1
|
Taylor OB, DeGroff N, El-Hodiri HM, Gao C, Fischer AJ. Sphingosine-1-phosphate signaling regulates the ability of Müller glia to become neurogenic, proliferating progenitor-like cells. eLife 2025; 13:RP102151. [PMID: 40047533 PMCID: PMC11884796 DOI: 10.7554/elife.102151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025] Open
Abstract
The purpose of these studies is to investigate how Sphingosine-1-phosphate (S1P) signaling regulates glial phenotype, dedifferentiation of Müller glia (MG), reprogramming into proliferating MG-derived progenitor cells (MGPCs), and neuronal differentiation of the progeny of MGPCs in the chick retina. We found that S1P-related genes are highly expressed by retinal neurons and glia, and levels of expression were dynamically regulated following retinal damage. Drug treatments that activate S1P receptor 1 (S1PR1) or increase levels of S1P suppressed the formation of MGPCs. Conversely, treatments that inhibit S1PR1 or decrease levels of S1P stimulated the formation of MGPCs. Inhibition of S1P receptors or S1P synthesis significantly enhanced the neuronal differentiation of the progeny of MGPCs. We report that S1P-related gene expression in MG is modulated by microglia and inhibition of S1P receptors or S1P synthesis partially rescues the loss of MGPC formation in damaged retinas missing microglia. Finally, we show that TGFβ/Smad3 signaling in the resting retina maintains S1PR1 expression in MG. We conclude that the S1P signaling is dynamically regulated in MG and MGPCs in the chick retina, and activation of S1P signaling depends, in part, on signals produced by reactive microglia.
Collapse
Affiliation(s)
- Olivia B Taylor
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
- Neuroscience Graduate Program, The Ohio State UniversityColumbusUnited States
| | - Nicholas DeGroff
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
| | - Heithem M El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
| | - Chengyu Gao
- Campus Chemical Instrument Center, Mass Spectrometry and Proteomics Facility, The Ohio State UniversityColumbusUnited States
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
| |
Collapse
|
2
|
McKenna S, Jung KI, Wolf JJ, Seo YJ, Hahm B. Multiple sphingolipid-metabolizing enzymes modulate influenza virus replication. Virology 2025; 603:110367. [PMID: 39754863 PMCID: PMC11793951 DOI: 10.1016/j.virol.2024.110367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 01/06/2025]
Abstract
The sphingolipid network is sustained principally by the balance of bioactive sphingolipid molecules and their regulation by sphingolipid-metabolizing enzymes. The components in the lipid system display key functions in numerous cellular and disease conditions including virus infections. During the COVID-19 pandemic, there was a fruitful effort to use an inhibitor that blocks the activity of sphingosine kinase (SphK) 2 to cure the devastating disease. Support for the inhibitor came from pre-clinical research on influenza where the inhibitor demonstrated effective protection of mice from influenza-induced morbidity and mortality. This highlights the importance of basic and translational research on the sphingolipid system for improving human health. Multiple sphingolipid-metabolizing enzymes have been reported to regulate influenza virus replication and propagation. In this review, the emphasis is placed on the roles of these enzymes that impact influenza virus life cycle and the conceivable mechanisms for the interplay between influenza virus and the sphingolipid pathway.
Collapse
Affiliation(s)
- Savannah McKenna
- Departments of Surgery & Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA
| | - Kwang Il Jung
- Departments of Surgery & Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA
| | - Jennifer J Wolf
- Departments of Surgery & Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA
| | - Young-Jin Seo
- Department of Life Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| | - Bumsuk Hahm
- Departments of Surgery & Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
3
|
Taylor O, DeGroff N, El-Hodiri H, Gao C, Fischer AJ. Sphingosine-1-phosphate signaling regulates the ability of Müller glia to become neurogenic, proliferating progenitor-like cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.06.606815. [PMID: 39149287 PMCID: PMC11326190 DOI: 10.1101/2024.08.06.606815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The purpose of these studies is to investigate how Sphingosine-1-phosphate (S1P) signaling regulates glial phenotype, dedifferentiation of Müller glia (MG), reprogramming into proliferating MG-derived progenitor cells (MGPCs), and neuronal differentiation of the progeny of MGPCs in the chick retina. We found that S1P-related genes are highly expressed by retinal neurons and glia, and levels of expression were dynamically regulated following retinal damage. Drug treatments that activate S1P receptor 1 (S1PR1) or increase levels of S1P suppressed the formation of MGPCs. Conversely, treatments that inhibit S1PR1 or decrease levels of S1P stimulated the formation of MGPCs. Inhibition of S1P receptors or S1P synthesis significantly enhanced the neuronal differentiation of the progeny of MGPCs. We report that S1P-related gene expression in MG is modulated by microglia and inhibition of S1P receptors or S1P synthesis partially rescues the loss of MGPC formation in damaged retinas missing microglia. Finally, we show that TGFβ/Smad3 signaling in the resting retina maintains S1PR1 expression in MG. We conclude that the S1P signaling is dynamically regulated in MG and MGPCs in the chick retina, and activation of S1P signaling depends, in part, on signals produced by reactive microglia.
Collapse
Affiliation(s)
- Olivia Taylor
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Nick DeGroff
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Heithem El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Chengyu Gao
- Campus Chemical Instrument Center, Mass Spectrometry & Proteomics Facility, The Ohio State University, Columbus, OH 43210, USA
| | - Andy J. Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
4
|
Ahmed T, Suzuki T, Terao R, Yamagishi R, Fujino R, Azuma K, Soga H, Ueta T, Honjo M, Watanabe S, Yoshioka K, Takuwa Y, Aihara M. Roles of Sphingosine Kinase and Sphingosine-1-Phosphate Receptor 2 in Endotoxin-Induced Acute Retinal Inflammation. Ocul Immunol Inflamm 2024; 32:1633-1647. [PMID: 38100527 DOI: 10.1080/09273948.2023.2273963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 12/17/2023]
Abstract
PURPOSE To investigate the roles of sphingosine kinases (SphKs) and sphingosine-1-phosphate receptors (S1PRs) in endotoxin-induced uveitis (EIU) mice. METHODS EIU model was induced using an intraperitoneal injection of lipopolysaccharide (LPS). The expression of SphKs and S1PRs in the retina was assessed using quantitative polymerase chain reaction (qPCR) and immunofluorescence. The effects of S1PR antagonists on the expression of inflammatory cytokines in the retina were evaluated using qPCR and western blotting. Effects of leukocyte infiltration of the retinal vessels were evaluated to determine the effects of the S1PR2 antagonist and genetic deletion of S1PR2 on retinal inflammation. RESULTS Retinal SphK1 expression was significantly upregulated in EIU. SphK1 was expressed in the GCL, IPL, and OPL and S1PR2 was expressed in the GCL, INL, and OPL. Positive cells in IPL and OPL of EIU retina were identified as endothelial cells. S1PR2 antagonist and genetic deletion of S1PR2 significantly suppressed the expression of IL-1α, IL-6, TNF-α, and ICAM-1, whereas S1PR1/3 antagonist did not. Use of S1PR2 antagonist and S1PR2 knockout in mice significantly ameliorated leukocyte adhesion induced by LPS. CONCLUSION SphK1/S1P/S1PR2 signaling was upregulated in EIU and S1PR2 inhibition suppressed inflammatory response. Targeting this signaling pathway has potential for treating retinal inflammatory diseases.
Collapse
Affiliation(s)
- Tazbir Ahmed
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takafumi Suzuki
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Ryo Terao
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Reiko Yamagishi
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Ryosuke Fujino
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kunihiro Azuma
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hirotsugu Soga
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takashi Ueta
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Megumi Honjo
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Sumiko Watanabe
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kazuaki Yoshioka
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Ishikawa, Japan
| | - Yoh Takuwa
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Ishikawa, Japan
| | - Makoto Aihara
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
5
|
Kataoka T, Ito H, Mori T, Hotta Y, Sanagawa A, Maeda Y, Furukawa-Hibi Y, Kimura K. Testosterone improved erectile function by upregulating transcriptional expression of growth factors in late androgen replacement therapy model rats. Int J Impot Res 2024; 36:437-442. [PMID: 36310186 DOI: 10.1038/s41443-022-00627-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 09/27/2022] [Indexed: 11/09/2022]
Abstract
We previously showed that castration of rats reduced erectile function over time; when testosterone replacement therapy was started 4 weeks after castration, erectile function improved. In this study, we examined the mechanism of improvement in erectile function following testosterone replacement therapy in rats. Thirty 12-week-old rats were divided into castrated (Cast), castrated with subcutaneous administration of testosterone (Cast + T), and sham (Sham) groups. Erectile function and mRNA and protein expression were evaluated in the rats by using standard methods. To assess erectile function, we measured the intracavernosal pressure, mean arterial pressure, mRNA expression of endothelial growth factors, and protein expression of endothelial nitric oxide synthase (eNOS). The intracavernosal pressure/mean arterial pressure ratio was significantly lower in the Cast group, and testosterone administration significantly improved (P = 0.017). Compared to the Cast group, the Cast+T group exhibited significantly increased mRNA expressions of vascular endothelial growth factor A (VEGF-A), intercellular adhesion molecule 1 (ICAM-1), transforming growth factor-β (TGF-β), nerve growth factor (NGF), α-smooth muscle actin (α-SMA), caveolae associated protein 1 (Cavin-1), Cavin-2, Cavin-3, sirtuin 1 (Sirt-1), sphingosine-1-phosphate 1 (S1P1), S1P2, and S1P3 and eNOS protein expression. Testosterone replacement therapy improved erectile function in castrated rats by increasing growth factors and eNOS protein.
Collapse
Affiliation(s)
- Tomoya Kataoka
- Department of Clinical Pharmaceutics, Graduate School of Medical Sciences, Nagoya City University, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
- Department of Pharmacology, Kataoka's lab, Graduate School of Pharmaceutical Sciences, Chiba Institute of Science, 15-8 Shiomi-cho, Choshi, Chiba, 288-0025, Japan.
| | - Hiroto Ito
- Department of Hospital Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Taiki Mori
- Department of Hospital Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Yuji Hotta
- Department of Hospital Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Akimasa Sanagawa
- Department of Hospital Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Yasuhiro Maeda
- Department of Hospital Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Yoko Furukawa-Hibi
- Department of Clinical Pharmaceutics, Graduate School of Medical Sciences, Nagoya City University, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
- Department of Hospital Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Kazunori Kimura
- Department of Clinical Pharmaceutics, Graduate School of Medical Sciences, Nagoya City University, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
- Department of Hospital Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan.
| |
Collapse
|
6
|
Velagapudi S, Wang D, Poti F, Feuerborn R, Robert J, Schlumpf E, Yalcinkaya M, Panteloglou G, Potapenko A, Simoni M, Rohrer L, Nofer JR, von Eckardstein A. Sphingosine-1-phosphate receptor 3 regulates the transendothelial transport of high-density lipoproteins and low-density lipoproteins in opposite ways. Cardiovasc Res 2024; 120:476-489. [PMID: 38109696 PMCID: PMC11060483 DOI: 10.1093/cvr/cvad183] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/08/2023] [Accepted: 10/20/2023] [Indexed: 12/20/2023] Open
Abstract
AIMS The entry of lipoproteins from blood into the arterial wall is a rate-limiting step in atherosclerosis. It is controversial whether this happens by filtration or regulated transendothelial transport.Because sphingosine-1-phosphate (S1P) preserves the endothelial barrier, we investigated in vivo and in vitro, whether S1P and its cognate S1P-receptor 3 (S1P3) regulate the transendothelial transport of lipoproteins. METHODS AND RESULTS Compared to apoE-haploinsufficient mice (CTRL), apoE-haploinsufficient mice with additional endothelium-specific knock-in of S1P3 (S1P3-iECKI) showed decreased transport of LDL and Evan's Blue but increased transport of HDL from blood into the peritoneal cave. After 30 weeks of high-fat diet feeding, S1P3-iECKI mice had lower levels of non-HDL-cholesterol and less atherosclerosis than CTRL mice. In vitro stimulation with an S1P3 agonist increased the transport of 125I-HDL but decreased the transport of 125I-LDL through human aortic endothelial cells (HAECs). Conversely, inhibition or knock-down of S1P3 decreased the transport of 125I-HDL but increased the transport of 125I-LDL. Silencing of SCARB1 encoding scavenger receptor B1 (SR-BI) abrogated the stimulation of 125I-HDL transport by the S1P3 agonist. The transendothelial transport of 125I-LDL was decreased by silencing of SCARB1 or ACVLR1 encoding activin-like kinase 1 but not by interference with LDLR. None of the three knock-downs prevented the stimulatory effect of S1P3 inhibition on transendothelial 125I-LDL transport. CONCLUSION S1P3 regulates the transendothelial transport of HDL and LDL oppositely by SR-BI-dependent and SR-BI-independent mechanisms, respectively. This divergence supports a contention that lipoproteins pass the endothelial barrier by specifically regulated mechanisms rather than passive filtration.
