is the sole source of the pleiotropic lipid mediator, sphingosine-1-phosphate (S1P). S1P has been implicated in a variety of diseases such as cancer, Alzheimer's disease, sickle cell disease and fibrosis and thus the biosynthetic route to S1P is a logical target for drug discovery. Areas covered: In this review, the authors consider the SphK inhibitor patent literature from 2006-2016 Q1 with the emphasis on composition of matter utility patents. The Espacenet database was queried with the search term 'sphingosine AND kinase' to identify relevant literature. Expert opinion: Early inhibitor discovery focused on SphK1 with a bias towards oncology indications. Structurally, the reported inhibitors occupy the sphingosine 'J-shaped' binding pocket. The lack of cytotoxicity with improved SphK1 inhibitors raises doubt about the enzyme as an oncology target. SphK2 inhibitors are featured in more recent patent applications. Interestingly, both SphK1 and SphK2 inhibition and gene 'knockout' share opposing effects on circulating S1P levels: SphK1 inhibition/gene ablation decreases, while SphK2 inhibition/gene ablation increases, blood S1P. As understanding of S1P's physiological roles increases and more drug-like SphK inhibitors emerge, inhibiting one or both SphK isotypes could provide unique strategies for treating disease.
Collapse
Affiliation(s)
- Kevin R Lynch
- a Department of Pharmacology , University of Virginia , Charlottesville , VA , USA
| | | | - Webster L Santos
- c Department of Chemistry and Virginia Tech Center for Drug Discovery , Virginia Tech , Blacksburg , VA , USA
| |
Collapse
|
35
|
Vogt D, Stark H. Therapeutic Strategies and Pharmacological Tools Influencing S1P Signaling and Metabolism. Med Res Rev 2016; 37:3-51. [PMID: 27480072 DOI: 10.1002/med.21402] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 06/01/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023]
Abstract
During the last two decades the study of the sphingolipid anabolic, catabolic, and signaling pathways has attracted enormous interest. Especially the introduction of fingolimod into market as first p.o. therapeutic for the treatment of multiple sclerosis has boosted this effect. Although the complex regulation of sphingosine-1-phosphate (S1P) and other catabolic and anabolic sphingosine-related compounds is not fully understood, the influence on different (patho)physiological states from inflammation to cytotoxicity as well as the availability of versatile pharmacological tools that represent new approaches to study these states are described. Here, we have summarized various aspects concerning the many faces of sphingolipid function modulation by different pharmacological tools up to clinical candidates. Due to the immense heterogeneity of physiological or pharmacological actions and complex cross regulations, it is difficult to predict their role in upcoming therapeutic approaches. Currently, inflammatory, immunological, and/or antitumor aspects are discussed.
Collapse
Affiliation(s)
- Dominik Vogt
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, D-60438, Frankfurt, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225, Düsseldorf, Germany
| |
Collapse
|
36
|
Development of hydroxy-based sphingosine kinase inhibitors and anti-inflammation in dextran sodium sulfate induced colitis in mice. Bioorg Med Chem 2016; 24:3218-30. [DOI: 10.1016/j.bmc.2016.05.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/20/2016] [Accepted: 05/21/2016] [Indexed: 01/01/2023]
|
37
|
MacRitchie N, Volpert G, Al Washih M, Watson DG, Futerman AH, Kennedy S, Pyne S, Pyne NJ. Effect of the sphingosine kinase 1 selective inhibitor, PF-543 on arterial and cardiac remodelling in a hypoxic model of pulmonary arterial hypertension. Cell Signal 2016; 28:946-55. [PMID: 27063355 PMCID: PMC4913619 DOI: 10.1016/j.cellsig.2016.03.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 03/29/2016] [Accepted: 03/29/2016] [Indexed: 12/20/2022]
Abstract
Recent studies have demonstrated that the expression of sphingosine kinase 1, the enzyme that catalyses formation of the bioactive lipid, sphingosine 1-phosphate, is increased in lungs from patients with pulmonary arterial hypertension. In addition, Sk1−/− mice are protected from hypoxic-induced pulmonary arterial hypertension. Therefore, we assessed the effect of the sphingosine kinase 1 selective inhibitor, PF-543 and a sphingosine kinase 1/ceramide synthase inhibitor, RB-005 on pulmonary and cardiac remodelling in a mouse hypoxic model of pulmonary arterial hypertension. Administration of the potent sphingosine kinase 1 inhibitor, PF-543 in a mouse hypoxic model of pulmonary hypertension had no effect on vascular remodelling but reduced right ventricular hypertrophy. The