1
|
Schwalm S, Manaila R, Oftring A, Schaefer L, von Gunten S, Pfeilschifter J. The contribution of the sphingosine 1-phosphate signaling pathway to chronic kidney diseases: recent findings and new perspectives. Pflugers Arch 2024:10.1007/s00424-024-03029-5. [PMID: 39384640 DOI: 10.1007/s00424-024-03029-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/11/2024]
Abstract
Chronic kidney disease (CKD) is a multifactorial condition with diverse etiologies, such as diabetes mellitus, hypertension, and genetic disorders, often culminating in end-stage renal disease (ESRD). A hallmark of CKD progression is kidney fibrosis, characterized by the excessive accumulation of extracellular matrix components, for which there is currently no effective anti-fibrotic therapy. Recent literature highlights the critical role of sphingosine 1-phosphate (S1P) signaling in CKD pathogenesis and renal fibrosis. This review provides an in-depth analysis of the latest findings on S1P metabolism and signaling in renal fibrosis and in specific CKDs, including diabetic nephropathy (DN), lupus nephritis (LN), focal segmental glomerulosclerosis (FSGS), Fabry disease (FD), and IgA nephropathy (IgAN). Emerging studies underscore the therapeutic potential of modulating S1P signaling with receptor modulators and inhibitors, such as fingolimod (FTY720) and more selective agents like ozanimod and cenerimod. Additionally, the current knowledge about the effects of established kidney protective therapies such as glucocorticoids and SGLT2 and ACE inhibitors on S1P signaling will be summarized. Furthermore, the review highlights the potential role of S1P as a biomarker for disease progression in CKD models, particularly in Fabry disease and diabetic nephropathy. Advanced technologies, including spatial transcriptomics, are further refining our understanding of S1P's role within specific kidney compartments. Collectively, these insights emphasize the need for continued research into S1P signaling pathways as promising targets for CKD treatment strategies.
Collapse
Affiliation(s)
- Stephanie Schwalm
- Institut für Allgemeine Pharmakologie und Toxikologie, Goethe-Universität Frankfurt Am Main, Theodor-Stern-Kai 7, 60590, Frankfurt Am Main, Germany.
| | - Roxana Manaila
- Institut für Pharmakologie, Universität Bern, Inselspital, INO-F, CH-3011, Bern, Switzerland
| | - Anke Oftring
- Institut für Allgemeine Pharmakologie und Toxikologie, Goethe-Universität Frankfurt Am Main, Theodor-Stern-Kai 7, 60590, Frankfurt Am Main, Germany
| | - Liliana Schaefer
- Institut für Allgemeine Pharmakologie und Toxikologie, Goethe-Universität Frankfurt Am Main, Theodor-Stern-Kai 7, 60590, Frankfurt Am Main, Germany
| | - Stephan von Gunten
- Institut für Pharmakologie, Universität Bern, Inselspital, INO-F, CH-3011, Bern, Switzerland
| | - Josef Pfeilschifter
- Institut für Allgemeine Pharmakologie und Toxikologie, Goethe-Universität Frankfurt Am Main, Theodor-Stern-Kai 7, 60590, Frankfurt Am Main, Germany
| |
Collapse
|
2
|
Glueck M, Lucaciu A, Subburayalu J, Kestner RI, Pfeilschifter W, Vutukuri R, Pfeilschifter J. Atypical sphingosine-1-phosphate metabolites-biological implications of alkyl chain length. Pflugers Arch 2024:10.1007/s00424-024-03018-8. [PMID: 39297971 DOI: 10.1007/s00424-024-03018-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/21/2024]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive lipid signaling molecule with pleiotropic implications by both auto- and paracrine signaling. Signaling occurs by engaging five G protein-coupled receptors (S1P1-5) or intracellular pathways. While the extensively studied S1P with a chain length of 18 carbon atoms (d18:1 S1P) affects lymphocyte trafficking, immune cell survival and inflammatory responses, the biological implication of atypical S1Ps such as d16:1 or d20:1 remains elusive. As S1P lipids have far-reaching implications in health and disease states in mammalian organisms, the previous contrasting results may be attributed to differences in S1P's alkyl chain length. Current research is beginning to appreciate these less abundant atypical S1P moieties. This review provides an up-to-date foundation of recent findings on the biological implications of atypical S1P chain lengths and offers a perspective on future research endeavors on S1P alkyl chain length-influenced signaling and its implications for drug discovery.
Collapse
Affiliation(s)
- Melanie Glueck
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, Goethe University, Frankfurt am Main, 60596, Frankfurt, Germany
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Goethe University Hospital, 60528, Frankfurt Am Main, Germany
| | - Alexandra Lucaciu
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, Goethe University, Frankfurt am Main, 60596, Frankfurt, Germany
- Department of Neurology, University Hospital Frankfurt, Frankfurt, Goethe University, Frankfurt am Main, 60528, Frankfurt, Germany
| | - Julien Subburayalu
- Department of Internal Medicine, University Hospital Carl Gustav Carus TU Dresden, Fetscherstraße 74, 01307, Dresden, Saxony, Germany
- Center of Regenerative Therapies Dresden, TU Dresden, Fetscherstraße 74, 01307, Dresden, Saxony, Germany
| | - Roxane Isabelle Kestner
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, Goethe University, Frankfurt am Main, 60596, Frankfurt, Germany
- Department of Neurology, University Hospital Frankfurt, Frankfurt, Goethe University, Frankfurt am Main, 60528, Frankfurt, Germany
| | - Waltraud Pfeilschifter
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, Goethe University, Frankfurt am Main, 60596, Frankfurt, Germany
- Department of Neurology and Clinical Neurophysiology, Städtisches Klinikum Lüneburg, 21339, Lüneburg, Germany
| | - Rajkumar Vutukuri
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, Goethe University, Frankfurt am Main, 60596, Frankfurt, Germany.
| | - Josef Pfeilschifter
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, Goethe University, Frankfurt am Main, 60596, Frankfurt, Germany.
| |
Collapse
|
3
|
Liu J, Liu X, Luo Y, Huang F, Xie Y, Zheng S, Jia B, Xiao Z. Sphingolipids: drivers of cardiac fibrosis and atrial fibrillation. J Mol Med (Berl) 2024; 102:149-165. [PMID: 38015241 PMCID: PMC10858135 DOI: 10.1007/s00109-023-02391-8] [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: 06/29/2023] [Revised: 10/13/2023] [Accepted: 10/23/2023] [Indexed: 11/29/2023]
Abstract
Sphingolipids (SLs) are vital constituents of the plasma membrane of animal cells and concurrently regulate numerous cellular processes. An escalating number of research have evinced that SLs assume a crucial part in the progression of tissue fibrosis, a condition for which no efficacious cure exists as of now. Cardiac fibrosis, and in particular, atrial fibrosis, is a key factor in the emergence of atrial fibrillation (AF). AF has become one of the most widespread cardiac arrhythmias globally, with its incidence continuing to mount, thereby propelling it to the status of a major public health concern. This review expounds on the structure and biosynthesis pathways of several pivotal SLs, the pathophysiological mechanisms of AF, and the function of SLs in cardiac fibrosis. Delving into the influence of sphingolipid levels in the alleviation of cardiac fibrosis offers innovative therapeutic strategies to address cardiac fibrosis and AF.
Collapse
Affiliation(s)
- Junjie Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ximao Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yucheng Luo
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fangze Huang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Xie
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| | - Zezhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Image-Based Longitudinal Characterization of Corneal Wound to Understand the Role of Sphingosine-1-Phosphate. Methods Mol Biol 2023; 2625:337-345. [PMID: 36653655 DOI: 10.1007/978-1-0716-2966-6_28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Since its discovery, the bioactive sphingolipid sphingosine 1-phosphate (S1P) has been shown to involve in a myriad of cellular and physiological processes. In the process of tissue healing, S1P plays an important role in both normal and pathological healing, leading to fibrosis in multiple tissues including the cornea. Cornea covers the anterior portion of the eye and is responsible for the refraction of light. Corneal transparency is essential to obtain a clear vision, and a proper wound healing process is necessary for a clear cornea. Even though S1P is indicated to be a critical player in corneal fibrosis, we lack a detailed understanding of the role of S1P signaling in corneal wound healing and fibrosis. Herein, we describe a methodology to characterize the in-vivo wound healing process of the cornea using an easy and affordable imaging-based assay. This gives a consistent and easy way to characterize the wound and also the longitudinal healing process.