Collapse
Affiliation(s)
- Srividya Velagapudi
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Dongdong Wang
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Francesco Poti
- Department of Medicine and Surgery—Unit of Neurosciences, University of Parma, Parma, Italy
- Department of Biomedical, Metabolic and Neural Sciences—Unit of Endocrinology, University of Modena and Reggio Emilia, Modena, Italy
| | - Renata Feuerborn
- Central Laboratory Facility, University Hospital of Münster, Münster, Germany
| | - Jerome Robert
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Eveline Schlumpf
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Mustafa Yalcinkaya
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Grigorios Panteloglou
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Anton Potapenko
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences—Unit of Endocrinology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Jerzy-Roch Nofer
- Central Laboratory Facility, University Hospital of Münster, Münster, Germany
- Institute of Laboratory Medicine, Marien-Hospital Osnabrück, Niels-Stensen-Kliniken, Osnabrück, Germany
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| |
Collapse
|
7
|
Alshaikh RA, Zaki RGE, El Din RAS, Ryan KB, Waeber C. Siponimod As a Novel Inhibitor of Retinal Angiogenesis: In Vitro and In Vivo Evidence of Therapeutic Efficacy. J Pharmacol Exp Ther 2023; 386:224-241. [PMID: 37188532 DOI: 10.1124/jpet.122.001529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/13/2023] [Accepted: 05/05/2023] [Indexed: 05/17/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) receptors control endothelial cell proliferation, migration, and survival. Evidence of the ability of S1P receptor modulators to influence multiple endothelial cell functions suggests their potential use for antiangiogenic effect. The main purpose of our study was to investigate the potential of siponimod for the inhibition of ocular angiogenesis in vitro and in vivo. We investigated the effects of siponimod on the metabolic activity (thiazolyl blue tetrazolium bromide assay), cell toxicity (lactate dehydrogenase release), basal proliferation and growth factor-induced proliferation (bromodeoxyuridine assay), and migration (transwell migration assay) of human umbilical vein endothelial cells (HUVEC) and retinal microvascular endothelial cells (HRMEC). The effects of siponimod on HRMEC monolayer integrity, barrier function under basal conditions, and tumor necrosis factor alpha (TNF-α)-induced disruption were assessed using the transendothelial electrical resistance and fluorescein isothiocyanate-dextran permeability assays. Siponimod's effect on TNF-α-induced distribution of barrier proteins in HRMEC was investigated using immunofluorescence. Finally, the effect of siponimod on ocular neovascularization in vivo was assessed using suture-induced corneal neovascularization in albino rabbits. Our results show that siponimod did not affect endothelial cell proliferation or metabolic activity but significantly inhibited endothelial cell migration, increased HRMEC barrier integrity, and reduced TNF-α-induced barrier disruption. Siponimod also protected against TNF-α-induced disruption of claudin-5, zonula occludens-1, and vascular endothelial-cadherin in HRMEC. These actions are mainly mediated by sphingosine-1-phosphate receptor 1 modulation. Finally, siponimod prevented the progression of suture-induced corneal neovascularization in albino rabbits. In conclusion, the effects of siponimod on various processes known to be involved in angiogenesis support its therapeutic potential in disorders associated with ocular neovascularization. SIGNIFICANCE STATEMENT: Siponimod is an extensively characterized sphingosine-1-phosphate receptor modulator already approved for the treatment of multiple sclerosis. It inhibited retinal endothelial cell migration, potentiated endothelial barrier function, protected against tumor necrosis factor alpha-induced barrier disruption, and also inhibited suture-induced corneal neovascularization in rabbits. These results support its use for a novel therapeutic indication in the management of ocular neovascular diseases.
Collapse
Affiliation(s)
- Rasha A Alshaikh
- School of Pharmacy (R.A.A., K.B.R., C.W.), SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy (K.B.R.), and Department of Pharmacology and Therapeutics (C.W.), University College Cork, Cork, Ireland; Department of Ophthalmology (R.G.E.Z.) and Department of Anatomy and Embryology (R.A.S.E.D.), Faculty of Medicine, Ain Shams University, Cairo, Egypt; and Department of Anatomy and Embryology, Faculty of Medicine, Newgiza University (R.A.S.E.D.)
| | - Rania Gamal Eldin Zaki
- School of Pharmacy (R.A.A., K.B.R., C.W.), SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy (K.B.R.), and Department of Pharmacology and Therapeutics (C.W.), University College Cork, Cork, Ireland; Department of Ophthalmology (R.G.E.Z.) and Department of Anatomy and Embryology (R.A.S.E.D.), Faculty of Medicine, Ain Shams University, Cairo, Egypt; and Department of Anatomy and Embryology, Faculty of Medicine, Newgiza University (R.A.S.E.D.)
| | - Rania A Salah El Din
- School of Pharmacy (R.A.A., K.B.R., C.W.), SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy (K.B.R.), and Department of Pharmacology and Therapeutics (C.W.), University College Cork, Cork, Ireland; Department of Ophthalmology (R.G.E.Z.) and Department of Anatomy and Embryology (R.A.S.E.D.), Faculty of Medicine, Ain Shams University, Cairo, Egypt; and Department of Anatomy and Embryology, Faculty of Medicine, Newgiza University (R.A.S.E.D.)
| | - Katie B Ryan
- School of Pharmacy (R.A.A., K.B.R., C.W.), SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy (K.B.R.), and Department of Pharmacology and Therapeutics (C.W.), University College Cork, Cork, Ireland; Department of Ophthalmology (R.G.E.Z.) and Department of Anatomy and Embryology (R.A.S.E.D.), Faculty of Medicine, Ain Shams University, Cairo, Egypt; and Department of Anatomy and Embryology, Faculty of Medicine, Newgiza University (R.A.S.E.D.)
| | - Christian Waeber
- School of Pharmacy (R.A.A., K.B.R., C.W.), SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy (K.B.R.), and Department of Pharmacology and Therapeutics (C.W.), University College Cork, Cork, Ireland; Department of Ophthalmology (R.G.E.Z.) and Department of Anatomy and Embryology (R.A.S.E.D.), Faculty of Medicine, Ain Shams University, Cairo, Egypt; and Department of Anatomy and Embryology, Faculty of Medicine, Newgiza University (R.A.S.E.D.)
| |
Collapse
|
8
|
Hach T, Shakeri-Nejad K, Bigaud M, Dahlke F, de Micco M, Petricoul O, Graham G, Piani-Meier D, Turrini R, Brinkmann V, Nicoletti F. Rationale for Use of Sphingosine-1-Phosphate Receptor Modulators in COVID-19 Patients: Overview of Scientific Evidence. J Interferon Cytokine Res 2022. [DOI: 10.1089/jir.2022.0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Thomas Hach
- Patient Engagement, Novartis Pharma AG, Basel, Switzerland
| | - Kasra Shakeri-Nejad
- Department of Clinical Pharmacology; Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Marc Bigaud
- Department of Autoimmunity, Transplantation & Inflammation; Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Frank Dahlke
- Patient Engagement, Novartis Pharma AG, Basel, Switzerland
| | | | - Olivier Petricoul
- Department of Neuroscience; Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Gordon Graham
- Patient Engagement, Novartis Pharma AG, Basel, Switzerland
| | | | - Renato Turrini
- Department of Autoimmunity, Transplantation & Inflammation; Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University Sapienza of Rome, Rome, Italy
- Department of Molecular Neuropharmacology, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
9
|
Sphingosine-1-Phosphate Alleviates Irradiation Induced Salivary Gland Hypofunction through Preserving Endothelial Cells and Resident Macrophages. Antioxidants (Basel) 2022; 11:antiox11102050. [PMID: 36290773 PMCID: PMC9598384 DOI: 10.3390/antiox11102050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/25/2022] Open
Abstract
Radiotherapy for head-and-neck cancers frequently causes long-term hypofunction of salivary glands that severely compromises quality of life and is difficult to treat. Here, we studied effects and mechanisms of Sphingosine-1-phosphate (S1P), a versatile signaling sphingolipid, in preventing irreversible dry mouth caused by radiotherapy. Mouse submandibular glands (SMGs) were irradiated with or without intra-SMG S1P pretreatment. The saliva flow rate was measured following pilocarpine stimulation. The expression of genes related to S1P signaling and radiation damage was examined by flow cytometry, immunohistochemistry, quantitative RT-PCR, Western blotting, and/or single-cell RNA-sequencing. S1P pretreatment ameliorated irradiation-induced salivary dysfunction in mice through a decrease in irradiation-induced oxidative stress and consequent apoptosis and cellular senescence, which is related to the enhancement of Nrf2-regulated anti-oxidative response. In mouse SMGs, endothelial cells and resident macrophages are the major cells capable of producing S1P and expressing the pro-regenerative S1P receptor S1pr1. Both mouse SMGs and human endothelial cells are protected from irradiation damage by S1P pretreatment, likely through the S1pr1/Akt/eNOS axis. Moreover, intra-SMG-injected S1P did not affect the growth and radiosensitivity of head-and-neck cancer in a mouse model. These data indicate that S1P signaling pathway is a promising target for alleviating irradiation-induced salivary gland hypofunction.
Collapse
|
10
|
Adipose Tissue Development Relies on Coordinated Extracellular Matrix Remodeling, Angiogenesis, and Adipogenesis. Biomedicines 2022; 10:biomedicines10092227. [PMID: 36140327 PMCID: PMC9496222 DOI: 10.3390/biomedicines10092227] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/24/2022] Open
Abstract
Despite developing prenatally, the adipose tissue is unique in its ability to undergo drastic growth even after reaching its mature size. This development and subsequent maintenance rely on the proper coordination between the vascular niche and the adipose compartment. In this review, the process of adipose tissue development is broken down to explain (1) the ultrastructural matrix remodeling that is undertaken during simultaneous adipogenesis and angiogenesis, (2) the paracrine crosstalk involved during adipose development, (3) the mechanical regulators involved in adipose growth, and (4) the proteolytic and paracrine oversight for matrix remodeling during adipose development. It is crucial to gain a better understanding of the complex relationships that exist between adipose tissue and the vasculature during tissue development to provide insights into the pathological tissue expansion of obesity and to develop improved soft-tissue reconstruction techniques.
Collapse
|
11
|
Alshaikh RA, Ryan KB, Waeber C. Sphingosine 1-phosphate, a potential target in neovascular retinal disease. Br J Ophthalmol 2022; 106:1187-1195. [PMID: 33962970 DOI: 10.1136/bjophthalmol-2021-319115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/17/2021] [Accepted: 04/23/2021] [Indexed: 06/12/2023]
Abstract
Neovascular ocular diseases (such as age-related macular degeneration, diabetic retinopathy and retinal vein occlusion) are characterised by common pathological processes that contribute to disease progression. These include angiogenesis, oedema, inflammation, cell death and fibrosis. Currently available therapies target the effects of vascular endothelial growth factor (VEGF), the main mediator of pathological angiogenesis. Unfortunately, VEGF blockers are expensive biological therapeutics that necessitate frequent intravitreal administration and are associated with multiple adverse effects. Thus, alternative treatment options associated with fewer side effects are required for disease management. This review introduces sphingosine 1-phosphate (S1P) as a potential pharmacological target for the treatment of neovascular ocular pathologies. S1P is a sphingolipid mediator that controls cellular growth, differentiation, survival and death. S1P actions are mediated by five G protein-coupled receptors (S1P1-5 receptors) which are abundantly expressed in all retinal and subretinal structures. The action of S1P on S1P1 receptors can reduce angiogenesis, increase endothelium integrity, reduce photoreceptor apoptosis and protect the retina against neurodegeneration. Conversely, S1P2 receptor signalling can increase neovascularisation, disrupt endothelial junctions, stimulate VEGF release, and induce retinal cell apoptosis and degeneration of neural retina. The aim of this review is to thoroughly discuss the role of S1P and its different receptor subtypes in angiogenesis, inflammation, apoptosis and fibrosis in order to determine which of these S1P-mediated processes may be targeted therapeutically.