latter was associated with a significant reduction in cardiomyocyte death. The protection involves a reduction in the expression of p53 (that promotes cardiomyocyte death) and an increase in the expression of anti-oxidant nuclear factor (erythroid-derived 2)-like 2 (Nrf-2). In contrast, RB-005 lacked effects on right ventricular hypertrophy, suggesting that sphingosine kinase 1 inhibition might be nullified by concurrent inhibition of ceramide synthase. Therefore, our findings with PF-543 suggest an important role for sphingosine kinase 1 in the development of hypertrophy in pulmonary arterial hypertension. PF-543, a sphingosine kinase 1 inhibitor reduces cardiac hypertrophy in a mouse pulmonary arterial hypertension (PAH) model This results in reduced cardiomyocyte apoptosis PF-543 reduces PARP processing and p53 expression and increases Nrf-2 expression in the right ventricle of mice with PAH
Collapse
Affiliation(s)
- Neil MacRitchie
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, UK
| | - Giora Volpert
- Department of Biological Chemistry, Weizmann Insitute of Science, Rehovot 76100, Israel
| | - Mohammed Al Washih
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, UK
| | - David G Watson
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, UK
| | - Anthony H Futerman
- Department of Biological Chemistry, Weizmann Insitute of Science, Rehovot 76100, Israel
| | - Simon Kennedy
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, G12 8QQ, UK
| | - Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, UK
| | - Nigel J Pyne
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, UK.
| |
Collapse
|
38
|
A reflection of the lasting contributions from Dr. Robert Bittman to sterol trafficking, sphingolipid and phospholipid research. Prog Lipid Res 2015; 61:19-29. [PMID: 26584871 DOI: 10.1016/j.plipres.2015.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/21/2015] [Accepted: 10/28/2015] [Indexed: 10/22/2022]
Abstract
With the passing of Dr. Robert Bittman from pancreatic cancer on the 1st October 2014, the lipid research field lost one of the most influential and significant personalities. Robert Bittman's genius was in chemical design and his contribution to the lipid research field was truly immense. The reagents and chemicals he designed and synthesised allowed interrogation of the role of lipids in constituting complex biophysical membranes, sterol transfer and in cellular communication networks. Here we provide a review of these works which serve as a lasting memory to his life.
Collapse
|
39
|
Sanllehí P, Abad JL, Casas J, Delgado A. Inhibitors of sphingosine-1-phosphate metabolism (sphingosine kinases and sphingosine-1-phosphate lyase). Chem Phys Lipids 2015. [PMID: 26200919 DOI: 10.1016/j.chemphyslip.2015.07.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Sphingolipids (SLs) are essential structural and signaling molecules of eukaryotic cells. Among them, sphingosine 1 phosphate (S1P) is a recognized promoter of cell survival, also involved, inter alia, in inflammation and tumorigenesis processes. The knowledge and modulation of the enzymes implicated in the biosynthesis and degradation of S1P are capital to control the intracellular levels of this lipid and, ultimately, to determine the cell fate. Starting with a general overview of the main metabolic pathways involved in SL metabolism, this review is mainly focused on the description of the most relevant findings concerning the development of modulators of S1P, namely inhibitors of the enzymes regulating S1P synthesis (sphingosine kinases) and degradation (sphingosine 1 phosphate phosphatase and lyase). In addition, a brief overview of the most significant agonists and antagonists at the S1P receptors is also addressed.
Collapse
Affiliation(s)
- Pol Sanllehí
- Research Unit on Bioactive Molecules, Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, E-08034 Barcelona, Spain; University of Barcelona (UB), Faculty of Pharmacy, Department of Pharmacology and Medicinal Chemistry, Unit of Pharmaceutical Chemistry (Associated Unit to CSIC), Avga. Joan XXIII s/n, E-08028 Barcelona, Spain
| | - José-Luis Abad
- Research Unit on Bioactive Molecules, Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, E-08034 Barcelona, Spain
| | - Josefina Casas
- Research Unit on Bioactive Molecules, Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, E-08034 Barcelona, Spain
| | - Antonio Delgado
- Research Unit on Bioactive Molecules, Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, E-08034 Barcelona, Spain; University of Barcelona (UB), Faculty of Pharmacy, Department of Pharmacology and Medicinal Chemistry, Unit of Pharmaceutical Chemistry (Associated Unit to CSIC), Avga. Joan XXIII s/n, E-08028 Barcelona, Spain.