Collapse
|
5
|
Wilkerson JL, Basu SK, Stiles MA, Prislovsky A, Grambergs RC, Nicholas SE, Karamichos D, Allegood JC, Proia RL, Mandal N. Ablation of Sphingosine Kinase 1 Protects Cornea from Neovascularization in a Mouse Corneal Injury Model. Cells 2022; 11:cells11182914. [PMID: 36139489 PMCID: PMC9497123 DOI: 10.3390/cells11182914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/03/2022] [Accepted: 09/10/2022] [Indexed: 11/24/2022] Open
Abstract
The purpose of this study was to investigate the role of sphingosine kinase 1 (SphK1), which generates sphingosine-1-phosphate (S1P), in corneal neovascularization (NV). Wild-type (WT) and Sphk1 knockout (Sphk1−/−) mice received corneal alkali-burn treatment to induce corneal NV by placing a 2 mm round piece of Whatman No. 1 filter paper soaked in 1N NaOH on the center of the cornea for 20 s. Corneal sphingolipid species were extracted and identified using liquid chromatography/mass spectrometry (LC/MS). The total number of tip cells and those positive for ethynyl deoxy uridine (EdU) were quantified. Immunocytochemistry was done to examine whether pericytes were present on newly forming blood vessels. Cytokine signaling and angiogenic markers were compared between the two groups using multiplex assays. Data were analyzed using appropriate statistical tests. Here, we show that ablation of SphK1 can significantly reduce NV invasion in the cornea following injury. Corneal sphingolipid analysis showed that total levels of ceramides, monohexosyl ceramides (HexCer), and sphingomyelin were significantly elevated in Sphk−/− corneas compared to WT corneas, with a comparable level of sphingosine among the two genotypes. The numbers of total and proliferating endothelial tip cells were also lower in the Sphk1−/− corneas following injury. This study underscores the role of S1P in post-injury corneal NV and raises further questions about the roles played by ceramide, HexCer, and sphingomyelin in regulating corneal NV. Further studies are needed to unravel the role played by bioactive sphingolipids in maintenance of corneal transparency and clear vision.
Collapse
Affiliation(s)
- Joseph L. Wilkerson
- Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Sandip K. Basu
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Megan A. Stiles
- Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Amanda Prislovsky
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Richard C. Grambergs
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Sarah E. Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Jeremy C. Allegood
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Richard L. Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nawajes Mandal
- Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
- Departments of Anatomy and Neurobiology, and Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
- Memphis VA Medical Center, Memphis, TN 38104, USA
- Correspondence:
| |
Collapse
|
6
|
Sphk1 and Sphk2 Differentially Regulate Erythropoietin Synthesis in Mouse Renal Interstitial Fibroblast-like Cells. Int J Mol Sci 2022; 23:ijms23115882. [PMID: 35682566 PMCID: PMC9180811 DOI: 10.3390/ijms23115882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Erythropoietin (Epo) is a crucial hormone regulating red blood cell number and consequently the hematocrit. Epo is mainly produced in the kidney by interstitial fibroblast-like cells. Previously, we have shown that in cultures of the immortalized mouse renal fibroblast-like cell line FAIK F3-5, sphingosine 1-phosphate (S1P), by activating S1P1 and S1P3 receptors, can stabilize hypoxia-inducible factor (HIF)-2α and upregulate Epo mRNA and protein synthesis. In this study, we have addressed the role of intracellular iS1P derived from sphingosine kinases (Sphk) 1 and 2 on Epo synthesis in F3-5 cells and in mouse primary cultures of renal fibroblasts. We show that stable knockdown of Sphk2 in F3-5 cells increases HIF-2α protein and Epo mRNA and protein levels, while Sphk1 knockdown leads to a reduction of hypoxia-stimulated HIF-2α and Epo protein. A similar effect was obtained using primary cultures of renal fibroblasts isolated from wildtype mice, Sphk1−/−, or Sphk2−/− mice. Furthermore, selective Sphk2 inhibitors mimicked the effect of genetic Sphk2 depletion and also upregulated HIF-2α and Epo protein levels. The combined blockade of Sphk1 and Sphk2, using Sphk2−/− renal fibroblasts treated with the Sphk1 inhibitor PF543, resulted in reduced HIF-2α and Epo compared to the untreated Sphk2−/− cells. Exogenous sphingosine (Sph) enhanced HIF-2α and Epo, and this was abolished by the combined treatment with the selective S1P1 and S1P3 antagonists NIBR-0213 and TY52156, suggesting that Sph was taken up by cells and converted to iS1P and exported to then act in an autocrine manner through S1P1 and S1P3. The upregulation of HIF-2α and Epo synthesis by Sphk2 knockdown was confirmed in the human hepatoma cell line Hep3B, which is well-established to upregulate Epo production under hypoxia. In summary, these data show that sphingolipids have diverse effects on Epo synthesis. While accumulation of intracellular Sph reduces Epo synthesis, iS1P will be exported to act through S1P1+3 to enhance Epo synthesis. Furthermore, these data suggest that selective inhibition of Sphk2 is an attractive new option to enhance Epo synthesis and thereby to reduce anemia development in chronic kidney disease.
Collapse
|
7
|
Extracellular Lipids in the Lung and Their Role in Pulmonary Fibrosis. Cells 2022; 11:cells11071209. [PMID: 35406772 PMCID: PMC8997955 DOI: 10.3390/cells11071209] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 02/04/2023] Open
Abstract
Lipids are major actors and regulators of physiological processes within the lung. Initial research has described their critical role in tissue homeostasis and in orchestrating cellular communication to allow respiration. Over the past decades, a growing body of research has also emphasized how lipids and their metabolism may be altered, contributing to the development and progression of chronic lung diseases such as pulmonary fibrosis. In this review, we first describe the current working model of the mechanisms of lung fibrogenesis before introducing lipids and their cellular metabolism. We then summarize the evidence of altered lipid homeostasis during pulmonary fibrosis, focusing on their extracellular forms. Finally, we highlight how lipid targeting may open avenues to develop therapeutic options for patients with lung fibrosis.
Collapse
|
8
|
Abstract
The relationship between sphingolipid levels and NAFLD pathology has been recognized for some time. Numerous studies using pharmacological and genetic approaches in vitro and in animal models of NAFLD have demonstrated that modifications to sphingolipid metabolism can attenuate various facets of NAFLD pathology. However, a more precise understanding of the role of sphingolipids and NAFLD pathology is essential to creating therapeutics that target this pathway. This chapter touches on the scale and variety of sphingolipid metabolites at play in NAFLD, which vary widely in their chemical structures and biological functions. With advances in liquid chromatography and tandem mass spectrometry approaches, each of thousands of individual sphingolipid species and sphingolipid metabolites can be identified and precisely quantified. These approaches are beginning to reveal specific sub-classes and species of sphingolipids that change in NAFLD, and as such, enzymes that generate them can be identified and potentially serve as therapeutic targets. Advances in lipidomics technology have been, and will continue to be, critical to these gains in our understanding of NAFLD.