Collapse
Affiliation(s)
- Rasha A Alshaikh
- School of Pharmacy, University College Cork, Cork, Ireland
- Department of Pharmaceutical Technology, Tanta University, Tanta, Egypt
| | - Katie B Ryan
- School of Pharmacy, University College Cork, Cork, Ireland
- SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork, Ireland
| | - Christian Waeber
- School of Pharmacy, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| |
Collapse
|
12
|
Decoding Functional High-Density Lipoprotein Particle Surfaceome Interactions. Int J Mol Sci 2022; 23:ijms23169506. [PMID: 36012766 PMCID: PMC9409371 DOI: 10.3390/ijms23169506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/10/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
High-density lipoprotein (HDL) is a mixture of complex particles mediating reverse cholesterol transport (RCT) and several cytoprotective activities. Despite its relevance for human health, many aspects of HDL-mediated lipid trafficking and cellular signaling remain elusive at the molecular level. During HDL’s journey throughout the body, its functions are mediated through interactions with cell surface receptors on different cell types. To characterize and better understand the functional interplay between HDL particles and tissue, we analyzed the surfaceome-residing receptor neighborhoods with which HDL potentially interacts. We applied a combination of chemoproteomic technologies including automated cell surface capturing (auto-CSC) and HATRIC-based ligand–receptor capturing (HATRIC-LRC) on four different cellular model systems mimicking tissues relevant for RCT. The surfaceome analysis of EA.hy926, HEPG2, foam cells, and human aortic endothelial cells (HAECs) revealed the main currently known HDL receptor scavenger receptor B1 (SCRB1), as well as 155 shared cell surface receptors representing potential HDL interaction candidates. Since vascular endothelial growth factor A (VEGF-A) was recently found as a regulatory factor of transendothelial transport of HDL, we next analyzed the VEGF-modulated surfaceome of HAEC using the auto-CSC technology. VEGF-A treatment led to the remodeling of the surfaceome of HAEC cells, including the previously reported higher surfaceome abundance of SCRB1. In total, 165 additional receptors were found on HAEC upon VEGF-A treatment representing SCRB1 co-regulated receptors potentially involved in HDL function. Using the HATRIC-LRC technology on human endothelial cells, we specifically aimed for the identification of other bona fide (co-)receptors of HDL beyond SCRB1. HATRIC-LRC enabled, next to SCRB1, the identification of the receptor tyrosine-protein kinase Mer (MERTK). Through RNA interference, we revealed its contribution to endothelial HDL binding and uptake. Furthermore, subsequent proximity ligation assays (PLAs) demonstrated the spatial vicinity of MERTK and SCRB1 on the endothelial cell surface. The data shown provide direct evidence for a complex and dynamic HDL receptome and that receptor nanoscale organization may influence binding and uptake of HDL.
Collapse
|
13
|
Velagapudi S, Rohrer L, Poti F, Feuerborn R, Perisa D, Wang D, Panteloglou G, Potapenko A, Yalcinkaya M, Hülsmeier AJ, Hesse B, Lukasz A, Liu M, Parks JS, Christoffersen C, Stoffel M, Simoni M, Nofer JR, von Eckardstein A. Apolipoprotein M and Sphingosine-1-Phosphate Receptor 1 Promote the Transendothelial Transport of High-Density Lipoprotein. Arterioscler Thromb Vasc Biol 2021; 41:e468-e479. [PMID: 34407633 PMCID: PMC8458249 DOI: 10.1161/atvbaha.121.316725] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective: ApoM enriches S1P (sphingosine-1-phosphate) within HDL (high-density lipoproteins) and facilitates the activation of the S1P1 (S1P receptor type 1) by S1P, thereby preserving endothelial barrier function. Many protective functions exerted by HDL in extravascular tissues raise the question of how S1P regulates transendothelial HDL transport. Approach and Results: HDL were isolated from plasma of wild-type mice, Apom knockout mice, human apoM transgenic mice or humans and radioiodinated to trace its binding, association, and transport by bovine or human aortic endothelial cells. We also compared the transport of fluorescently-labeled HDL or Evans Blue, which labels albumin, from the tail vein into the peritoneal cavity of apoE-haploinsufficient mice with (apoE-haploinsufficient mice with endothelium-specific knockin of S1P1) or without (control mice, ie, apoE-haploinsufficient mice without endothelium-specific knockin of S1P1) endothelium-specific knockin of S1P1. The binding, association, and transport of HDL from Apom knockout mice and human apoM-depleted HDL by bovine aortic endothelial cells was significantly lower than that of HDL from wild-type mice and human apoM-containing HDL, respectively. The binding, uptake, and transport of 125I-HDL by human aortic endothelial cells was increased by an S1P1 agonist but decreased by an S1P1 inhibitor. Silencing of SR-BI (scavenger receptor BI) abrogated the stimulation of 125I-HDL transport by the S1P1 agonist. Compared with control mice, that is, apoE-haploinsufficient mice without endothelium-specific knockin of S1P1, apoE-haploinsufficient mice with endothelium-specific knockin of S1P1 showed decreased transport of Evans Blue but increased transport of HDL from blood into the peritoneal cavity and SR-BI expression in the aortal endothelium. Conclusions: ApoM and S1P1 promote transendothelial HDL transport. Their opposite effect on transendothelial transport of albumin and HDL indicates that HDL passes endothelial barriers by specific mechanisms rather than passive filtration.
Collapse
Affiliation(s)
- Srividya Velagapudi
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Francesco Poti
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, Italy (F.P.)
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy (F.P., M. Simoni, J.-R.N.)
| | - Renate Feuerborn
- Central Laboratory Facility, University Hospital of Münster, Germany (R.F., J.-R.N.)
| | - Damir Perisa
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Dongdong Wang
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Grigorios Panteloglou
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Anton Potapenko
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Mustafa Yalcinkaya
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Andreas J Hülsmeier
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| | - Bettina Hesse
- Department of Medicine D, Division of General Internal Medicine, Nephrology, and Rheumatology, University Hospital Münster, Germany (B.H., A.L.)
| | - Alexander Lukasz
- Department of Medicine D, Division of General Internal Medicine, Nephrology, and Rheumatology, University Hospital Münster, Germany (B.H., A.L.)
| | - Mingxia Liu
- Department of Internal Medicine/Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (M.L., J.S.P.)
| | - John S Parks
- Department of Internal Medicine/Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (M.L., J.S.P.)
| | - Christina Christoffersen
- Department of Biomedical Science, University of Copenhagen, Denmark (C.C.)
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark (C.C.)
| | - Markus Stoffel
- Institute of Molecular Health Sciences, ETH Zurich, Switzerland (M. Stoffel)
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy (F.P., M. Simoni, J.-R.N.)
| | - Jerzy-Roch Nofer
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy (F.P., M. Simoni, J.-R.N.)
- Central Laboratory Facility, University Hospital of Münster, Germany (R.F., J.-R.N.)
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (J.-R.N.)
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Switzerland (S.V., L.R., D.P., D.W., G.P., A.P., M.Y., A.J.H., A.v.E.)
| |
Collapse
|
14
|
Goel K, Beatman EL, Egersdorf N, Scruggs A, Cao D, Berdyshev EV, Schweitzer KS, Petrache I. Sphingosine 1 Phosphate (S1P) Receptor 1 Is Decreased in Human Lung Microvascular Endothelial Cells of Smokers and Mediates S1P Effect on Autophagy. Cells 2021; 10:cells10051200. [PMID: 34068927 PMCID: PMC8156252 DOI: 10.3390/cells10051200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 01/26/2023] Open
Abstract
Destruction of alveoli by apoptosis induced by cigarette smoke (CS) is a major driver of emphysema pathogenesis. However, when compared to cells isolated from non-smokers, primary human lung microvascular endothelial cells (HLMVECs) isolated from chronic smokers are more resilient when exposed to apoptosis-inducing ceramide. Whether this adaptation restores homeostasis is unknown. To better understand the phenotype of HLMVEC in smokers, we interrogated a major pro-survival pathway supported by sphingosine-1-phosphate (S1P) signaling via S1P receptor 1 (S1P1). Primary HLMVECs from lungs of non-smoker or smoker donors were isolated and studied in culture for up to five passages. S1P1 mRNA and protein abundance were significantly decreased in HLMVECs from smokers compared to non-smokers. S1P1 was also decreased in situ in lungs of mice chronically exposed to CS. Levels of S1P1 expression tended to correlate with those of autophagy markers, and increasing S1P (via S1P lyase knockdown with siRNA) stimulated baseline macroautophagy with lysosomal degradation. In turn, loss of S1P1 (siRNA) inhibited these effects of S1P on HLMVECs autophagy. These findings suggest that the anti-apoptotic phenotype of HLMVECs from smokers may be maladaptive, since it is associated with decreased S1P1 expression that may impair their autophagic response to S1P.
Collapse
Affiliation(s)
- Khushboo Goel
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA;
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - Erica L. Beatman
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - Nicholas Egersdorf
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - April Scruggs
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - Danting Cao
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - Evgeny V. Berdyshev
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
| | - Kelly S. Schweitzer
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Irina Petrache
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA; (E.L.B.); (N.E.); (A.S.); (D.C.); (E.V.B.); (K.S.S.)
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: ; Tel.: +1-303-398-1355
| |
Collapse
|
15
|
Qi Y, Wang W, Song Z, Aji G, Liu XT, Xia P. Role of Sphingosine Kinase in Type 2 Diabetes Mellitus. Front Endocrinol (Lausanne) 2021; 11:627076. [PMID: 33633691 PMCID: PMC7899982 DOI: 10.3389/fendo.2020.627076] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
Sphingolipids are a class of essential lipids, functioning as both cell membrane constituents and signaling messengers. In the sphingolipid metabolic network, ceramides serve as the central hub that is hydrolyzed to sphingosine, followed by phosphorylation to sphingosine 1-phosphate (S1P) by sphingosine kinase (SphK). SphK is regarded as a "switch" of the sphingolipid rheostat, as it catalyzes the conversion of ceramide/sphingosine to S1P, which often exhibit opposing biological roles in the cell. Besides, SphK is an important signaling enzyme that has been implicated in the regulation of a wide variety of biological functions. In recent years, an increasing body of evidence has suggested a critical role of SphK in type 2 diabetes mellitus (T2D), although a certain level of controversy remains. Herein, we review recent findings related to SphK in the field of T2D research with a focus on peripheral insulin resistance and pancreatic β-cell failure. It is expected that a comprehensive understanding of the role of SphK and the associated sphingolipids in T2D will help to identify druggable targets for future anti-diabetes therapy.
Collapse
Affiliation(s)
- Yanfei Qi
- Lipid Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Sydney, NSW, Australia
| | - Wei Wang
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ziyu Song
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gulibositan Aji
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Tracy Liu
- Lipid Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Sydney, NSW, Australia
| | - Pu Xia
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Balaji Ragunathrao VA, Anwar M, Akhter MZ, Chavez A, Mao DY, Natarajan V, Lakshmikanthan S, Chrzanowska-Wodnicka M, Dudek AZ, Claesson-Welsh L, Kitajewski JK, Wary KK, Malik AB, Mehta D. Sphingosine-1-Phosphate Receptor 1 Activity Promotes Tumor Growth by Amplifying VEGF-VEGFR2 Angiogenic Signaling. Cell Rep 2020; 29:3472-3487.e4. [PMID: 31825830 PMCID: PMC6927555 DOI: 10.1016/j.celrep.2019.11.036] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/06/2019] [Accepted: 11/07/2019] [Indexed: 12/24/2022] Open
Abstract
The vascular endothelial growth factor-A (VEGF-A)-VEGFR2 pathway drives tumor vascularization by activating proangiogenic signaling in endothelial cells (ECs). Here, we show that EC-sphingosine-1-phosphate receptor 1 (S1PR1) amplifies VEGFR2-mediated angiogenic signaling to enhance tumor growth. We show that cancer cells induce S1PR1 activity in ECs, and thereby, conditional deletion of S1PR1 in ECs (EC-S1pr1−/− mice) impairs tumor vascularization and growth. Mechanistically, we show that S1PR1 engages the heterotrimeric G-protein Gi, which amplifies VEGF-VEGFR2 signaling due to an increase in the activity of the tyrosine kinase c-Abl1. c-Abl1, by phosphorylating VEGFR2 at tyrosine-951, prolongs VEGFR2 retention on the plasmalemma to sustain Rac1 activity and EC migration. Thus, S1PR1 or VEGFR2 antagonists, alone or in combination, reverse the tumor growth in control mice to the level seen in EC-S1pr1−/− mice. Our findings suggest that blocking S1PR1 activity in ECs has the potential to suppress tumor growth by preventing amplification of VEGF-VEGFR2 signaling. Vijay Avin et al. demonstrate an essential role of endothelial cell (EC)-S1PR1 signaling in amplifying VEGFR2-mediated tumor growth. S1PR1 by Gi and c-Abl1 phosphorylates VEGFR2 at Y951, which retains VEGFR2 at EC plasmalemma, thus enabling EC migration, tumor angiogenesis, and growth.