| |
Collapse
|
40
|
Woodcock JM, Coolen C, Goodwin KL, Baek DJ, Bittman R, Samuel MS, Pitson SM, Lopez AF. Destabilisation of dimeric 14-3-3 proteins as a novel approach to anti-cancer therapeutics. Oncotarget 2015; 6:14522-36. [PMID: 25971334 PMCID: PMC4546484 DOI: 10.18632/oncotarget.3995] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 04/11/2015] [Indexed: 12/24/2022] Open
Abstract
14-3-3 proteins play a pivotal role in controlling cell proliferation and survival, two commonly dysregulated hallmarks of cancers. 14-3-3 protein expression is enhanced in many human cancers and correlates with more aggressive tumors and poor prognosis, suggesting a role for 14-3-3 proteins in tumorigenesis and/or progression. We showed previously that the dimeric state of 14-3-3 proteins is regulated by the lipid sphingosine, a physiological inducer of apoptosis. As the functions of 14-3-3 proteins are dependent on their dimeric state, this sphingosine-mediated 14-3-3 regulation provides a possible means to target dimeric 14-3-3 for therapeutic effect. However, sphingosine mimics are needed that are not susceptible to sphingolipid metabolism. We show here the identification and optimization of sphingosine mimetics that render dimeric 14-3-3 susceptible to phosphorylation at a site buried in the dimer interface and induce mitochondrial-mediated apoptosis. Two such compounds, RB-011 and RB-012, disrupt 14-3-3 dimers at low micromolar concentrations and induce rapid down-regulation of Raf-MAPK and PI3K-Akt signaling in Jurkat cells. Importantly, both RB-011 and RB-012 induce apoptosis of human A549 lung cancer cells and RB-012, through disruption of MAPK signaling, reduces xenograft growth in mice. Thus, these compounds provide proof-of-principle for this novel 14-3-3-targeting approach for anti-cancer drug discovery.
Collapse
Affiliation(s)
- Joanna M. Woodcock
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Carl Coolen
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Katy L. Goodwin
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Dong Jae Baek
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, NY, USA
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, NY, USA
| | - Michael S. Samuel
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Stuart M. Pitson
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Angel F. Lopez
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
41
|
Pulkoski-Gross MJ, Donaldson JC, Obeid LM. Sphingosine-1-phosphate metabolism: A structural perspective. Crit Rev Biochem Mol Biol 2015; 50:298-313. [PMID: 25923252 DOI: 10.3109/10409238.2015.1039115] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Sphingolipids represent an important class of bioactive signaling lipids which have key roles in numerous cellular processes. Over the last few decades, the levels of bioactive sphingolipids and/or their metabolizing enzymes have been realized to be important factors involved in disease development and progression, most notably in cancer. Targeting sphingolipid-metabolizing enzymes in disease states has been the focus of many studies and has resulted in a number of pharmacological inhibitors, with some making it into the clinic as therapeutics. In order to better understand the regulation of sphingolipid-metabolizing enzymes as well as to develop much more potent and specific inhibitors, the field of sphingolipids has recently taken a turn toward structural biology. The last decade has seen the structural determination of a number of sphingolipid enzymes and effector proteins. In these terms, one of the most complete arms of the sphingolipid pathway is the sphingosine-1-phosphate (S1P) arm. The structures of proteins involved in the function and regulation of S1P are being used to investigate further the regulation of said proteins as well as in the design and development of inhibitors as potential therapeutics.
Collapse
Affiliation(s)
| | - Jane C Donaldson
- b Department of Medicine , Stony Brook University , Stony Brook , NY , USA .,c Stony Brook Cancer Center , Stony Brook , NY , USA , and
| | - Lina M Obeid
- b Department of Medicine , Stony Brook University , Stony Brook , NY , USA .,c Stony Brook Cancer Center , Stony Brook , NY , USA , and.,d Northport Veterans Affairs Medical Center , Northport , NY , USA
| |
Collapse
|
42
|
Wang X, Sun C, Fang L, Yin D. Discovery of novel sphingosine kinase 1 inhibitorsvia structure-based hierarchical virtual screening. MEDCHEMCOMM 2015. [DOI: 10.1039/c4md00312h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hierarchical structure-based virtual screening against the sphingosine kinase 1(SphK1) binding pocket was performed. 25 compounds were selected for biological evaluation. Compound 25 exhibited comparable SphK1 and SphK2 inhibitory activities and anti-proliferative effects on U937 cells to the positive control N,N-dimethylsphingosine (DMS) 1. Further molecule dynamic (MD) simulations revealed the binding mode between SphK1 and 25.