Collapse
Affiliation(s)
- David Montefusco
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA.
| | - Johana Lambert
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrea Anderson
- Department of Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Jeremy Allegood
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
9
|
Hafizi R, Imeri F, Wenger RH, Huwiler A. S1P Stimulates Erythropoietin Production in Mouse Renal Interstitial Fibroblasts by S1P 1 and S1P 3 Receptor Activation and HIF-2α Stabilization. Int J Mol Sci 2021; 22:ijms22179467. [PMID: 34502385 PMCID: PMC8430949 DOI: 10.3390/ijms22179467] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 02/06/2023] Open
Abstract
Erythropoietin (Epo) is the critical hormone for erythropoiesis. In adults, Epo is mainly produced by a subset of interstitial fibroblasts in the kidney, with minor amounts being produced in the liver and the brain. In this study, we used the immortalized renal interstitial fibroblast cell line FAIK F3-5 to investigate the ability of the bioactive sphingolipid sphingosine 1-phosphate (S1P) to stimulate Epo production and to reveal the mechanism involved. Stimulation of cells with exogenous S1P under normoxic conditions (21% O2) led to a dose-dependent increase in Epo mRNA and protein levels and subsequent release of Epo into the medium. S1P also enhanced the stabilization of HIF-2α, a key transcription factor for Epo expression. S1P-stimulated Epo mRNA and protein expression was abolished by HIF-2α mRNA knockdown or by the HIF-2 inhibitor compound 2. Furthermore, the approved S1P receptor modulator FTY720, and its active form FTY720-phosphate, both exerted a similar effect on Epo expression as S1P. The effect of S1P on Epo was antagonized by the selective S1P1 and S1P3 antagonists NIBR-0213 and TY-52156, but not by the S1P2 antagonist JTE-013. Moreover, inhibitors of the classical MAPK/ERK, the p38-MAPK, and inhibitors of protein kinase (PK) C and D all blocked the effect of S1P on Epo expression. Finally, the S1P and FTY720 effects were recapitulated in the Epo-producing human neuroblastoma cell line Kelly, suggesting that S1P receptor-dependent Epo synthesis is of general relevance and not species-specific. In summary, these data suggest that, in renal interstitial fibroblasts, which are the primary source of plasma Epo, S1P1 and 3 receptor activation upregulates Epo under normoxic conditions. This may have a therapeutic impact on disease situations such as chronic kidney disease, where Epo production is impaired, causing anemia, but it may also have therapeutic value as Epo can mediate additional tissue-protective effects in various organs.
Collapse
Affiliation(s)
- Redona Hafizi
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010 Bern, Switzerland; (R.H.); (F.I.)
| | - Faik Imeri
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010 Bern, Switzerland; (R.H.); (F.I.)
| | - Roland H. Wenger
- Institute of Physiology, University of Zürich, CH-8057 Zürich, Switzerland;
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010 Bern, Switzerland; (R.H.); (F.I.)
- Correspondence: ; Tel.: +41-316-323-214
| |
Collapse
|
10
|
Negative regulators of TGF-β1 signaling in renal fibrosis; pathological mechanisms and novel therapeutic opportunities. Clin Sci (Lond) 2021; 135:275-303. [PMID: 33480423 DOI: 10.1042/cs20201213] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/23/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023]
Abstract
Elevated expression of the multifunctional cytokine transforming growth factor β1 (TGF-β1) is causatively linked to kidney fibrosis progression initiated by diabetic, hypertensive, obstructive, ischemic and toxin-induced injury. Therapeutically relevant approaches to directly target the TGF-β1 pathway (e.g., neutralizing antibodies against TGF-β1), however, remain elusive in humans. TGF-β1 signaling is subjected to extensive negative control at the level of TGF-β1 receptor, SMAD2/3 activation, complex assembly and promoter engagement due to its critical role in tissue homeostasis and numerous pathologies. Progressive kidney injury is accompanied by the deregulation (loss or gain of expression) of several negative regulators of the TGF-β1 signaling cascade by mechanisms involving protein and mRNA stability or epigenetic silencing, further amplifying TGF-β1/SMAD3 signaling and fibrosis. Expression of bone morphogenetic proteins 6 and 7 (BMP6/7), SMAD7, Sloan-Kettering Institute proto-oncogene (Ski) and Ski-related novel gene (SnoN), phosphate tensin homolog on chromosome 10 (PTEN), protein phosphatase magnesium/manganese dependent 1A (PPM1A) and Klotho are dramatically decreased in various nephropathies in animals and humans albeit with different kinetics while the expression of Smurf1/2 E3 ligases are increased. Such deregulations frequently initiate maladaptive renal repair including renal epithelial cell dedifferentiation and growth arrest, fibrotic factor (connective tissue growth factor (CTGF/CCN2), plasminogen activator inhibitor type-1 (PAI-1), TGF-β1) synthesis/secretion, fibroproliferative responses and inflammation. This review addresses how loss of these negative regulators of TGF-β1 pathway exacerbates renal lesion formation and discusses the therapeutic value in restoring the expression of these molecules in ameliorating fibrosis, thus, presenting novel approaches to suppress TGF-β1 hyperactivation during chronic kidney disease (CKD) progression.
Collapse
|
11
|
Barochia AV, Kaler M, Weir N, Gordon EM, Figueroa DM, Yao X, WoldeHanna ML, Sampson M, Remaley AT, Grant G, Barnett SD, Nathan SD, Levine SJ. Serum levels of small HDL particles are negatively correlated with death or lung transplantation in an observational study of idiopathic pulmonary fibrosis. Eur Respir J 2021; 58:13993003.04053-2020. [PMID: 34289973 DOI: 10.1183/13993003.04053-2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/13/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Serum lipoproteins, such as high density lipoproteins (HDL), may influence disease severity in idiopathic pulmonary fibrosis (IPF). Here, we investigated associations between serum lipids and lipoproteins and clinical endpoints in IPF. METHODS Clinical data and serum lipids were analyzed from a discovery cohort (59 IPF subjects, 56 healthy volunteers) and validated using an independent, multicenter cohort (207 IPF subjects) from the Pulmonary Fibrosis Foundation registry. Associations between lipids and clinical endpoints (FVC, forced vital capacity; 6MWD, 6 min walk distance; GAP (Gender Age Physiology) index; death or lung transplantation) were examined using Pearson's correlation and multivariable analyses. RESULTS Serum concentrations of small HDL particles (S-HDLPNMR), measured by nuclear magnetic resonance (NMR) spectroscopy, correlated negatively with the GAP index in the discovery cohort of IPF subjects. The negative correlation of S-HDLPNMR with GAP index was confirmed in the validation cohort of IPF subjects. Higher levels of S-HDLPNMR were associated with lower odds of death or its competing outcome, lung transplantation (OR of 0.9 for each 1 μmol·L-1 increase in S-HDLPNMR, p<0.05), at 1, 2, and 3 years from study entry in a combined cohort of all IPF subjects. CONCLUSIONS Higher serum levels of S-HDLPNMR are negatively correlated with the GAP index, as well as with lower observed mortality or lung transplantation in IPF subjects. These findings support the hypothesis that S-HDLPNMR may modify mortality risk in patients with IPF.