Collapse
Affiliation(s)
- Vijay Avin Balaji Ragunathrao
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Mumtaz Anwar
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Md Zahid Akhter
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Alejandra Chavez
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - De Yu Mao
- Department of Physiology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Viswanathan Natarajan
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA; Department of Medicine, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | | | - Arkadiusz Z Dudek
- Department of Medicine, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Jan K Kitajewski
- Department of Physiology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Kishore K Wary
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Asrar B Malik
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Dolly Mehta
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
17
|
Zheng Z, Chopp M, Chen J. Multifaceted roles of pericytes in central nervous system homeostasis and disease. J Cereb Blood Flow Metab 2020; 40:1381-1401. [PMID: 32208803 PMCID: PMC7308511 DOI: 10.1177/0271678x20911331] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pericytes, the mural cells surrounding microcirculation, are gaining increasing attention for their roles in health and disease of the central nervous system (CNS). As an essential part of the neurovascular unit (NVU), pericytes are actively engaged in interactions with neighboring cells and work in synergy with them to maintain homeostasis of the CNS, such as maintaining the blood-brain barrier (BBB), regulating cerebral blood flow (CBF) and the glymphatic system as well as mediating immune responses. However, the dysfunction of pericytes may contribute to the progression of various pathologies. In this review, we discuss: (1) origin of pericytes and different pericyte markers; (2) interactions of pericytes with endothelial cells (ECs), astrocytes, microglia, oligodendrocytes, and neurons; (3) physiological roles of pericytes in the CNS; (4) effects of pericytes in different CNS diseases; (5) relationship of pericytes with extracellular vesicles (EVs) and microRNAs (miRs); (6) recent advances in pericytes studies and future perspective.
Collapse
Affiliation(s)
- Zhitong Zheng
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Physics, Oakland University, Rochester, MI, USA
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
18
|
Abstract
Sphingosine-1-phosphate (S1P) can regulate several physiological and pathological processes. S1P signaling via its cell surface receptor S1PR1 has been shown to enhance tumorigenesis and stimulate growth, expansion, angiogenesis, metastasis, and survival of cancer cells. S1PR1-mediated tumorigenesis is supported and amplified by activation of downstream effectors including STAT3, interleukin-6, and NF-κB networks. S1PR1 signaling can also trigger various other signaling pathways involved in carcinogenesis including activation of PI3K/AKT, MAPK/ERK1/2, Rac, and PKC/Ca, as well as suppression of cyclic adenosine monophosphate (cAMP). It also induces immunological tolerance in the tumor microenvironment, while the immunosuppressive function of S1PR1 can also lead to the generation of pre-metastatic niches. Some tumor cells upregulate S1PR1 signaling pathways, which leads to drug resistant cancer cells, mainly through activation of STAT3. This signaling pathway is also implicated in some inflammatory conditions leading to the instigation of inflammation-driven cancers. Furthermore, it can also increase survival via induction of anti-apoptotic pathways, for instance, in breast cancer cells. Therefore, S1PR1 and its signaling pathways can be considered as potential anti-tumor therapeutic targets, alone or in combination therapies. Given the oncogenic nature of S1PR1 and its distribution in a variety of cancer cell types along with its targeting advantages over other molecules of this family, S1PR1 should be considered a favorable target in therapeutic approaches to cancer. This review describes the role of S1PR1 in cancer development and progression, specifically addressing breast cancer, glioma, and hematopoietic malignancies. We also discuss the potential use of S1P signaling modulators as therapeutic targets in cancer therapy.
Collapse
|
19
|
Riboni L, Abdel Hadi L, Navone SE, Guarnaccia L, Campanella R, Marfia G. Sphingosine-1-Phosphate in the Tumor Microenvironment: A Signaling Hub Regulating Cancer Hallmarks. Cells 2020; 9:E337. [PMID: 32024090 PMCID: PMC7072483 DOI: 10.3390/cells9020337] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
As a key hub of malignant properties, the cancer microenvironment plays a crucial role intimately connected to tumor properties. Accumulating evidence supports that the lysophospholipid sphingosine-1-phosphate acts as a key signal in the cancer extracellular milieu. In this review, we have a particular focus on glioblastoma, representative of a highly aggressive and deleterious neoplasm in humans. First, we highlight recent advances and emerging concepts for how tumor cells and different recruited normal cells contribute to the sphingosine-1-phosphate enrichment in the cancer microenvironment. Then, we describe and discuss how sphingosine-1-phosphate signaling contributes to favor cancer hallmarks including enhancement of proliferation, stemness, invasion, death resistance, angiogenesis, immune evasion and, possibly, aberrant metabolism. We also discuss the potential of how sphingosine-1-phosphate control mechanisms are coordinated across distinct cancer microenvironments. Further progress in understanding the role of S1P signaling in cancer will depend crucially on increasing knowledge of its participation in the tumor microenvironment.
Collapse
Affiliation(s)
- Laura Riboni
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Loubna Abdel Hadi
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
- Department of Clinical Sciences and Community Health, University of Milan, 20100 Milan, Italy
| | - Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| |
Collapse
|
20
|
Kovilakath A, Cowart LA. Sphingolipid Mediators of Myocardial Pathology. J Lipid Atheroscler 2020; 9:23-49. [PMID: 32821720 PMCID: PMC7379069 DOI: 10.12997/jla.2020.9.1.23] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 09/25/2019] [Accepted: 10/09/2019] [Indexed: 12/15/2022] Open
Abstract
Cardiomyopathy is the leading cause of mortality worldwide. While the causes of cardiomyopathy continue to be elucidated, current evidence suggests that aberrant bioactive lipid signaling plays a crucial role as a component of cardiac pathophysiology. Sphingolipids have been implicated in the pathophysiology of cardiovascular disease, as they regulate numerous cellular processes that occur in primary and secondary cardiomyopathies. Experimental evidence gathered over the last few decades from both in vitro and in vivo model systems indicates that inhibitors of sphingolipid synthesis attenuate a variety of cardiomyopathic symptoms. In this review, we focus on various cardiomyopathies in which sphingolipids have been implicated and the potential therapeutic benefits that could be gained by targeting sphingolipid metabolism.
Collapse
Affiliation(s)
- Anna Kovilakath
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - L. Ashley Cowart
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
- Hunter Holmes McGuire Veteran's Affairs Medical Center, Richmond, VA, USA
| |
Collapse
|
21
|
S1P/S1P Receptor Signaling in Neuromuscolar Disorders. Int J Mol Sci 2019; 20:ijms20246364. [PMID: 31861214 PMCID: PMC6941007 DOI: 10.3390/ijms20246364] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/06/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022] Open
Abstract
The bioactive sphingolipid metabolite, sphingosine 1-phosphate (S1P), and the signaling pathways triggered by its binding to specific G protein-coupled receptors play a critical regulatory role in many pathophysiological processes, including skeletal muscle and nervous system degeneration. The signaling transduced by S1P binding appears to be much more complex than previously thought, with important implications for clinical applications and for personalized medicine. In particular, the understanding of S1P/S1P receptor signaling functions in specific compartmentalized locations of the cell is worthy of being better investigated, because in various circumstances it might be crucial for the development or/and the progression of neuromuscular diseases, such as Charcot-Marie-Tooth disease, myasthenia gravis, and Duchenne muscular dystrophy.
Collapse
|
22
|
Panta CR, Ruisanchez É, Móré D, Dancs PT, Balogh A, Fülöp Á, Kerék M, Proia RL, Offermanns S, Tigyi GJ, Benyó Z. Sphingosine-1-Phosphate Enhances α 1-Adrenergic Vasoconstriction via S1P2-G 12/13-ROCK Mediated Signaling. Int J Mol Sci 2019; 20:ijms20246361. [PMID: 31861195 PMCID: PMC6941080 DOI: 10.3390/ijms20246361] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/04/2019] [Accepted: 12/13/2019] [Indexed: 01/21/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) has been implicated recently in the physiology and pathology of the cardiovascular system including regulation of vascular tone. Pilot experiments showed that the vasoconstrictor effect of S1P was enhanced markedly in the presence of phenylephrine (PE). Based on this observation, we hypothesized that S1P might modulate α1-adrenergic vasoactivity. In murine aortas, a 20-minute exposure to S1P but not to its vehicle increased the Emax and decreased the EC50 of PE-induced contractions indicating a hyperreactivity to α1-adrenergic stimulation. The potentiating effect of S1P disappeared in S1P2 but not in S1P3 receptor-deficient vessels. In addition, smooth muscle specific conditional deletion of G12/13 proteins or pharmacological inhibition of the Rho-associated protein kinase (ROCK) by Y-27632 or fasudil abolished the effect of S1P on α1-adrenergic vasoconstriction. Unexpectedly, PE-induced contractions remained enhanced markedly as late as three hours after S1P-exposure in wild-type (WT) and S1P3 KO but not in S1P2 KO vessels. In conclusion, the S1P–S1P2–G12/13–ROCK signaling pathway appears to have a major influence on α1-adrenergic vasoactivity. This cooperativity might lead to sustained vasoconstriction when increased sympathetic tone is accompanied by increased S1P production as it occurs during acute coronary syndrome and stroke.
Collapse
Affiliation(s)
- Cecília R. Panta
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
- Correspondence: (C.R.P.); (Z.B.)
| | - Éva Ruisanchez
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Dorottya Móré
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Péter T. Dancs
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Andrea Balogh
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Ágnes Fülöp
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Margit Kerék
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Richard L. Proia
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, MD 20892, USA;
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany;
| | - Gábor J. Tigyi
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
- Correspondence: (C.R.P.); (Z.B.)
| |
Collapse
|
23
|
Sunitha P, Raju R, Sajil CK, Abhinand CS, Nair AS, Oommen OV, Sugunan VS, Sudhakaran PR. Temporal VEGFA responsive genes in HUVECs: Gene signatures and potential ligands/receptors fine-tuning angiogenesis. J Cell Commun Signal 2019; 13:561-571. [PMID: 31840205 DOI: 10.1007/s12079-019-00541-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
Vascular Endothelial Growth Factor-A (VEGFA) signaling is crucial to the cellular processes involved in angiogenesis. Previously, we assembled a network of molecular reactions induced by VEGFA in human umbilical vein endothelial cell populations. Considering transcriptome as a read-out of the transcriptional and epigenomic regulatory network, we now present an analysis of VEGFA-induced temporal transcriptome datasets from 6 non-synchronized studies. From these datasets, applying a confidence criterion, a set of early VEGFA-responsive signature genes were derived and evaluated for their co-expression potential with respect to multiple cancer gene expression datasets. Further, inclusive of a set of ligand-receptor pairs, a list of ligand and receptor signaling systems that potentially fine-tune the endothelial cell functions subsequent to VEGFA signaling were also derived. We believe that a number of these signaling systems would concurrently and/or hierarchically fine-tune the signaling network of endothelial cell populations towards the processes associated with angiogenesis through autocrine, paracrine, juxtacrine, and matricrine modes. By further analysis of published literature on VEGFA signaling, we also present an improved update-version of our previous VEGFA signaling network model in endothelial cells as a platform for analysis of cross-talk with these signaling systems.