Collapse
Affiliation(s)
- Xiaojian Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines
- Institute of Materia Medica
- Peking Union Medical College
- Chinese Academy of Medical Sciences
- Beijing 100050
| | - Chenbin Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines
- Institute of Materia Medica
- Peking Union Medical College
- Chinese Academy of Medical Sciences
- Beijing 100050
| | - Liang Fang
- Department of Medicinal Chemistry
- Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation
- Institute of Materia Medica
- Peking Union Medical College
- Chinese Academy of Medical Sciences
| | - Dali Yin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines
- Institute of Materia Medica
- Peking Union Medical College
- Chinese Academy of Medical Sciences
- Beijing 100050
| |
Collapse
|
43
|
Cingolani F, Casasampere M, Sanllehí P, Casas J, Bujons J, Fabrias G. Inhibition of dihydroceramide desaturase activity by the sphingosine kinase inhibitor SKI II. J Lipid Res 2014; 55:1711-20. [PMID: 24875537 PMCID: PMC4109765 DOI: 10.1194/jlr.m049759] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/27/2014] [Indexed: 01/05/2023] Open
Abstract
Sphingosine kinase inhibitor (SKI) II has been reported as a dual inhibitor of sphingosine kinases (SKs) 1 and 2 and has been extensively used to prove the involvement of SKs and sphingosine-1-phosphate (S1P) in cellular processes. Dihydroceramide desaturase (Des1), the last enzyme in the de novo synthesis of ceramide (Cer), regulates the balance between dihydroceramides (dhCers) and Cers. Both SKs and Des1 have interest as therapeutic targets. Here we show that SKI II is a noncompetitive inhibitor (Ki = 0.3 μM) of Des1 activity with effect also in intact cells without modifying Des1 protein levels. Molecular modeling studies support that the SKI II-induced decrease in Des1 activity could result from inhibition of NADH-cytochrome b5 reductase. SKI II, but not the SK1-specific inhibitor PF-543, provoked a remarkable accumulation of dhCers and their metabolites, while both SKI II and PF-543 reduced S1P to almost undetectable levels. SKI II, but not PF543, reduced cell proliferation with accumulation of cells in the G0/G1 phase. SKI II, but not PF543, induced autophagy. These overall findings should be taken into account when using SKI II as a pharmacological tool, as some of the effects attributed to decreased S1P may actually be caused by augmented dhCers and/or their metabolites.
Collapse
Affiliation(s)
- Francesca Cingolani
- Research Unit on BioActive Molecules (RUBAM), Departments of Biomedicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - Mireia Casasampere
- Research Unit on BioActive Molecules (RUBAM), Departments of Biomedicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - Pol Sanllehí
- Research Unit on BioActive Molecules (RUBAM), Departments of Biomedicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
- Faculty of Pharmacy, Unit of Pharmaceutical Chemistry (Associated Unit to CSIC), University of Barcelona, E-08028 Barcelona, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Departments of Biomedicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - Jordi Bujons
- Biological Chemistry and Molecular Modeling, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - Gemma Fabrias
- Research Unit on BioActive Molecules (RUBAM), Departments of Biomedicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| |
Collapse
|
44
|
Plano D, Amin S, Sharma AK. Importance of sphingosine kinase (SphK) as a target in developing cancer therapeutics and recent developments in the synthesis of novel SphK inhibitors. J Med Chem 2014; 57:5509-24. [PMID: 24471412 DOI: 10.1021/jm4011687] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Sphingosine kinase (SphK) is an oncogenic lipid kinase that regulates the sphingolipid metabolic pathway that has been shown to play a role in numerous hyperproliferative/inflammatory diseases. The SphK isoforms (SphK1 and SphK2) catalyze the conversion of the proapoptotic substrate d-erythrosphingosine to the promitogenic/migratory product sphingosine 1-phosphate (S1P). Accumulation of S1P has been linked to the development/progression of cancer and various other diseases including, but not limited to, asthma, inflammatory bowel disease, rheumatoid arthritis, and diabetic nephropathy. SphK therefore represents a potential new target for developing novel therapeutics for cancer and other diseases. This finding has stimulated the development and evaluation of numerous SphK inhibitors over the past decade or so. In this review, we highlight the recent advancement in the field of SphK inhibitors including SphK1 and SphK2 specific inhibitors. Both sphingolipid based and nolipidic small molecule inhibitors and their importance in treatment of cancer and other diseases are discussed.