Collapse
Affiliation(s)
- Amisha V Barochia
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, NHLBI, NIH, Bethesda, MD, USA
| | - Maryann Kaler
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, NHLBI, NIH, Bethesda, MD, USA
| | - Nargues Weir
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, NHLBI, NIH, Bethesda, MD, USA.,Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, VA, USA
| | - Elizabeth M Gordon
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, NHLBI, NIH, Bethesda, MD, USA
| | - Debbie M Figueroa
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, NHLBI, NIH, Bethesda, MD, USA
| | - Xianglan Yao
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, NHLBI, NIH, Bethesda, MD, USA
| | - Merte Lemma WoldeHanna
- Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, VA, USA
| | | | - Alan T Remaley
- Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD, USA
| | | | - Scott D Barnett
- Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, VA, USA
| | - Steven D Nathan
- Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, VA, USA
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, NHLBI, NIH, Bethesda, MD, USA
| |
Collapse
|
12
|
Galal SA, Omar MA, Khairat SHM, Ragab FAF, Roy S, Naqvi AAT, Hassan MI, El Diwani HI. Design and synthesis of new pyrazolylbenzimidazoles as sphingosine kinase-1 inhibitors. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02760-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
13
|
Khairat SHM, Omar MA, Ragab FAF, Roy S, Turab Naqvi AA, Abdelsamie AS, Hirsch AKH, Galal SA, Hassan MI, El Diwani HI. Design, synthesis, and biological evaluation of novel benzimidazole derivatives as sphingosine kinase 1 inhibitor. Arch Pharm (Weinheim) 2021; 354:e2100080. [PMID: 34128259 DOI: 10.1002/ardp.202100080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/17/2021] [Accepted: 04/23/2021] [Indexed: 11/08/2022]
Abstract
Sphingosine kinase 1 (SphK1) has emerged as an attractive drug target for different diseases. Recently, discovered SphK1 inhibitors have been recommended in cancer therapeutics; however, selectivity and potency are great challenges. In this study, a novel series of benzimidazoles was synthesized and evaluated as SphK1 inhibitors. Our design strategy is twofold: It aimed first to study the effect of replacing the 5-position of the benzimidazole ring with a polar carboxylic acid group on the SphK1-inhibitory activity and cytotoxicity. Our second aim was to optimize the structures of the benzimidazoles through the elongation of the chain. The enzyme inhibition potentials against all the synthesized compounds toward SphK1 were evaluated, and the results revealed that most of the studied compounds inhibited SphK1 effectively. The binding affinity of the benzimidazole derivatives toward SphK1 was measured by fluorescence binding and molecular docking. Compounds 33, 37, 39, 41, 42, 43, and 45 showed an appreciable binding affinity. Therefore, the SphK1-inhibitory potentials of compounds 33, 37, 39, 41, 42, 43, and 45 were studied and IC50 values were determined, to reveal high potency. The study showed that these compounds inhibited SphK1 with effective IC50 values. Among the studied compounds, compound 41 was the most effective one with the lowest IC50 value and a high cytotoxicity on a wide spectrum of cell lines. Molecular docking revealed that most of these compounds fit well into the ATP-binding site of SphK1 and form hydrogen bond interactions with catalytically important residues. Overall, the findings suggest the therapeutic potential of benzimidazoles in the clinical management of SphK1-associated diseases.
Collapse
Affiliation(s)
- Sarah H M Khairat
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries Research, National Research Centre, Cairo, Egypt
| | - Mohamed A Omar
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries Research, National Research Centre, Cairo, Egypt
| | - Fatma A F Ragab
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Sonam Roy
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Ahmad A Turab Naqvi
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Ahmed S Abdelsamie
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries Research, National Research Centre, Cairo, Egypt.,Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Anna K H Hirsch
- Department of Pharmacy, Saarland University, Saarbrücken, Germany.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Shadia A Galal
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries Research, National Research Centre, Cairo, Egypt
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Hoda I El Diwani
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries Research, National Research Centre, Cairo, Egypt
| |
Collapse
|
14
|
Mammoliti O, Palisse A, Joannesse C, El Bkassiny S, Allart B, Jaunet A, Menet C, Coornaert B, Sonck K, Duys I, Clément-Lacroix P, Oste L, Borgonovi M, Wakselman E, Christophe T, Houvenaghel N, Jans M, Heckmann B, Sanière L, Brys R. Discovery of the S1P2 Antagonist GLPG2938 (1-[2-Ethoxy-6-(trifluoromethyl)-4-pyridyl]-3-[[5-methyl-6-[1-methyl-3-(trifluoromethyl)pyrazol-4-yl]pyridazin-3-yl]methyl]urea), a Preclinical Candidate for the Treatment of Idiopathic Pulmonary Fibrosis. J Med Chem 2021; 64:6037-6058. [PMID: 33939425 DOI: 10.1021/acs.jmedchem.1c00138] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Mounting evidence from the literature suggests that blocking S1P2 receptor (S1PR2) signaling could be effective for the treatment of idiopathic pulmonary fibrosis (IPF). However, only a few antagonists have been so far disclosed. A chemical enablement strategy led to the discovery of a pyridine series with good antagonist activity. A pyridazine series with improved lipophilic efficiency and with no CYP inhibition liability was identified by scaffold hopping. Further optimization led to the discovery of 40 (GLPG2938), a compound with exquisite potency on a phenotypic IL8 release assay, good pharmacokinetics, and good activity in a bleomycin-induced model of pulmonary fibrosis.
Collapse
Affiliation(s)
- Oscar Mammoliti
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Adeline Palisse
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | | | - Brigitte Allart
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Alex Jaunet
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Christel Menet
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | - Kathleen Sonck
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Inge Duys
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | - Line Oste
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Monica Borgonovi
- Galapagos SASU, 102 avenue Gaston Roussel, 93230 Romainville, France
| | | | | | | | - Mia Jans
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Bertrand Heckmann
- Galapagos SASU, 102 avenue Gaston Roussel, 93230 Romainville, France
| | - Laurent Sanière
- Galapagos SASU, 102 avenue Gaston Roussel, 93230 Romainville, France
| | - Reginald Brys
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| |
Collapse
|
15
|
Magaye RR, Savira F, Hua Y, Xiong X, Huang L, Reid C, Flynn BL, Kaye D, Liew D, Wang BH. Attenuating PI3K/Akt- mTOR pathway reduces dihydrosphingosine 1 phosphate mediated collagen synthesis and hypertrophy in primary cardiac cells. Int J Biochem Cell Biol 2021; 134:105952. [PMID: 33609744 DOI: 10.1016/j.biocel.2021.105952] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/15/2022]
Abstract
Cardiac fibrosis and myocyte hypertrophy play contributory roles in the progression of diseases such as heart Failure (HF) through what is collectively termed cardiac remodelling. The phosphoinositide 3- kinase (PI3K), protein kinase B (Akt) and mammalian target for rapamycin (mTOR) signalling pathway (PI3K/Akt- mTOR) is an important pathway in protein synthesis, cell growth, cell proliferation, and lipid metabolism. The sphingolipid, dihydrosphingosine 1 phosphate (dhS1P) has been shown to bind to high density lipids in plasma. Unlike its analog, spingosine 1 phosphate (S1P), the role of dhS1P in cardiac fibrosis is still being deciphered. This study was conducted to investigate the effect of dhS1P on PI3K/Akt signalling in primary cardiac fibroblasts and myocytes. Our findings demonstrate that inhibiting PI3K reduced collagen synthesis in neonatal cardiac fibroblasts (NCFs), and hypertrophy in neonatal cardiac myocytes (NCMs) induced by dhS1P, in vitro. Reduced activation of the PI3K/Akt- mTOR signalling pathway led to impaired translation of fibrotic proteins such as collagen 1 (Coll1) and transforming growth factor β (TGFβ) and inhibited the transcription and translation of tissue inhibitor of matrix metalloproteinase 1 (TIMP1). PI3K inhibition also affected the gene expression of S1P receptors and enzymes such as the dihydroceramide delta 4 desaturase (DEGS1) and sphingosine kinase 1 (SK1) in the de novo sphingolipid pathway. While in myocytes, PI3K inhibition reduced myocyte hypertrophy induced by dhS1P by reducing skeletal muscle α- actin (αSKA) mRNA expression, and protein translation due to increased glycogen synthase kinase 3β (GSK3β) mRNA expression. Our findings show a relationship between the PI3K/Akt- mTOR signalling cascade and exogenous dhS1P induced collagen synthesis and myocyte hypertrophy in primary neonatal cardiac cells.