Collapse
Affiliation(s)
- P Sunitha
- Department of Computational Biology & Bioinformatics, Research Centre, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - Rajesh Raju
- Rajiv Gandhi Centre for Biotechnology, KINFRA Campus, Thiruvananthapuram, Kerala, 695585, India.
| | - C K Sajil
- Department of Computational Biology & Bioinformatics, Research Centre, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - C S Abhinand
- Department of Computational Biology & Bioinformatics, Research Centre, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - Achuthsankar S Nair
- Department of Computational Biology & Bioinformatics, Research Centre, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - Oommen V Oommen
- Department of Computational Biology & Bioinformatics, Research Centre, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - V S Sugunan
- Department of Computational Biology & Bioinformatics, Research Centre, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - P R Sudhakaran
- Department of Computational Biology & Bioinformatics, Research Centre, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India.
| |
Collapse
|
24
|
Park KM, Wang JW, Yoo YM, Choi MJ, Hwang KC, Jeung EB, Jeong YW, Hwang WS. Sphingosine-1-phosphate (S1P) analog phytosphingosine-1-phosphate (P1P) improves the in vitro maturation efficiency of porcine oocytes via regulation of oxidative stress and apoptosis. Mol Reprod Dev 2019; 86:1705-1719. [PMID: 31490595 DOI: 10.1002/mrd.23264] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/03/2019] [Indexed: 12/11/2022]
Abstract
Phytosphingosine-1-phosphate (P1P) is a signaling sphingolipid that regulates various physiological activities. However, little is known about the effect of P1P in the context of reproduction. Thus, we aimed to investigate the influence of P1P on oocyte maturation during porcine in vitro maturation (IVM). Here, we report the expression of S1PR1-3 among P1P receptors (S1PR1-4) in cumulus cells and oocytes. When P1P was administered at concentrations of 10, 50, 100, and 1,000 nM during IVM, the metaphase II rate was significantly increased in the 1,000 nM (1 μM) P1P treatment group. Maturation rate improvement by P1P supplementation was observed only in the presence of epidermal growth factor (EGF). Oocytes under the influence of P1P showed decreased intracellular reactive oxygen species levels but no significant differences in glutathione levels. In our molecular studies, P1P treatment upregulated gene expression involved in cumulus expansion (Has2 and EGF), antioxidant enzymes (SOD3 and Cat), and developmental competence (Oct4) while activating extracellular signal-regulated kinase1/2 and Akt signaling. P1P treatment also influenced oocyte survival by shifting the ratio of Bcl-2 to Bax while inactivating JNK signaling. We further demonstrated that oocytes matured with P1P displayed significantly higher developmental competence (cleavage and blastocyst [BL] formation rate) and greater BL quality (total cell number and the ratio of apoptotic cells) when activated via parthenogenetic activation (PA) and in vitro fertilization. Despite the low levels of endogenous P1P found in animals, exogenous P1P influenced animal reproduction, as shown by increased porcine oocyte maturation as well as preimplantation embryo development. This study and its findings are potentially relevant for both human and animal-assisted reproduction.
Collapse
Affiliation(s)
- Kyu-Mi Park
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Jae Woong Wang
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Yeong-Min Yoo
- Laboratory of Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | | | - Kyu Chan Hwang
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Yeon Woo Jeong
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Woo Suk Hwang
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| |
Collapse
|
25
|
Volpi C, Orabona C, Macchiarulo A, Bianchi R, Puccetti P, Grohmann U. Preclinical discovery and development of fingolimod for the treatment of multiple sclerosis. Expert Opin Drug Discov 2019; 14:1199-1212. [PMID: 31389262 DOI: 10.1080/17460441.2019.1646244] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Introduction: Fingolimod, the first oral disease-modifying treatment (DMT) in multiple sclerosis (MS), is a sphingosine 1-phosphate receptor (S1PR) ligand. Approved in 2010, fingolimod has been extensively studied and has been credited with several mechanisms of actions that contribute to its efficacy in MS, among which is the regulation of lymphocyte circulation between the central nervous system and the periphery. Concerns about toxicity, off-target effects, and real-life performance have been raised over time in post-marketing studies of such that next-generation sphingosine-1 phosphate receptor ligands are now being developed. Areas covered: Herein, the authors expand upon previous systematic reviews obtained via PubMed and through their expert opinion on fingolimod use in clinical practice. Long-term data including long-term efficacy, safety, tolerability, and management especially within growing DMT options and pre-treatment constellation in MS patients are discussed, together with the results of an increased understanding of the chemistry underlying the structure-activity relationship. Expert opinion: Despite the limitations illustrated in this article, fingolimod still constitutes a paradigm shift in MS treatment. However, although immunomodulation via S1PRs on lymphocytes has represented a major breakthrough in the clinical management of MS, modifying the evolution of progressive MS will likely require the development of approaches other than merely targeting S1PRs.
Collapse
Affiliation(s)
- Claudia Volpi
- Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Ciriana Orabona
- Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia , Perugia , Italy
| | - Roberta Bianchi
- Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia , Perugia , Italy
| |
Collapse
|
26
|
Iwazu Y, Muto S, Ioka T, Watanabe Y, Iwazu K, Kusano E, Nagata D. Multiple Sclerosis Drug Fingolimod Induces Thrombotic Microangiopathy in Deoxycorticosterone Acetate/Salt Hypertension. Hypertension 2019; 72:776-784. [PMID: 30354753 DOI: 10.1161/hypertensionaha.117.10655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We examined whether fingolimod (FTY720), an S1PR (sphingosine-1-phosphate receptor) modulator, has beneficial or harmful effects on mineralocorticoid/salt-induced renal injury. Uninephrectomized rats on 0.9% NaCl/0.3% KCl drinking solution were randomly divided into control, control+FTY720, deoxycorticosterone acetate (DOCA), and DOCA+FTY720 groups and administered vehicle, vehicle+FTY720, DOCA+vehicle, and DOCA+FTY720 for 4 weeks, respectively. Only the DOCA+FTY720 group had reduced survival rates and showed hemolysis because of intravascular mechanical fragmentation of erythrocytes and thrombocytopenia. Both the DOCA+FTY720 and DOCA groups developed malignant hypertension, which was more severe in the DOCA+FTY720 group. In the DOCA+FTY720 group only, thrombotic microangiopathy involving severe renal arteriole endothelial cell injury was observed and was characterized by fibrinoid necrosis and onion-skin lesions in arterioles. There were fewer circulating endothelial progenitor cells in the DOCA+FTY720 group but more in the DOCA group compared with the control group. Expression levels of VEGF (vascular endothelial growth factor), S1PR1, and S1PR3 in renal arteriole endothelial cells were significantly greater in the DOCA+FTY720 and DOCA groups compared with the control group, with levels being similar between the 2 groups. Expression levels of endothelial nitric oxide synthase in renal arteriole endothelial cells were significantly lower in the DOCA+FTY720 group only. The control+FTY720 group showed reduced circulating endothelial progenitor cells but no significant functional or pathological changes in kidneys or changes in blood pressure. Exposure of uninephrectomized rats to DOCA/salt+FTY720 for 4 weeks induced renal arteriolar endothelial cell injury, resulting in the development of thrombotic microangiopathy. Consideration of this possibility is recommended when prescribing FTY720.
Collapse
Affiliation(s)
- Yoshitaka Iwazu
- From the Department of Nephrology (Y.I., S.M., T.I., Y.W., K.I., E.K., D.N.).,Clinical Laboratory Medicine (Y.I.), Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Shigeaki Muto
- From the Department of Nephrology (Y.I., S.M., T.I., Y.W., K.I., E.K., D.N.)
| | - Takashi Ioka
- From the Department of Nephrology (Y.I., S.M., T.I., Y.W., K.I., E.K., D.N.)
| | - Yuko Watanabe
- From the Department of Nephrology (Y.I., S.M., T.I., Y.W., K.I., E.K., D.N.)
| | - Kana Iwazu
- From the Department of Nephrology (Y.I., S.M., T.I., Y.W., K.I., E.K., D.N.)
| | - Eiji Kusano
- From the Department of Nephrology (Y.I., S.M., T.I., Y.W., K.I., E.K., D.N.)
| | - Daisuke Nagata
- From the Department of Nephrology (Y.I., S.M., T.I., Y.W., K.I., E.K., D.N.)
| |
Collapse
|
27
|
Smith CJ, Allard DE, Wang Y, Howard JF, Montgomery SA, Su MA. IL-10 Paradoxically Promotes Autoimmune Neuropathy through S1PR1-Dependent CD4 + T Cell Migration. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:1580-1592. [PMID: 29367208 PMCID: PMC5821539 DOI: 10.4049/jimmunol.1701280] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/28/2017] [Indexed: 01/13/2023]
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP) is a debilitating condition caused by autoimmune demyelination of peripheral nerves. CIDP is associated with increased IL-10, a cytokine with well-described anti-inflammatory effects. However, the role of IL-10 in CIDP is unclear. In this study, we demonstrate that IL-10 paradoxically exacerbates autoimmunity against peripheral nerves. In IL-10-deficient mice, protection from neuropathy was associated with an accrual of highly activated CD4+ T cells in draining lymph nodes and absence of infiltrating immune cells in peripheral nerves. Accumulated CD4+ T cells in draining lymph nodes of IL-10-deficient mice expressed lower sphingosine-1-phosphate receptor 1 (S1pr1), a protein important in lymphocyte egress. Additionally, IL-10 stimulation in vitro induced S1pr1 expression in lymph node cells in a STAT3-dependent manner. Together, these results delineate a novel mechanism in which IL-10-induced STAT3 increases S1pr1 expression and CD4+ T cell migration to accelerate T cell-mediated destruction of peripheral nerves.
Collapse
Affiliation(s)
- Collin-Jamal Smith
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Denise E Allard
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Yan Wang
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - James F Howard
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Stephanie A Montgomery
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Maureen A Su
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599;
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
28
|
Du X, Li W, Du G, Cho H, Yu M, Fang Q, Lee LP, Fang J. Droplet Array-Based 3D Coculture System for High-Throughput Tumor Angiogenesis Assay. Anal Chem 2018; 90:3253-3261. [DOI: 10.1021/acs.analchem.7b04772] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Xiaohui Du
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, China
| | - Wanming Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, China
| | - Guansheng Du
- Institute of Microanalytical System, Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Hansang Cho
- Department of Bioengineering, University of California, Berkeley, California 94720, United States
| | - Min Yu
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, China
| | - Qun Fang
- Institute of Microanalytical System, Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Luke P. Lee
- Department of Bioengineering, University of California, Berkeley, California 94720, United States
| | - Jin Fang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, China
| |
Collapse
|
29
|
Combined use of bone marrow-derived mesenchymal stromal cells (BM-MSCs) and platelet rich plasma (PRP) stimulates proliferation and differentiation of myoblasts in vitro: new therapeutic perspectives for skeletal muscle repair/regeneration. Cell Tissue Res 2018; 372:549-570. [PMID: 29404727 DOI: 10.1007/s00441-018-2792-3] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 01/06/2018] [Indexed: 10/18/2022]
Abstract
Satellite cell-mediated skeletal muscle repair/regeneration is compromised in cases of extended damage. Bone marrow mesenchymal stromal cells (BM-MSCs) hold promise for muscle healing but some criticisms hamper their clinical application, including the need to avoid animal serum contamination for expansion and the scarce survival after transplant. In this context, platelet-rich plasma (PRP) could offer advantages. Here, we compare the effects of PRP or standard culture media on C2C12 myoblast, satellite cell and BM-MSC viability, survival, proliferation and myogenic differentiation and evaluate PRP/BM-MSC combination effects in promoting myogenic differentiation. PRP induced an increase of mitochondrial activity and Ki67 expression comparable or even greater than that elicited by standard media and promoted AKT signaling activation in myoblasts and BM-MSCs and Notch-1 pathway activation in BM-MSCs. It stimulated MyoD, myogenin, α-sarcomeric actin and MMP-2 expression in myoblasts and satellite cell activation. Notably, PRP/BM-MSC combination was more effective than PRP alone. We found that BM-MSCs influenced myoblast responses through a paracrine activation of AKT signaling, contributing to shed light on BM-MSC action mechanisms. Our results suggest that PRP represents a good serum substitute for BM-MSC manipulation in vitro and could be beneficial towards transplanted cells in vivo. Moreover, it might influence muscle resident progenitors' fate, thus favoring the endogenous repair/regeneration mechanisms. Finally, within the limitations of an in vitro experimentation, this study provides an experimental background for considering the PRP/BM-MSC combination as a potential therapeutic tool for skeletal muscle damage, combining the beneficial effects of BM-MSCs and PRP on muscle tissue, while potentiating BM-MSC functionality.