Collapse
Affiliation(s)
- Daniel Plano
- Department of Pharmacology, Penn State Hershey Cancer Institute, CH72, Penn State College of Medicine , 500 University Drive, Hershey, Pennsylvania 17033, United States
| | | | | |
Collapse
|
45
|
Ohotski J, Rosen H, Bittman R, Pyne S, Pyne NJ. Sphingosine kinase 2 prevents the nuclear translocation of sphingosine 1-phosphate receptor-2 and tyrosine 416 phosphorylated c-Src and increases estrogen receptor negative MDA-MB-231 breast cancer cell growth: The role of sphingosine 1-phosphate receptor-4. Cell Signal 2014; 26:1040-7. [PMID: 24486401 DOI: 10.1016/j.cellsig.2014.01.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 01/24/2014] [Indexed: 10/25/2022]
Abstract
We demonstrate that pre-treatment of estrogen receptor negative MDA-MB-231 breast cancer cells containing ectopically expressed HA-tagged sphingosine 1-phosphate receptor-2 (S1P2) with the sphingosine kinase 1/2 inhibitor SKi (2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole) or the sphingosine kinase 2 selective inhibitor (R)-FTY720 methyl ether (ROMe) or sphingosine kinase 2 siRNA induced the translocation of HA-tagged S1P2 and Y416 phosphorylated c-Src to the nucleus of these cells. This is associated with reduced growth of HA-tagged S1P2 over-expressing MDA-MB-231 cells. Treatment of HA-S1P2 over-expressing MDA-MB-231 cells with the sphingosine 1-phosphate receptor-4 (S1P4) antagonist CYM50367 or with S1P4 siRNA also promoted nuclear translocation of HA-tagged S1P2. These findings identify for the first time a signaling pathway in which sphingosine 1-phosphate formed by sphingosine kinase 2 binds to S1P4 to prevent nuclear translocation of S1P2 and thereby promote the growth of estrogen receptor negative breast cancer cells.
Collapse
Affiliation(s)
- Jan Ohotski
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Hugh Rosen
- The Scripps Research Institute, Department of Chemistry, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, United States
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College, The City University of New York, Flushing, NY 11367-1597, United States
| | - Susan Pyne
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Nigel J Pyne
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, United Kingdom.
| |
Collapse
|
46
|
Pyne NJ, Ohotski J, Bittman R, Pyne S. The role of sphingosine 1-phosphate in inflammation and cancer. Adv Biol Regul 2014; 54:121-129. [PMID: 24070975 DOI: 10.1016/j.jbior.2013.08.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 08/31/2013] [Accepted: 08/31/2013] [Indexed: 06/02/2023]
Abstract
The enzymes that catalyze formation of the bioactive sphingolipid, sphingosine 1-phosphate, sphingosine kinase 1 and 2, are predictive markers in inflammatory diseases and cancer as evidenced by data from patients, knockout mice and the use of available molecular and chemical inhibitors. Thus, there is a compelling case for therapeutic targeting of sphingosine kinase. In addition, there are several examples of functional interaction between sphingosine 1-phosphate receptors and sphingosine kinase 1 that can drive malicious amplification loops that promote cancer cell growth. These novel aspects of sphingosine 1-phosphate pathobiology are reviewed herein.