Collapse
Affiliation(s)
- Ruth R Magaye
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Feby Savira
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Yue Hua
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xin Xiong
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; Shanghai Institute of Heart Failure, Research Centre for Translational Medicine, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai 200120, China
| | - Li Huang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Christopher Reid
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; School of Public Health School, Curtin University, Perth, Australia
| | - Bernard L Flynn
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - David Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Danny Liew
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Bing H Wang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia.
| |
Collapse
|
16
|
Amargant F, Manuel SL, Larmore MJ, Johnson BW, Lawson M, Pritchard MT, Zelinski MB, Duncan FE. Sphingosine-1-phosphate and its mimetic FTY720 do not protect against radiation-induced ovarian fibrosis in the nonhuman primate†. Biol Reprod 2021; 104:1058-1070. [PMID: 33524104 DOI: 10.1093/biolre/ioab012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/18/2020] [Accepted: 01/29/2021] [Indexed: 12/26/2022] Open
Abstract
Oocytes are highly radiosensitive, so agents that prevent radiation-induced ovarian follicle destruction are important fertility preservation strategies. A previous study in rhesus macaques demonstrated that ovarian treatment with antiapoptotic agents, sphingosine-1-phosphate (S1P) and FTY720, its long-acting mimetic, preserved follicles following a single dose of 15 Gy X-ray radiation, and live offspring were obtained from FTY720-treated animals. However, it is unknown whether these antiapoptotic agents also protected the ovarian stroma from late effects of radiation, including vascular damage and fibrosis. Using ovarian histological sections from this study, we evaluated the vasculature and extracellular matrix in the following cohorts: vehicle + sham irradiation, vehicle + irradiation (OXI), S1P + irradiation (S1P), and FTY720 + irradiation (FTY720). One ovary from each animal was harvested prior to radiation whereas the contralateral ovary was harvested 10 months post-treatment. We assessed vasculature by immunohistochemistry with a PECAM1 antibody, hyaluronan by a hyaluronan binding protein assay, and collagen by picrosirius red and Masson's trichrome staining. Disorganized vessels were observed in the medulla in the OXI and S1P cohorts relative to the sham, but the vasculature in the FTY720 cohort appeared intact, which may partially explain fertoprotection. There were no differences in the hyaluronan matrix among the cohorts, but there was thickening of the tunica albuginea and fibrosis in the OXI cohort relative to the sham, which was not mitigated by either S1P or FTY720 treatment. Thus, the fertoprotective properties of S1P and FTY720 may be limited given their inability to protect the ovarian stroma against the late effects of radiation-induced fibrosis.
Collapse
Affiliation(s)
- Farners Amargant
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sharrón L Manuel
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Megan J Larmore
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Brian W Johnson
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Maralee Lawson
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Michele T Pritchard
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mary B Zelinski
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, USA.,Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
17
|
Schwalm S, Beyer S, Hafizi R, Trautmann S, Geisslinger G, Adams DR, Pyne S, Pyne N, Schaefer L, Huwiler A, Pfeilschifter J. Validation of highly selective sphingosine kinase 2 inhibitors SLM6031434 and HWG-35D as effective anti-fibrotic treatment options in a mouse model of tubulointerstitial fibrosis. Cell Signal 2020; 79:109881. [PMID: 33301900 DOI: 10.1016/j.cellsig.2020.109881] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/04/2020] [Accepted: 12/06/2020] [Indexed: 01/17/2023]
Abstract
Renal fibrosis is characterized by chronic inflammation and excessive accumulation of extracellular matrix and progressively leads to functional insufficiency and even total loss of kidney function. In this study we investigated the anti-fibrotic potential of two highly selective and potent SK2 inhibitors, SLM6031434 and HWG-35D, in unilateral ureter obstruction (UUO), a model for progressive renal fibrosis, in mice. In both cases, treatment with SLM6031434 or HWG-35D resulted in an attenuated fibrotic response to UUO in comparison to vehicle-treated mice as demonstrated by reduced collagen accumulation and a decreased expression of collagen-1 (Col1), fibronectin-1 (FN-1), connective tissue growth factor (CTGF), and α-smooth muscle actin (α-SMA). Similar to our previous study in Sphk2-/- mice, we found an increased protein expression of Smad7, a negative regulator of the pro-fibrotic TGFβ/Smad signalling cascade, accompanied by a strong accumulation of sphingosine in SK2 inhibitor-treated kidneys. Treatment of primary renal fibroblasts with SLM6031434 or HWG-35D dose-dependently increased Smad7 expression and ameliorated the expression of Col1, FN-1 and CTGF. In summary, these data prove the anti-fibrotic potential of SK2 inhibition in a mouse model of renal fibrosis, thereby validating SK2 as pharmacological target for the treatment of fibrosis in chronic kidney disease.
Collapse
Affiliation(s)
- Stephanie Schwalm
- Pharmazentrum Frankfurt/ZAFES, Institute of General Pharmacology and Toxicology, Universitätsklinikum and Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Sandra Beyer
- Pharmazentrum Frankfurt/ZAFES, Institute of General Pharmacology and Toxicology, Universitätsklinikum and Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Redona Hafizi
- Institute of Pharmacology, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland
| | - Sandra Trautmann
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Universitätsklinikum and Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Universitätsklinikum and Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - David R Adams
- School of Engineering & Physical Sciences, Heriot-Watt University, Edinburgh, UK
| | - Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Nigel Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institute of General Pharmacology and Toxicology, Universitätsklinikum and Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, Institute of General Pharmacology and Toxicology, Universitätsklinikum and Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
18
|
Fan X, Liu L, Shi Y, Guo F, He X, Zhao X, Zhong D, Li G. Recent advances of the function of sphingosine 1-phosphate (S1P) receptor S1P3. J Cell Physiol 2020; 236:1564-1578. [PMID: 33410533 DOI: 10.1002/jcp.29958] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/18/2022]
Abstract
Known as a variety of sphingolipid metabolites capable of performing various biological activities, sphingosine 1-phosphate (S1P) is commonly found in platelets, red blood cells, neutrophils, lymph fluid, and blood, as well as other cells and body fluids. S1P comprises five receptors, namely, S1P1-S1P5, with the distribution of S1P receptors exhibiting tissue selectivity to some degree. S1P1, S1P2, and S1P3 are extensively expressed in a wide variety of different tissues. The expression of S1P4 is restricted to lymphoid and hematopoietic tissues, while S1P5 is primarily expressed in the nervous system. S1P3 plays an essential role in the pathophysiological processes related to inflammation, cell proliferation, cell migration, tumor invasion and metastasis, ischemia-reperfusion, tissue fibrosis, and vascular tone. In this paper, the relevant mechanism in the role of S1P3 is summarized.
Collapse
Affiliation(s)
- Xuehui Fan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lili Liu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Shi
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fanghan Guo
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiao He
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiuli Zhao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Di Zhong
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
19
|
Xie T, Chen C, Peng Z, Brown BC, Reisz JA, Xu P, Zhou Z, Song A, Zhang Y, Bogdanov MV, Kellems RE, D'Alessandro A, Zhang W, Xia Y. Erythrocyte Metabolic Reprogramming by Sphingosine 1-Phosphate in Chronic Kidney Disease and Therapies. Circ Res 2020; 127:360-375. [PMID: 32284030 DOI: 10.1161/circresaha.119.316298] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RATIONALE Hypoxia promotes renal damage and progression of chronic kidney disease (CKD). The erythrocyte is the only cell type for oxygen (O2) delivery. Sphingosine 1-phosphate (S1P)-a highly enriched biolipid in erythrocytes-is recently reported to be induced under high altitude in normal humans to enhance O2 delivery. However, nothing is known about erythrocyte S1P in CKD. OBJECTIVE To investigate the function and metabolic basis of erythrocyte S1P in CKD with a goal to explore potential therapeutics. METHODS AND RESULTS Using erythrocyte-specific SphK1 (sphingosine kinase 1; the only enzyme to produce S1P in erythrocytes) knockout mice (eSphK1-/-) in an experimental model of hypertensive CKD with Ang II (angiotensin II) infusion, we found severe renal hypoxia, hypertension, proteinuria, and fibrosis in Ang II-infused eSphk1-/- mice compared with controls. Untargeted metabolomics profiling and in vivo U-13C6 isotopically labeled glucose flux analysis revealed that SphK1 is required for channeling glucose metabolism toward glycolysis versus pentose phosphate pathway, resulting in enhanced erythroid-specific Rapoport-Luebering shunt in Ang II-infused mice. Mechanistically, increased erythrocyte S1P functioning intracellularly activates AMPK (AMP-activated protein kinase) 1α and BPGM (bisphosphoglycerate mutase) by reducing ceramide/S1P ratio and inhibiting PP2A (protein phosphatase 2A), leading to increased 2,3-bisphosphoglycerate (an erythrocyte-specific metabolite negatively regulating Hb [hemoglobin]-O2-binding affinity) production and thus more O2 delivery to counteract kidney hypoxia and progression to CKD. Preclinical studies revealed that an AMPK agonist or a PP2A inhibitor rescued the severe CKD phenotype in Ang II-infused eSphK1-/- mice and prevented development of CKD in the control mice by inducing 2,3-bisphosphoglycerate production and thus enhancing renal oxygenation. Translational research validated mouse findings in erythrocytes of hypertensive CKD patients and cultured human erythrocytes. CONCLUSIONS Our study elucidates the beneficial role of eSphk1-S1P in hypertensive CKD by channeling glucose metabolism toward Rapoport-Luebering shunt and inducing 2,3-bisphosphoglycerate production and O2 delivery via a PP2A-AMPK1α signaling pathway. These findings reveal the metabolic and molecular basis of erythrocyte S1P in CKD and new therapeutic avenues.