Collapse
|
30
|
Korbecki J, Gutowska I, Kojder I, Jeżewski D, Goschorska M, Łukomska A, Lubkowska A, Chlubek D, Baranowska-Bosiacka I. New extracellular factors in glioblastoma multiforme development: neurotensin, growth differentiation factor-15, sphingosine-1-phosphate and cytomegalovirus infection. Oncotarget 2018; 9:7219-7270. [PMID: 29467963 PMCID: PMC5805549 DOI: 10.18632/oncotarget.24102] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 01/02/2018] [Indexed: 11/25/2022] Open
Abstract
Recent years have seen considerable progress in understanding the biochemistry of cancer. For example, more significance is now assigned to the tumor microenvironment, especially with regard to intercellular signaling in the tumor niche which depends on many factors secreted by tumor cells. In addition, great progress has been made in understanding the influence of factors such as neurotensin, growth differentiation factor-15 (GDF-15), sphingosine-1-phosphate (S1P), and infection with cytomegalovirus (CMV) on the 'hallmarks of cancer' in glioblastoma multiforme. Therefore, in the present work we describe the influence of these factors on the proliferation and apoptosis of neoplastic cells, cancer stem cells, angiogenesis, migration and invasion, and cancer immune evasion in a glioblastoma multiforme tumor. In particular, we discuss the effect of neurotensin, GDF-15, S1P (including the drug FTY720), and infection with CMV on tumor-associated macrophages (TAM), microglial cells, neutrophil and regulatory T cells (Treg), on the tumor microenvironment. In order to better understand the role of the aforementioned factors in tumoral processes, we outline the latest models of intratumoral heterogeneity in glioblastoma multiforme. Based on the most recent reports, we discuss the problems of multi-drug therapy in treating glioblastoma multiforme.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland.,Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biała, 43-309 Bielsko-Biała, Poland
| | - Izabela Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Ireneusz Kojder
- Department of Applied Neurocognitivistics, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland.,Department of Neurosurgery, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Dariusz Jeżewski
- Department of Applied Neurocognitivistics, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland.,Department of Neurosurgery, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Marta Goschorska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Agnieszka Łukomska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Anna Lubkowska
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| |
Collapse
|
31
|
Apolipoprotein M Inhibits Angiogenic and Inflammatory Response by Sphingosine 1-Phosphate on Retinal Pigment Epithelium Cells. Int J Mol Sci 2017; 19:ijms19010112. [PMID: 29301231 PMCID: PMC5796061 DOI: 10.3390/ijms19010112] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/20/2017] [Accepted: 12/28/2017] [Indexed: 01/04/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is a potent lipid mediator that modulates inflammatory responses and proangiogenic factors. It has been suggested that S1P upregulates choroidal neovascularization (CNV) and may be deeply involved in the pathogenesis of exudative age-related macular degeneration (AMD). Recent studies have suggested that apolipoprotein M (ApoM), a carrier protein for S1P, modulates the biological properties of S1P in the pathogenesis of atherosclerosis. However, the role of ApoM/S1P in AMD has not been explored. We investigated the effect of S1P on proangiogenic factors in human retinal pigment epithelium (RPE) cell lines in vitro. S1P promoted the expression of vascular endothelial growth factor in RPE cells. Hypoxia inducible factor-1α expression was also upregulated. These S1P-induced enhancements in growth factors and chemotactic cytokines in RPE cells were significantly inhibited by ApoM treatment. Additionally, in vivo experiments using a laser-induced CNV murine model demonstrated that intravitreal ApoM injection significantly reduced the progression of CNV formation. Although the detailed mechanisms remain to be elucidated, the present results provide a novel potential therapeutic target for AMD, and demonstrate a suppressive role for ApoM and S1P in the pathology of CNV progression.
Collapse
|
32
|
Abou Daher A, El Jalkh T, Eid AA, Fornoni A, Marples B, Zeidan YH. Translational Aspects of Sphingolipid Metabolism in Renal Disorders. Int J Mol Sci 2017; 18:E2528. [PMID: 29186855 PMCID: PMC5751131 DOI: 10.3390/ijms18122528] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 12/13/2022] Open
Abstract
Sphingolipids, long thought to be passive components of biological membranes with merely a structural role, have proved throughout the past decade to be major players in the pathogenesis of many human diseases. The study and characterization of several genetic disorders like Fabry's and Tay Sachs, where sphingolipid metabolism is disrupted, leading to a systemic array of clinical symptoms, have indeed helped elucidate and appreciate the importance of sphingolipids and their metabolites as active signaling molecules. In addition to being involved in dynamic cellular processes like apoptosis, senescence and differentiation, sphingolipids are implicated in critical physiological functions such as immune responses and pathophysiological conditions like inflammation and insulin resistance. Interestingly, the kidneys are among the most sensitive organ systems to sphingolipid alterations, rendering these molecules and the enzymes involved in their metabolism, promising therapeutic targets for numerous nephropathic complications that stand behind podocyte injury and renal failure.
Collapse
Affiliation(s)
- Alaa Abou Daher
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Tatiana El Jalkh
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Alessia Fornoni
- Department of Medicine, Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miami, FL 33136, USA.
| | - Brian Marples
- Department of Radiation Oncology, Miller School of Medicine/Sylvester Cancer Center, University of Miami, Miami, FL 33136, USA.
| | - Youssef H Zeidan
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon.
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon.
| |
Collapse
|
33
|
Smith P, O'Sullivan C, Gergely P. Sphingosine 1-Phosphate Signaling and Its Pharmacological Modulation in Allogeneic Hematopoietic Stem Cell Transplantation. Int J Mol Sci 2017; 18:ijms18102027. [PMID: 28934113 PMCID: PMC5666709 DOI: 10.3390/ijms18102027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/10/2017] [Accepted: 09/18/2017] [Indexed: 12/16/2022] Open
Abstract
Allogeneic haemopoietic stem cell transplantation (HSCT) is increasingly used to treat haematological malignant diseases via the graft-versus-leukaemia (GvL) or graft-versus-tumour effects. Although improvements in infectious disease prophylaxis, immunosuppressive treatments, supportive care, and molecular based tissue typing have contributed to enhanced outcomes, acute graft-versus-host disease and other transplant related complications still contribute to high mortality and significantly limit the more widespread use of HSCT. Sphingosine 1-phosphate (S1P) is a zwitterionic lysophospholipid that has been implicated as a crucial signaling regulator in many physiological and pathophysiological processes including multiple cell types such as macrophages, dendritic cells, T cells, T regulatory cells and endothelial cells. Recent data suggested important roles for S1P signaling in engraftment, graft-versus-host disease (GvHD), GvL and other processes that occur during and after HSCT. Based on such data, pharmacological intervention via S1P modulation may have the potential to improve patient outcome by regulating GvHD and enhancing engraftment while permitting effective GvL.
Collapse
Affiliation(s)
- Philip Smith
- Novartis Institutes for BioMedical Research, WSJ-386, CH-4002 Basel, Switzerland.
| | - Catherine O'Sullivan
- Novartis Institutes for BioMedical Research, WSJ-386, CH-4002 Basel, Switzerland.
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.
| | - Peter Gergely
- Novartis Institutes for BioMedical Research, WSJ-386, CH-4002 Basel, Switzerland.
| |
Collapse
|
34
|
Identification of the S100 fused-type protein hornerin as a regulator of tumor vascularity. Nat Commun 2017; 8:552. [PMID: 28916756 PMCID: PMC5601918 DOI: 10.1038/s41467-017-00488-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 07/04/2017] [Indexed: 02/03/2023] Open
Abstract
Sustained angiogenesis is essential for the development of solid tumors and metastatic disease. Disruption of signaling pathways that govern tumor vascularity provide a potential avenue to thwart cancer progression. Through phage display-based functional proteomics, immunohistochemical analysis of human pancreatic ductal carcinoma (PDAC) specimens, and in vitro validation, we reveal that hornerin, an S100 fused-type protein, is highly expressed on pancreatic tumor endothelium in a vascular endothelial growth factor (VEGF)-independent manner. Murine-specific hornerin knockdown in PDAC xenografts results in tumor vessels with decreased radii and tortuosity. Hornerin knockdown tumors have significantly reduced leakiness, increased oxygenation, and greater apoptosis. Additionally, these tumors show a significant reduction in growth, a response that is further heightened when therapeutic inhibition of VEGF receptor 2 (VEGFR2) is utilized in combination with hornerin knockdown. These results indicate that hornerin is highly expressed in pancreatic tumor endothelium and alters tumor vessel parameters through a VEGF-independent mechanism.Angiogenesis is essential for solid tumor progression. Here, the authors interrogate the proteome of pancreatic cancer endothelium via phage display and identify hornerin as a critical protein whose expression is essential to maintain the pancreatic cancer vasculature through a VEGF-independent mechanism.
Collapse
|
35
|
Sphingosine kinase 1/sphingosine-1-phosphate (S1P)/S1P receptor axis is involved in ovarian cancer angiogenesis. Oncotarget 2017; 8:74947-74961. [PMID: 29088837 PMCID: PMC5650392 DOI: 10.18632/oncotarget.20471] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/25/2017] [Indexed: 12/15/2022] Open
Abstract
Sphingosine kinase (SphK)/sphingosine-1-phosphate (S1P)/S1P receptor (S1PR) signaling pathway has been implicated in a variety of pathological processes of ovarian cancer. However, the function of this axis in ovarian cancer angiogenesis remains incompletely defined. Here we provided the first evidence that SphK1/S1P/S1PR1/3 pathway played key roles in ovarian cancer angiogenesis. The expression level of SphK1, but not SphK2, was closely correlated with the microvascular density (MVD) of ovarian cancer tissue. In vitro, the angiogenic potential and angiogenic factor secretion of ovarian cancer cells could be attenuated by SphK1, but not SphK2, blockage and were restored by the addition of S1P. Moreover, in these cells, we found S1P stimulation induced the angiogenic factor secretion via S1PR1 and S1PR3, but not S1PR2. Furthermore, inhibition of S1PR1/3, but not S1PR2, attenuated the angiogenic potential and angiogenic factor secretion of the cells. in vivo, blockage of SphK or S1PR1/3 could attenuate ovarian cancer angiogenesis and inhibit angiogenic factor expression in mouse models. Collectively, the current study showed a novel role of SphK1/S1P/S1PR1/3 axis within the ovarian cancer, suggesting a new target to block ovarian cancer angiogenesis.
Collapse
|
36
|
Rojas-Canales D, Penko D, Myo Min KK, Parham KA, Peiris H, Haberberger RV, Pitson SM, Drogemuller C, Keating DJ, Grey ST, Coates PT, Bonder CS, Jessup CF. Local Sphingosine Kinase 1 Activity Improves Islet Transplantation. Diabetes 2017; 66:1301-1311. [PMID: 28174291 DOI: 10.2337/db16-0837] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 02/02/2017] [Indexed: 11/13/2022]
Abstract
Pancreatic islet transplantation is a promising clinical treatment for type 1 diabetes, but success is limited by extensive β-cell death in the immediate posttransplant period and impaired islet function in the longer term. Following transplantation, appropriate vascular remodeling is crucial to ensure the survival and function of engrafted islets. The sphingosine kinase (SK) pathway is an important regulator of vascular beds, but its role in the survival and function of transplanted islets is unknown. We observed that donor islets from mice deficient in SK1 (Sphk1 knockout) contain a reduced number of resident intraislet vascular endothelial cells. Furthermore, we demonstrate that the main product of SK1, sphingosine-1-phosphate, controls the migration of intraislet endothelial cells in vitro. We reveal in vivo that Sphk1 knockout islets have an impaired ability to cure diabetes compared with wild-type controls. Thus, SK1-deficient islets not only contain fewer resident vascular cells that participate in revascularization, but likely also a reduced ability to recruit new vessels into the transplanted islet. Together, our data suggest that SK1 is important for islet revascularization following transplantation and represents a novel clinical target for improving transplant outcomes.