Collapse
Affiliation(s)
- Nigel J Pyne
- Cell Biology Research Group, Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Jan Ohotski
- Cell Biology Research Group, Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, New York 11367-1597, USA
| | - Susan Pyne
- Cell Biology Research Group, Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
47
|
Baek DJ, MacRitchie N, Anthony NG, Mackay SP, Pyne S, Pyne NJ, Bittman R. Structure-activity relationships and molecular modeling of sphingosine kinase inhibitors. J Med Chem 2013; 56:9310-27. [PMID: 24164513 PMCID: PMC3848335 DOI: 10.1021/jm401399c] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
![]()
The
design, synthesis, and evaluation of the potency of new isoform-selective
inhibitors of sphingosine kinases 1 and 2 (SK1 and SK2), the enzyme
that catalyzes the phosphorylation of d-erythro-sphingosine to produce the key signaling lipid, sphingosine 1-phosphate,
are described. Recently, we reported that 1-(4-octylphenethyl)piperidin-4-ol
(RB-005) is a selective inhibitor of SK1. Here we report
the synthesis of 43 new analogues of RB-005, in which
the lipophilic tail, polar headgroup, and linker region were modified
to extend the structure–activity relationship profile for this
lead compound, which we explain using modeling studies with the recently
published crystal structure of SK1. We provide a basis for the key
residues targeted by our profiled series and provide further evidence
for the ability to discriminate between the two isoforms using pharmacological
intervention.
Collapse
Affiliation(s)
- Dong Jae Baek
- Department of Chemistry and Biochemistry, Queens College, The City University of New York , Flushing, New York 11367-1597, United States
| | | | | | | | | | | | | |
Collapse
|
48
|
Liu Z, MacRitchie N, Pyne S, Pyne NJ, Bittman R. Synthesis of (S)-FTY720 vinylphosphonate analogues and evaluation of their potential as sphingosine kinase 1 inhibitors and activators. Bioorg Med Chem 2013; 21:2503-10. [PMID: 23541833 DOI: 10.1016/j.bmc.2013.02.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/19/2013] [Accepted: 02/27/2013] [Indexed: 11/30/2022]
Abstract
Sphingosine kinase 1 (SK1) is over-expressed in many cancers where it provides a selective growth and survival advantage to these cells. SK1 is thus a target for anti-cancer agents that can promote apoptosis of cancer cells. In previous work, we synthesized a novel allosteric SK1 inhibitor, (S)-FTY720 vinylphosphonate. We now report a more expeditious route to this inhibitor which features B-alkyl Suzuki coupling as a key step and show that replacement of the amino group in (S)-FTY720 vinylphosphonate with an azido group converts the vinylphosphonate from an allosteric inhibitor to an activator of SK1 at low micromolar concentrations. Our results demonstrate the feasibility of using the (S)-FTY720 vinylphosphonate scaffold to define structure-activity relationships in the allosteric site of SK1.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Chemistry and Biochemistry, Queens College, The City University of New York, Flushing, NY 11367-1597, USA
| | | | | | | | | |
Collapse
|
49
|
Byun HS, Pyne S, Macritchie N, Pyne NJ, Bittman R. Novel sphingosine-containing analogues selectively inhibit sphingosine kinase (SK) isozymes, induce SK1 proteasomal degradation and reduce DNA synthesis in human pulmonary arterial smooth muscle cells. MEDCHEMCOMM 2013; 4. [PMID: 24396570 DOI: 10.1039/c3md00201b] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Sphingosine 1-phosphate (S1P) is involved in hyper-proliferative diseases such as cancer and pulmonary arterial hypertension. We have synthesized inhibitors that are selective for the two isoforms of sphingosine kinase (SK1 and SK2) that catalyze the synthesis of S1P. A thiourea adduct of sphinganine (F02) is selective for SK2 whereas the 1-deoxysphinganines 55-21 and 77-7 are selective for SK1. (2S,3R)-1-Deoxysphinganine (55-21) induced the proteasomal degradation of SK1 in human pulmonary arterial smooth muscle cells and inhibited DNA synthesis, while the more potent SK1 inhibitors PF-543 and VPC96091 failed to inhibit DNA synthesis. These findings indicate that moderate potency inhibitors such as 55-21 are likely to have utility in unraveling the functions of SK1 in inflammatory and hyperproliferative disorders.
Collapse
Affiliation(s)
- Hoe-Sup Byun
- Department of Chemistry and Biochemistry, Queens College, The City University of New York, Flushing, NY 11367-1597, USA. Tel: +1 718-997-3279
| | - Susan Pyne
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, UK
| | - Neil Macritchie
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, UK
| | - Nigel J Pyne
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow G4 0RE, UK
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College, The City University of New York, Flushing, NY 11367-1597, USA. Tel: +1 718-997-3279
| |
Collapse
|