Collapse
Affiliation(s)
- Tingting Xie
- From the Rheumatology and Immunology (T.X.), Xiangya Hospital, Central South University, Changsha, Hunan, China.,Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Changhan Chen
- Otolaryngology Head and Neck Surgery (C.C.), Xiangya Hospital, Central South University, Changsha, Hunan, China.,Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Zhangzhe Peng
- Nephrology (Z.P.), Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Benjamin C Brown
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora (B.C.B., J.A.R., A.D.)
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora (B.C.B., J.A.R., A.D.)
| | - Ping Xu
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Zhen Zhou
- Division of Medical Genetics, Department of Internal Medicine (Z.Z.), University of Texas McGovern Medical School at Houston
| | - Anren Song
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Yujin Zhang
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Mikhail V Bogdanov
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Rodney E Kellems
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston.,MDAnderson-UTHealth Graduate School of Biomedical Science, Houston, TX (R.E.K., Y.X.)
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora (B.C.B., J.A.R., A.D.)
| | - Weiru Zhang
- General Medicine (W.Z.), Xiangya Hospital, Central South University, Changsha, Hunan, China.,Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Yang Xia
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston.,MDAnderson-UTHealth Graduate School of Biomedical Science, Houston, TX (R.E.K., Y.X.)
| |
Collapse
|
20
|
Zeng J, Jiang B, Xiao X, Zhang R. Inhibition of sphingosine kinase 2 attenuates hypertrophic scar formation via upregulation of Smad7 in human hypertrophic scar fibroblasts. Mol Med Rep 2020; 22:2573-2582. [PMID: 32705254 PMCID: PMC7411334 DOI: 10.3892/mmr.2020.11313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 05/20/2020] [Indexed: 12/18/2022] Open
Abstract
The aims of the present study were to investigate the role of sphingosine kinase 2 (Sphk2) in hypertrophic scar (HS) formation and its underlying mechanisms. The expression levels of Sphk2 and Smad7 in HS tissues and healthy skin tissues of patients undergoing plastic surgery were determined using immunohistochemical staining. Subsequently, the expression levels of Sphk2 and collagen I in human embryonic skin fibroblasts (control) and human HS fibroblasts (HSF) were detected using western blot analysis and immunofluorescence assay, respectively. Following Sphk2 silencing, Smad7 overexpression or both Sphk2 and Smad7 silencing, HSF proliferative ability was assessed using Cell Counting Kit‑8 assay and proliferation‑associated proteins were evaluated using western blot analysis. In addition, the level of apoptosis in HSF was assessed using flow cytometry and expression levels of apoptotic‑associated proteins were determined using western blotting. Furthermore, the expression levels of collagen I and proteins in the TGF‑β1/Smad signaling pathway were detected using western blot analysis. The results indicated that the expression of Sphk2 was significantly increased, while Smad7 expression was decreased in HS tissue. Moreover, the upregulation of Sphk2 and collagen I expression levels was identified in HSF. The present results also indicated that Sphk2 silencing or Smad7 overexpression inhibited proliferation, but promoted apoptosis of HSF, coupled with changes in the expression levels of proliferation‑associated proteins, with an increase in p21 and a decrease in cyclin D1 expression levels, and apoptosis‑associated proteins, with an increase in Bax and cleaved caspase‑3, and a decrease in Bcl‑2, which were reversed following transfection with both Sphk2 and Smad7 using small interfering RNA in HSF. In addition, the expression levels of transforming growth factor‑β1, phosphorylated (p)‑Smad2, p‑Smad3 and collagen I were reduced following Sphk2 silencing or Smad7 overexpression, which were abolished by silencing both Sphk2 and Smad7. Collectively, the present results indicated that inhibition of Sphk2 attenuated HS formation via upregulation of Smad7 expression, thus Sphk2 may serve as a potential therapeutic target for the treatment of HS.
Collapse
Affiliation(s)
- Jian Zeng
- Department of Medical Cosmetology, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Bin Jiang
- Department of Medical Cosmetology, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xia Xiao
- Department of Medical Cosmetology, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Rou Zhang
- Department of Medical Cosmetology, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
21
|
Park WJ, Song JH, Kim GT, Park TS. Ceramide and Sphingosine 1-Phosphate in Liver Diseases. Mol Cells 2020; 43:419-430. [PMID: 32392908 PMCID: PMC7264474 DOI: 10.14348/molcells.2020.0054] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/06/2020] [Accepted: 04/19/2020] [Indexed: 12/12/2022] Open
Abstract
The liver is an important organ in the regulation of glucose and lipid metabolism. It is responsible for systemic energy homeostasis. When energy need exceeds the storage capacity in the liver, fatty acids are shunted into nonoxidative sphingolipid biosynthesis, which increases the level of cellular ceramides. Accumulation of ceramides alters substrate utilization from glucose to lipids, activates triglyceride storage, and results in the development of both insulin resistance and hepatosteatosis, increasing the likelihood of major metabolic diseases. Another sphingolipid metabolite, sphingosine 1-phosphate (S1P) is a bioactive signaling molecule that acts via S1P-specific G protein coupled receptors. It regulates many cellular and physiological events. Since an increase in plasma S1P is associated with obesity, it seems reasonable that recent studies have provided evidence that S1P is linked to lipid pathophysiology, including hepatosteatosis and fibrosis. Herein, we review recent findings on ceramides and S1P in obesity-mediated liver diseases and the therapeutic potential of these sphingolipid metabolites.