Collapse
Affiliation(s)
- Darling Rojas-Canales
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Central Northern Adelaide Renal and Transplantation Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Daniella Penko
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Central Northern Adelaide Renal and Transplantation Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Kay K Myo Min
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Kate A Parham
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Heshan Peiris
- Department of Human Physiology, Flinders University, Bedford Park, Australia
- Centre for Neuroscience, Flinders University, Bedford Park, Australia
| | | | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Chris Drogemuller
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Central Northern Adelaide Renal and Transplantation Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Damien J Keating
- Department of Human Physiology, Flinders University, Bedford Park, Australia
- Centre for Neuroscience, Flinders University, Bedford Park, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Shane T Grey
- Garvan Medical Institute, Darlinghurst, Australia
| | - Patrick T Coates
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Central Northern Adelaide Renal and Transplantation Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Claudine S Bonder
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Claire F Jessup
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Centre for Neuroscience, Flinders University, Bedford Park, Australia
- Department of Anatomy & Histology, Flinders University, Bedford Park, Australia
| |
Collapse
|
37
|
4-Hydroxynonenal Contributes to Angiogenesis through a Redox-Dependent Sphingolipid Pathway: Prevention by Hydralazine Derivatives. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9172741. [PMID: 28479957 PMCID: PMC5396448 DOI: 10.1155/2017/9172741] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/01/2017] [Indexed: 12/23/2022]
Abstract
The neovascularization of atherosclerotic lesions is involved in plaque development and may contribute to intraplaque hemorrhage and plaque fragilization and rupture. Among the various proangiogenic agents involved in the neovascularization process, proatherogenic oxidized LDLs (oxLDLs) contribute to the formation of tubes via the generation of sphingosine 1-phosphate (S1P), a major mitogenic and proangiogenic sphingolipid mediator. In this study, we investigated whether 4-hydroxynonenal (4-HNE), an aldehydic lipid oxidation product abundantly present in oxLDLs, contributes to their proangiogenic properties. Immunofluorescence analysis of human atherosclerotic lesions from carotid endarterectomy showed the colocalization of HNE-adducts with CD31, a marker of endothelial cells, suggesting a close relationship between 4-HNE and neovessel formation. In vitro, low 4-HNE concentration (0.5-1 µM) elicited the formation of tubes by human microvascular endothelial cells (HMEC-1), whereas higher concentrations were not angiogenic. The formation of tubes by 4-HNE involved the generation of reactive oxygen species and the activation of the sphingolipid pathway, namely, the neutral type 2 sphingomyelinase and sphingosine kinase-1 (nSMase2/SK-1) pathway, indicating a role for S1P in the angiogenic signaling of 4-HNE. Carbonyl scavengers hydralazine and bisvanillyl-hydralazone inhibited the nSMase2/SK1 pathway activation and the formation of tubes on Matrigel® evoked by 4-HNE. Altogether, these results emphasize the role of 4-HNE in the angiogenic effect of oxLDLs and point out the potential interest of pharmacological carbonyl scavengers to prevent the neovascularization process.
Collapse
|
38
|
Patmanathan SN, Wang W, Yap LF, Herr DR, Paterson IC. Mechanisms of sphingosine 1-phosphate receptor signalling in cancer. Cell Signal 2017; 34:66-75. [PMID: 28302566 DOI: 10.1016/j.cellsig.2017.03.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/11/2017] [Accepted: 03/11/2017] [Indexed: 12/12/2022]
Abstract
S1P is a small bioactive lipid which exerts its effects following binding to a family of five G protein-coupled receptors, known as S1P1-5. Following receptor activation, multiple signalling cascades are activated, allowing S1P to regulate a range of cellular processes, such as proliferation, apoptosis, migration and angiogenesis. There is strong evidence implicating the involvement of S1P receptors (S1PRs) in cancer progression and the oncogenic effects of S1P can result from alterations in the expression of one or more of the S1PRs and/or the enzymes that regulate the levels of S1P. However, cooperativity between the individual S1PRs, functional interactions with receptor tyrosine kinases and the sub-cellular localisation of the S1PRs within tumour cells also appear to play a role in mediating the effects of S1PR signalling during carcinogenesis. Here we review what is known regarding the role of individual S1PRs in cancer and discuss the recent evidence to suggest cross-talk between the S1PRs and other cellular signalling pathways in cancer. We will also discuss the therapeutic potential of targeting the S1PRs and their downstream signalling pathways for the treatment of cancer.
Collapse
Affiliation(s)
- Sathya Narayanan Patmanathan
- Department of Oral and Craniofacial Sciences, Oral Cancer Research & Coordinating Centre, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Wei Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Lee Fah Yap
- Department of Oral and Craniofacial Sciences, Oral Cancer Research & Coordinating Centre, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Ian C Paterson
- Department of Oral and Craniofacial Sciences, Oral Cancer Research & Coordinating Centre, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
39
|
Tölle M, Klöckl L, Wiedon A, Zidek W, van der Giet M, Schuchardt M. Regulation of endothelial nitric oxide synthase activation in endothelial cells by S1P1 and S1P3. Biochem Biophys Res Commun 2016; 476:627-634. [DOI: 10.1016/j.bbrc.2016.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/01/2016] [Indexed: 12/16/2022]
|
40
|
Park-Windhol C, D'Amore PA. Disorders of Vascular Permeability. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 11:251-81. [PMID: 26907525 DOI: 10.1146/annurev-pathol-012615-044506] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The endothelial barrier maintains vascular and tissue homeostasis and modulates many physiological processes, such as angiogenesis. Vascular barrier integrity can be disrupted by a variety of soluble permeability factors, and changes in barrier function can exacerbate tissue damage during disease progression. Understanding endothelial barrier function is critical for vascular homeostasis. Many of the signaling pathways promoting vascular permeability can also be triggered during disease, resulting in prolonged or uncontrolled vascular leak. It is believed that recovery of the normal vasculature requires diminishing this hyperpermeable state. Although the molecular mechanisms governing vascular leak have been studied over the last few decades, recent advances have identified new therapeutic targets that have begun to show preclinical and clinical promise. These approaches have been successfully applied to an increasing number of disease conditions. New perspectives regarding how vascular leak impacts the progression of various diseases are highlighted in this review.
Collapse
Affiliation(s)
- Cindy Park-Windhol
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts 02114; , .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Patricia A D'Amore
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts 02114; , .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115.,Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
41
|
Azimi A. Hypothesis: Fingolimod explores new horizons in treatment of lymphoma. Med Hypotheses 2016; 86:71-5. [PMID: 26804601 DOI: 10.1016/j.mehy.2015.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/24/2015] [Accepted: 11/28/2015] [Indexed: 11/18/2022]
Abstract
Lymphomas (Hodgkin's (10%) and non-Hodgkin's (90%) lymphomas) are a group of blood cell tumors arising from lymphocytes and lymphadenopathy is the most common primary presentation of the disease. In non-Hodgkin lymphomas, the prognosis is worse. As the disease initiates, neoplastic cells may spread to and involve other lymph nodes and extra nodal regions. The disease is staged based on desperation of neoplastic cells and the prognosis highly depends on the stage of the disease at the time of diagnosis. Fingolimod is an immunomodulating drug, approved for treating relapsing forms of multiple sclerosis. Fingolimod impairs migration of lymphocytes from lymph nodes and it is hypothesized that Fingolimod could alleviate and decrease disease burden of lymphoma as it sequesters malignant cells within involved lymph nodes and it decelerates progression of the disease, increases efficacy of other treatment options and it is synergistic with anti-VEGF medications, it is an anti-metastatic, anti-inflammatory, cytostatic/cytotoxic agent and it boosts function of immune system in deterioration of neoplastic cells. Therefore, the agent can be used not only to treat lymphoma, but also to control and prevent relapse of the disease in those who are remitted.
Collapse
Affiliation(s)
- Arsalan Azimi
- Histomorphometry & Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
42
|
Sphingosine-1-Phosphate/Sphingosine-1-Phosphate Receptor 2 Axis Can Promote Mouse and Human Primary Mast Cell Angiogenic Potential through Upregulation of Vascular Endothelial Growth Factor-A and Matrix Metalloproteinase-2. Mediators Inflamm 2016; 2016:1503206. [PMID: 26884643 PMCID: PMC4738939 DOI: 10.1155/2016/1503206] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/30/2015] [Accepted: 12/21/2015] [Indexed: 12/18/2022] Open
Abstract
Mast cells (MC) are present in most vascularized tissues around the vasculature likely exerting immunomodulatory functions. Endowed with diverse mediators, resident MC represent first-line fine-tuners of local microenvironment. Sphingosine-1-phosphate (S1P) functions as a pluripotent signaling sphingolipid metabolite in health and disease. S1P formation occurs at low levels in resting MC and is upregulated upon activation. Its export can result in type 2 S1P receptor- (S1PR2-) mediated stimulation of MC, further fueling inflammation. However, the role of S1PR2 ligation in proangiogenic vascular endothelial growth factor- (VEGF-) A and matrix metalloproteinase- (MMP-) 2 release from MC is unknown. Using a preclinical MC-dependent model of acute allergic responses and in vitro stimulated primary mouse bone marrow-derived MC (BMMC) or human primary skin MC, we report that S1P signaling resulted in substantial amount of VEGF-A release. Similar experiments using S1pr2-deficient mice or BMMC or selective S1P receptor agonists or antagonists demonstrated that S1P/S1PR2 ligation on MC is important for VEGF-A secretion. Further, we show that S1P stimulation triggered transcriptional upregulation of VEGF-A and MMP-2 mRNA in human but not in mouse MC. S1P exposure also triggered MMP-2 secretion from human MC. These studies identify a novel proangiogenic axis encompassing MC/S1P/S1PR2 likely relevant to inflammation.
Collapse
|
43
|
Durham JT, Dulmovits BM, Cronk SM, Sheets AR, Herman IM. Pericyte chemomechanics and the angiogenic switch: insights into the pathogenesis of proliferative diabetic retinopathy? Invest Ophthalmol Vis Sci 2015; 56:3441-59. [PMID: 26030100 DOI: 10.1167/iovs.14-13945] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
PURPOSE To establish the regulatory roles that pericytes have in coordinating retinal endothelial cell (EC) growth and angiogenic potential. METHODS Pericytes were derived from donor diabetic (DHuRP) or normal (NHuRP) human retinae, and characterized using vascular markers, coculture, contraction, morphogenesis, and proliferation assays. To investigate capillary "cross-talk," pericyte-endothelial coculture growth, and connexin-43 (Cx43) expression assays were performed. Paracrine effects were examined via treating EC with pericyte-derived conditioned media (CM) in proliferation, angiogenesis, and angiocrine assays. The effects of sphingosine 1-phosphate (S1P) were assessed using receptor antagonists. RESULTS The DHuRP exhibit unique proliferative and morphologic properties, reflecting distinctive cytoskeletal and isoactin expression patterns. Unlike NHuRP, DHuRP are unable to sustain EC growth arrest in coculture and display reduced Cx43 expression. Further, CM from DHuRP (DPCM) markedly stimulates EC proliferation and tube formation. Treatment with S1P receptor antagonists mitigates DPCM growth-promotion in EC and S1P-mediated pericyte contraction. Angiocrine assays on normal and diabetic pericyte secretomes reveal factors involved in angiogenic control, inflammation, and metabolism. CONCLUSIONS Effects from the diabetic microenvironment appear sustainable in cell culture: pericytes derived from diabetic donor eyes seemingly possess a "metabolic memory" in vitro, which may be linked to original donor health status. Diabetes- and pericyte-dependent effects on EC growth and angiogenesis may reflect alterations in bioactive lipid, angiocrine, and chemomechanical signaling. Altogether, our results suggest that diabetes alters pericyte contractile phenotype and cytoskeletal signaling, which ultimately may serve as a key, initiating event required for retinal endothelial reproliferation, angiogenic activation, and the pathological neovascularization accompanying proliferative diabetic retinopathy.
Collapse
|
44
|
Williams PA, Stilhano RS, To VP, Tran L, Wong K, Silva EA. Hypoxia augments outgrowth endothelial cell (OEC) sprouting and directed migration in response to sphingosine-1-phosphate (S1P). PLoS One 2015; 10:e0123437. [PMID: 25875493 PMCID: PMC4398361 DOI: 10.1371/journal.pone.0123437] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 02/20/2015] [Indexed: 12/26/2022] Open
Abstract
Therapeutic angiogenesis provides a promising approach to treat ischemic cardiovascular diseases through the delivery of proangiogenic cells and/or molecules. Outgrowth endothelial cells (OECs) are vascular progenitor cells that are especially suited for therapeutic strategies given their ease of noninvasive isolation from umbilical cord or adult peripheral blood and their potent ability to enhance tissue neovascularization. These cells are recruited to sites of vascular injury or tissue ischemia and directly incorporate within native vascular endothelium to participate in neovessel formation. A better understanding of how OEC activity may be boosted under hypoxia with external stimulation by proangiogenic molecules remains a challenge to improving their therapeutic potential. While vascular endothelial growth factor (VEGF) is widely established as a critical factor for initiating angiogenesis, sphingosine-1-phosphate (S1P), a bioactive lysophospholipid, has recently gained great enthusiasm as a potential mediator in neovascularization strategies. This study tests the hypothesis that hypoxia and the presence of VEGF impact the angiogenic response of OECs to S1P stimulation in vitro. We found that hypoxia altered the dynamically regulated S1P receptor 1 (S1PR1) expression on OECs in the presence of S1P (1.0 μM) and/or VEGF (1.3 nM). The combined stimuli of S1P and VEGF together promoted OEC angiogenic activity as assessed by proliferation, wound healing, 3D sprouting, and directed migration under both normoxia and hypoxia. Hypoxia substantially augmented the response to S1P alone, resulting in ~6.5-fold and ~25-fold increases in sprouting and directed migration, respectively. Overall, this report highlights the importance of establishing hypoxic conditions in vitro when studying ischemia-related angiogenic strategies employing vascular progenitor cells.
Collapse
Affiliation(s)
- Priscilla A. Williams
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Roberta S. Stilhano
- Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | - Vivian P. To
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Lyndon Tran
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, California, United States of America
| | - Kevin Wong
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Eduardo A. Silva
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
45
|
Heo MJ, Kim YM, Koo JH, Yang YM, An J, Lee SK, Lee SJ, Kim KM, Park JW, Kim SG. microRNA-148a dysregulation discriminates poor prognosis of hepatocellular carcinoma in association with USP4 overexpression. Oncotarget 2015; 5:2792-806. [PMID: 24798342 PMCID: PMC4058045 DOI: 10.18632/oncotarget.1920] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is classified as a poor prognostic tumor, and becomes frequently aggressive. MicroRNAs emerge as key contributors to tumor progression. This study investigated whether miR-148a dysregulation differentiates poor prognosis of HCC, exploring new targets of miR-148a. miR-148a dysregulation discriminated not only the overall survival and recurrence free survival rates of HCC, but the microvascular invasion. In the human HCC samples, ubiquitin specific protease 4 (USP4) and sphingosine 1-phosphate receptor 1 (S1P1) were up-regulated as the new targets of miR-148a. USP4 and S1P1 were up-regulated in mesenchymal-type liver-tumor cells with miR-148a dysregulation, facilitating migration and proliferation of tumor cells. The inverse relationship between miR-148a and the identified targets was verified in a tumor xenograft model. In the analysis of human samples, the expression of USP4, but not S1P1, correlated with the decrease of miR-148a. In a heterotropic patient-derived HCC xenograft model, USP4 was also overexpressed in G1 and G2 tumors when miR-148a was dysregulated, reflecting the closer link between miR-148a and USP4 for a shift in the expansion phase of tumorgraft. In conclusion, miR-148a dysregulation affects the poor prognosis of HCC. Of the identified targets of miR-148a, USP4 overexpression may contribute to HCC progression towards more aggressive feature.
Collapse
Affiliation(s)
- Mi Jeong Heo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Rhee SH, Zhang P, Hunter K, Mama ST, Caraballo R, Holzberg AS, Seftel RH, Seftel AD, Echols KT, DiSanto ME. Pelvic organ prolapse is associated with alteration of sphingosine-1-phosphate/Rho-kinase signalling pathway in human vaginal wall. J OBSTET GYNAECOL 2015; 35:726-32. [DOI: 10.3109/01443615.2015.1004527] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
47
|
Li Q, Chen B, Zeng C, Fan A, Yuan Y, Guo X, Huang X, Huang Q. Differential activation of receptors and signal pathways upon stimulation by different doses of sphingosine-1-phosphate in endothelial cells. Exp Physiol 2014; 100:95-107. [PMID: 25557733 DOI: 10.1113/expphysiol.2014.082149] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 10/21/2014] [Indexed: 01/08/2023]
Abstract
NEW FINDINGS What is the central question of this study? Why do different doses of sphingosine-1-phosphate (S1P) induce distinct biological effects in endothelial cells? What is the main finding and its importance? S1P at physiological concentrations preserved endothelial barrier function by binding to S1P receptor 1, then triggering Ca(2+) release from endoplasmic reticulum through phosphoinositide phospholipase C and inositol triphosphate, and consequently strengthening tight junction and F-actin assembly through Rac1 activation. Excessive S1P induced endothelial malfunction by activating S1P receptor 2 and RhoA/ROCK pathway, causing F-actin and tight junction disorganisation. Extracellular Ca(2+) influx was involved in this process. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid in plasma, and its plasma concentration can be adjusted through a complex metabolic process. The alterations in S1P levels and the activation of receptors collaboratively regulate distinct biological effects. This study was performed to investigate comparatively the effect of different concentrations of S1P on endothelial barrier function and to explore the roles of S1P receptors (S1PRs), Rho GTPases and calcium in S1P-induced endothelial responses. Endothelial barrier function was studied using transendothelial electric resistance and a resistance meter in human umbilical vein endothelial cells. Specific agonists or antagonists were applied to control the activation of S1P receptors and the release of calcium from different cellular compartments. The results indicated that at physiological concentrations, S1P preserved endothelial barrier function by binding with S1PR1. The activation of S1PR1 triggered the release of intracellular Ca(2+) from the endoplasmic reticulum through the PI-phospholipase C and inositol trisphosphate pathways. Consequently, the Rho GTPase Rac1 was activated, strengthening the assembly of tight junction proteins and F-actin. However, excessive S1P induced endothelial barrier dysfunction by activating S1PR2 followed by the RhoA/RhoA kinase pathway, causing the disorganization of F-actin and the disassembly of the tight junction protein ZO-1. An influx of extracellular Ca(2+) was involved in this process. These data suggest that physiological and excessive amounts of S1P induce different responses in human umbilical vein endothelial cells; the activation of the 1PR1-PLC-IP3 R-Ca(2+) -Rac1 pathway governs the low-dose S1P-enhanced endothelial barrier integrity, and the activation of S1PR2-calcium influx-RhoA/ROCK dominates the high-dose S1P-induced endothelial monolayer hyperpermeability response.
Collapse
Affiliation(s)
- Qiang Li
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, 510515, PR China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Yu M, He P, Liu Y, He Y, Du Y, Wu M, Zhang G, Yang C, Gao F. Hyaluroan-regulated lymphatic permeability through S1P receptors is crucial for cancer metastasis. Med Oncol 2014; 32:381. [PMID: 25428387 DOI: 10.1007/s12032-014-0381-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 11/17/2014] [Indexed: 12/31/2022]
Abstract
Disruption of cancer lymphatic vessel barrier function occurs has been reported to involve in cancer lymphatic metastasis. Hyaluronan (HA), a major glycosaminoglycan component of the extracellular matrix, is associated with cancer metastasis. We investigated the effect of high/low molecular weight hyaluronan (HMW-HA/LMW-HA) on regulation of barrier function and tight junctions in cancer lymphatic endothelial cell (LEC) monolayer. Results showed that LMW-HA increased the permeability of cancer LEC monolayers and induced disruption of Zonula Occludens-1 (ZO-1)-mediated intercellular tight junction and actin stress fiber formation. HMW-HA treatment decreased permeability in cancer LEC monolayers and cortical actin ring formation. As reported, sphingosine 1-phosphate (S1P) receptors are involved in vascular integrity. After silencing of lymphatic vessel endothelial hyaluronan receptor (LYVE-1), upregulation of S1P receptors (S1P1 and S1P3) induced by HMW-HA/LMW-HA were inhibited, respectively. With S1P3 silenced, the disruption of ZO-1 as well as stress fiber formation and the ROCK1/RhoA signaling pathway induced by LMW-HA was not observed in cancer LEC. These results suggested that S1P receptors may play an important role in HMW-HA-/LMW-HA-mediated regulation of cancer lymphatic vessel integrity, which might be the initial step of cancer lymphatic metastasis and a useful intervention of cancer progression.
Collapse
Affiliation(s)
- Mengsi Yu
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sassoli C, Frati A, Tani A, Anderloni G, Pierucci F, Matteini F, Chellini F, Zecchi Orlandini S, Formigli L, Meacci E. Mesenchymal stromal cell secreted sphingosine 1-phosphate (S1P) exerts a stimulatory effect on skeletal myoblast proliferation. PLoS One 2014; 9:e108662. [PMID: 25264785 PMCID: PMC4181304 DOI: 10.1371/journal.pone.0108662] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/25/2014] [Indexed: 12/20/2022] Open
Abstract
Bone-marrow-derived mesenchymal stromal cells (MSCs) have the potential to significantly contribute to skeletal muscle healing through the secretion of paracrine factors that support proliferation and enhance participation of the endogenous muscle stem cells in the process of repair/regeneration. However, MSC-derived trophic molecules have been poorly characterized. The aim of this study was to investigate paracrine signaling effects of MSCs on skeletal myoblasts. It was found, using a biochemical and morphological approach that sphingosine 1-phosphate (S1P), a natural bioactive lipid exerting a broad range of muscle cell responses, is secreted by MSCs and represents an important factor by which these cells exert their stimulatory effects on C2C12 myoblast and satellite cell proliferation. Indeed, exposure to conditioned medium obtained from MSCs cultured in the presence of the selective sphingosine kinase inhibitor (iSK), blocked increased cell proliferation caused by the conditioned medium from untreated MSCs, and the addition of exogenous S1P in the conditioned medium from MSCs pre-treated with iSK further increased myoblast proliferation. Finally, we also demonstrated that the myoblast response to MSC-secreted vascular endothelial growth factor (VEGF) involves the release of S1P from C2C12 cells. Our data may have important implications in the optimization of cell-based strategies to promote skeletal muscle regeneration.
Collapse
Affiliation(s)
- Chiara Sassoli
- Department of Experimental and Clinical Medicine - Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Alessia Frati
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” - Unit of Biochemical Sciences and Molecular Biology, University of Florence, Florence, Italy
| | - Alessia Tani
- Department of Experimental and Clinical Medicine - Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Giulia Anderloni
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” - Unit of Biochemical Sciences and Molecular Biology, University of Florence, Florence, Italy
| | - Federica Pierucci
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” - Unit of Biochemical Sciences and Molecular Biology, University of Florence, Florence, Italy
| | - Francesca Matteini
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” - Unit of Biochemical Sciences and Molecular Biology, University of Florence, Florence, Italy
| | - Flaminia Chellini
- Department of Experimental and Clinical Medicine - Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Sandra Zecchi Orlandini
- Department of Experimental and Clinical Medicine - Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Lucia Formigli
- Department of Experimental and Clinical Medicine - Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Elisabetta Meacci
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” - Unit of Biochemical Sciences and Molecular Biology, University of Florence, Florence, Italy
- * E-mail:
| |
Collapse
|
50
|
Identification and characterization of a mirror-image oligonucleotide that binds and neutralizes sphingosine 1-phosphate, a central mediator of angiogenesis. Biochem J 2014; 462:153-62. [PMID: 24832383 PMCID: PMC4109837 DOI: 10.1042/bj20131422] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The sphingolipid S1P (sphingosine 1-phosphate) is known to be involved in a number of pathophysiological conditions such as cancer, autoimmune diseases and fibrosis. It acts extracellularly through a set of five G-protein-coupled receptors, but its intracellular actions are also well documented. Employing in vitro selection techniques, we identified an L-aptamer (Spiegelmer®) to S1P designated NOX-S93. The binding affinity of NOX-S93 to S1P had a Kd value of 4.3 nM. The Spiegelmer® shows equal binding to dihydro-S1P, but no cross-reactivity to the related lipids sphingosine, lysophosphatidic acid, ceramide, ceramide-1-phosphate or sphingosine phosphocholine. In stably transfected CHO (Chinese-hamster ovary) cell lines expressing the S1P receptors S1PR1 or S1PR3, NOX-S93 inhibits S1P-mediated β-arrestin recruitment and intracellular calcium release respectively, with IC50 values in the low nanomolar range. The pro-angiogenic activity of S1P, and of the growth factors VEGF-A (vascular endothelial growth factor-A), FGF-2 (fibroblast growth factor-2) and IGF-1 (insulin-like growth factor-1), was effectively blocked by NOX-S93 in a cellular angiogenesis assay employing primary human endothelial cells. These data provide further evidence for the relevance of extracellular S1P as a central mediator of angiogenesis, suggesting pharmacological S1P neutralization as a promising treatment alternative to current anti-angiogenesis approaches. The mirror-image aptamer NOX-S93, identified by in vitro selection to bind specifically to S1P, inhibits signalling at S1P receptors. NOX-S93 inhibits the pro-angiogenic activity of S1P and other growth factors, revealing S1P neutralization as a promising anti-angiogenesis approach.
Collapse
|