Collapse
Affiliation(s)
- Woo-Jae Park
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 2999, Korea
| | - Jae-Hwi Song
- Department of Life Science, Gachon University, Seongnam 1310, Korea
| | - Goon-Tae Kim
- Department of Life Science, Gachon University, Seongnam 1310, Korea
| | - Tae-Sik Park
- Department of Life Science, Gachon University, Seongnam 1310, Korea
| |
Collapse
|
22
|
Blankenbach KV, Bruno G, Wondra E, Spohner AK, Aster NJ, Vienken H, Trautmann S, Ferreirós N, Wieland T, Bruni P, Meyer Zu Heringdorf D. The WD40 repeat protein, WDR36, orchestrates sphingosine kinase-1 recruitment and phospholipase C-β activation by G q-coupled receptors. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158704. [PMID: 32244061 DOI: 10.1016/j.bbalip.2020.158704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 03/13/2020] [Accepted: 03/27/2020] [Indexed: 11/29/2022]
Abstract
Sphingosine kinases (SphK) catalyse the formation of sphingosine-1-phosphate (S1P) and play important roles in the cardiovascular, nervous and immune systems. We have shown before that Gq-coupled receptors induce a rapid and long-lasting translocation of SphK1 to the plasma membrane and cross-activation of S1P receptors. Here, we further addressed Gq regulation of SphK1 by analysing the influence of the WD40 repeat protein, WDR36. WDR36 has been described as a scaffold tethering Gαq to phospholipase C (PLC)-β and the thromboxane A2 receptor-β (TPβ receptor). Overexpression of WDR36 in HEK-293 cells enhanced TPβ receptor-induced inositol phosphate production, as reported (Cartier et al. 2011), but significantly attenuated inositol phosphate production induced by muscarinic M3 and bradykinin B2 receptors. In agreement with its effect on PLCβ, WDR36 augmented TPβ receptor-induced [Ca2+]i increases. Surprisingly, WDR36 also augmented M3 receptor-induced [Ca2+]i increases, which was due to increased Ca2+ mobilization while the Ca2+ content of thapsigargin-sensitive stores remained unaltered. Interestingly, overexpression of WDR36 significantly delayed SphK1 translocation by Gq-coupled M3, B2 and H1 receptors in HEK-293 cells, while TPβ receptor-induced SphK1 translocation was generally slow and not altered by WDR36 in these cells. Finally, in C2C12 myoblasts, overexpression of WDR36 delayed SphK1 translocation induced by B2 receptors. It is concluded that WDR36 reduces signalling of Gq-coupled receptors other than TPβ towards PLC and SphK1, most likely by scavenging Gαq and PLCβ. Our results support a role of WDR36 in orchestration of Gq signalling complexes, and might help to functionally unravel its genetic association with asthma and allergy.
Collapse
Affiliation(s)
- Kira Vanessa Blankenbach
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Gennaro Bruno
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università degli Studi di Firenze, Viale Morgagni 50, 50134 Firenze, Italy.
| | - Enrico Wondra
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Anna Katharina Spohner
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Natalie Judith Aster
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Hans Vienken
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Sandra Trautmann
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Nerea Ferreirós
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Thomas Wieland
- Experimentelle Pharmakologie, Medizinische Fakultät Mannheim der Universität Heidelberg, Ludolf Krehl-Str. 13-17, D-68167 Mannheim, Germany.
| | - Paola Bruni
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università degli Studi di Firenze, Viale Morgagni 50, 50134 Firenze, Italy.
| | - Dagmar Meyer Zu Heringdorf
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
23
|
Chen G, Zhang Q, Ai C, Huang S, Zhang H, Guo X, Wang W, Hua W, Bi H, Wang H. Serum metabolic profile characteristics of offspring rats before and after birth caused by prenatal caffeine exposure. Toxicology 2019; 427:152302. [PMID: 31568846 DOI: 10.1016/j.tox.2019.152302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/27/2019] [Accepted: 09/26/2019] [Indexed: 02/07/2023]
Abstract
Epidemiological investigations have confirmed that prenatal caffeine intake could increase the incidence rate of intrauterine growth retardation (IUGR) and multiple diseases after birth. Based on liquid chromatography-mass spectrometry, we analyzed serum metabolic profiles of offspring rats before and after birth in IUGR model induced by prenatal caffeine exposure (PCE). We discovered that differential metabolites in PCE fetuses mainly manifested as amino acids and lipid metabolism. In adulthood, PCE offspring showed less and inconsistent types of differential metabolites compared to those in utero, which still exhibited gender differences. The main differential metabolites induced by PCE, including phospholipids, platelet-activating factor, arachidonic acid, bile acid, sphingosine-1-phosphoric acid, indoxyl sulfuric acid, and cortexolone, may participate in the pathological and physiological processes of organ toxicities. This study demonstrated the short- and long-term developmental toxicity and gender differences of caffeine, providing new ideas for exploring the early warning and drug intervention targets of IUGR offspring.
Collapse
Affiliation(s)
- Guanghui Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Qi Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Can Ai
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Songqiang Huang
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Huizhen Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132# Waihuandong Road, Guangzhou University City, Guangzhou, 510006, China
| | - Xiaoyu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Wenju Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Weiying Hua
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Huichang Bi
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132# Waihuandong Road, Guangzhou University City, Guangzhou, 510006, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China.
| |
Collapse
|
24
|
Elwakeel E, Brüne B, Weigert A. PGE 2 in fibrosis and cancer: Insights into fibroblast activation. Prostaglandins Other Lipid Mediat 2019; 143:106339. [PMID: 31100473 DOI: 10.1016/j.prostaglandins.2019.106339] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/18/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023]
Abstract
Fibroblasts are the essential cellular architects of connective tissue and as such are crucial cells in contributing to organ homeostasis. While fulfilling important repair functions during tissue regeneration upon wounding, chronic fibroblast activation provokes pathological organ fibrosis and promotes neoplastic disease progression. Identifying targets that may serve to therapeutically terminate fibroblast activation is therefore desirable. Among the mediators that may be relevant in this context is the prostanoid prostaglandin E2 (PGE2) that is produced during inflammatory settings, where pathological fibrosis occurs. Here, we summarize current, in part controversial, concepts on the impact of PGE2 on fibroblast activation in fibrotic diseases including cancer, and discuss these findings in the context of the evolving concept of fibroblast heterogeneity.
Collapse
Affiliation(s)
- Eiman Elwakeel
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany.
| |
Collapse
|
25
|
Wang E, He X, Zeng M. The Role of S1P and the Related Signaling Pathway in the Development of Tissue Fibrosis. Front Pharmacol 2019; 9:1504. [PMID: 30687087 PMCID: PMC6338044 DOI: 10.3389/fphar.2018.01504] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/10/2018] [Indexed: 12/12/2022] Open
Abstract
Tissue fibrosis, including pulmonary fibrosis, hepatic fibrosis, and cardiac fibrosis, is an important stage in the development of many diseases. It can lead to structural damage and dysfunction and even severe carcinogenesis or death. There is currently no effective method for the treatment of fibrosis. At present, the molecular mechanism of tissue fibrosis has not yet been fully elucidated, but many studies have demonstrated that it is involved in conveying the complex messages between fibroblasts and various cytokines. Sphingosine 1-phosphate (S1P) is a naturally bioactive sphingolipid. S1P and the related signaling pathways are important intracellular metabolic pathways involved in many life activities, including cell proliferation, differentiation, apoptosis, and cellular signal transduction. Increasing evidence suggests that S1P and its signaling pathways play an important role in the development of tissue fibrosis; however, the mechanisms of these effects have not yet been fully elucidated, and even the role of S1P and its signaling pathways are still controversial. This article focuses on the role of S1P and the related signaling pathways in the development of fibrosis of lung, liver, heart, and other tissues, with emphasis on the application of inhibitors of some of molecules in the pathway in clinical treatment of fibrosis diseases.
Collapse
Affiliation(s)
- Erjin Wang
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xingxuan He
- Department of Human Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ming Zeng
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
26
|
Lan T, Li C, Yang G, Sun Y, Zhuang L, Ou Y, Li H, Wang G, Kisseleva T, Brenner D, Guo J. Sphingosine kinase 1 promotes liver fibrosis by preventing miR-19b-3p-mediated inhibition of CCR2. Hepatology 2018; 68:1070-1086. [PMID: 29572892 PMCID: PMC6174945 DOI: 10.1002/hep.29885] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/22/2018] [Accepted: 03/09/2018] [Indexed: 01/01/2023]
Abstract
UNLABELLED Chronic liver disease mediated by activation of hepatic stellate cells (HSCs) and Kupffer cells (KCs) leads to liver fibrosis. Here, we aimed to investigate the molecular mechanism and define the cell type involved in mediating the sphingosine kinase (SphK)1-dependent effect on liver fibrosis. The levels of expression and activity of SphK1 were significantly increased in fibrotic livers compared with the normal livers in human. SphK1 was coexpressed with a range of HSC/KC markers including desmin, α-smooth muscle actin (α-SMA) and F4/80 in fibrotic liver. Deficiency of SphK1 (SphK1-/- ) resulted in a marked amelioration of hepatic injury, including transaminase activities, histology, collagen deposition, α-SMA and inflammation, in CCl4 or bile duct ligation (BDL)-induced mice. Likewise, treatment with a specific inhibitor of SphK1, 5C, also significantly prevented liver injury and fibrosis in mice induced by CCl4 or BDL. In cellular levels, inhibition of SphK1 significantly blocked the activation and migration of HSCs and KCs. Moreover, SphK1 knockout in KCs reduced the secretion of CCL2, and SphK1 knockout in HSCs reduced C-C motif chemokine receptor 2 ([CCR2] CCL2 receptor) expression in HSCs. CCL2 in SphK1-/- mice was lower whereas microRNA-19b-3p in SphK1-/- mice was higher compared with wild-type (WT) mice. Furthermore, microRNA-19b-3p downregulated CCR2 in HSCs. The functional effect of SphK1 in HSCs on liver fibrosis was further strengthened by the results of animal experiments using a bone marrow transplantation (BMT) method. CONCLUSION SphK1 has distinct roles in the activation of KCs and HSCs in liver fibrosis. Mechanistically, SphK1 in KCs mediates CCL2 secretion, and SphK1 in HSCs upregulates CCR2 by downregulation of miR-19b-3p. (Hepatology 2018).
Collapse
Affiliation(s)
- Tian Lan
- Guangdong Pharmaceutical UniversityGuangzhouChina,Guangdong Metabolic Disease Research Center of Integrated Chinese and Western MedicineGuangzhouChina,Joint Laboratory of GuangdongHong Kong and Macao on Glycolipid Metabolic DiseasesGuangzhouChina
| | | | - Guizhi Yang
- Guangdong Pharmaceutical UniversityGuangzhouChina
| | - Yue Sun
- Guangdong Pharmaceutical UniversityGuangzhouChina,Guangdong Metabolic Disease Research Center of Integrated Chinese and Western MedicineGuangzhouChina,Joint Laboratory of GuangdongHong Kong and Macao on Glycolipid Metabolic DiseasesGuangzhouChina
| | - Lihang Zhuang
- Guangdong Pharmaceutical UniversityGuangzhouChina,Guangdong Metabolic Disease Research Center of Integrated Chinese and Western MedicineGuangzhouChina,Joint Laboratory of GuangdongHong Kong and Macao on Glycolipid Metabolic DiseasesGuangzhouChina
| | - Yitao Ou
- Guangdong Pharmaceutical UniversityGuangzhouChina
| | - Hui Li
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Genshu Wang
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | | | - David Brenner
- Department of MedicineUniversity of California San DiegoLa JollaCA
| | - Jiao Guo
- Guangdong Pharmaceutical UniversityGuangzhouChina,Guangdong Metabolic Disease Research Center of Integrated Chinese and Western MedicineGuangzhouChina,Joint Laboratory of GuangdongHong Kong and Macao on Glycolipid Metabolic DiseasesGuangzhouChina
| |
Collapse
|
27
|
Braetz J, Becker A, Geissen M, Larena-Avellaneda A, Schrepfer S, Daum G. Sphingosine-1-phosphate receptor 1 regulates neointimal growth in a humanized model for restenosis. J Vasc Surg 2018; 68:201S-207S. [PMID: 29804740 DOI: 10.1016/j.jvs.2018.02.053] [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] [Received: 11/07/2017] [Accepted: 02/28/2018] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The main objective of this study was to define a role of sphingosine-1-phosphate receptor 1 (S1PR1) in the arterial injury response of a human artery. The hypotheses were tested that injury induces an expansion of S1PR1-positive cells and that these cells accumulate toward the lumen because they follow the sphingosine-1-phosphate gradient from arterial wall tissue (low) to plasma (high). METHODS A humanized rat model was used in which denuded human internal mammary artery (IMA) was implanted into the position of the abdominal aorta of immunosuppressed Rowett nude rats. This injury model is characterized by medial as well as intimal hyperplasia, whereby intimal cells are of human origin. At 7, 14, and 28 days after implantation, grafts were harvested and processed for fluorescent immunostaining for S1PR1 and smooth muscle α-actin. Nuclei were stained with 4',6-diamidine-2'-phenylindole dihydrochloride. Using digitally reconstructed, complete cross sections of grafts, intimal and medial areas were measured, whereby the medial area had virtually been divided into an outer (toward adventitia) and inner (toward lumen) layer. The fraction of S1PR1-positive cells was determined in each layer by counting S1PR1-positive and S1PR1-negative cells. RESULTS The fraction of S1PR1-postive cells in naive IMA is 58.9% ± 6.0% (mean ± standard deviation). At day 28 after implantation, 81.6% ± 4.4% of medial cells were scored S1PR1 positive (P < .01). At day 14, the ratio between S1PR1-positive and S1PR1-negative cells was significantly higher in the lumen-oriented inner layer (9.3 ± 2.1 vs 6.0 ± 1.0; P < .01). Cells appearing in the intima at day 7 and day 14 were almost all S1PR1 positive. At day 28, however, about one-third of intimal cells were scored S1PR1 negative. CONCLUSIONS From these data, we conclude that denudation of IMA specifically induces the expansion of S1PR1-positive cells. Based on the nonrandom distribution of S1PR1-positive cells, we consider the possibility that much like lymphocytes, S1PR1-positive smooth muscle cells also use S1PR1 to recognize the sphingosine-1-phosphate gradient from tissue (low) to plasma (high) and so migrate out of the media toward the intima of the injured IMA.
Collapse
Affiliation(s)
- Julian Braetz
- Clinic and Polyclinic for Vascular Medicine, University Heart Center Hamburg-Eppendorf, Hamburg, Germany; Clinic and Polyclinic for General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Astrid Becker
- Clinic and Polyclinic for Vascular Medicine, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Geissen
- Clinic and Polyclinic for Vascular Medicine, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel Larena-Avellaneda
- Clinic and Polyclinic for Vascular Medicine, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonja Schrepfer
- Clinic and Polyclinic for Cardiovascular Surgery, Transplant and Stem Cell Immunobiology Laboratory, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guenter Daum
- Clinic and Polyclinic for Vascular Medicine, University Heart Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
28
|
Zhao J, Okamoto Y, Asano Y, Ishimaru K, Aki S, Yoshioka K, Takuwa N, Wada T, Inagaki Y, Takahashi C, Nishiuchi T, Takuwa Y. Sphingosine-1-phosphate receptor-2 facilitates pulmonary fibrosis through potentiating IL-13 pathway in macrophages. PLoS One 2018; 13:e0197604. [PMID: 29782549 PMCID: PMC5962071 DOI: 10.1371/journal.pone.0197604] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 05/04/2018] [Indexed: 01/26/2023] Open
Abstract
Idiopathic pulmonary fibrosis is a devastating disease with poor prognosis. The pathogenic role of the lysophospholipid mediator sphingosine-1-phosphate and its receptor S1PR2 in lung fibrosis is unknown. We show here that genetic deletion of S1pr2 strikingly attenuated lung fibrosis induced by repeated injections of bleomycin in mice. We observed by using S1pr2LacZ/+ mice that S1PR2 was expressed in alveolar macrophages, vascular endothelial cells and alveolar epithelial cells in the lung and that S1PR2-expressing cells accumulated in the fibrotic legions. Bone marrow chimera experiments suggested that S1PR2 in bone marrow–derived cells contributes to the development of lung fibrosis. Depletion of macrophages greatly attenuated lung fibrosis. Bleomycin administration stimulated the mRNA expression of the profibrotic cytokines IL-13 and IL-4 and the M2 markers including arginase 1, Fizz1/Retnla, Ccl17 and Ccl24 in cells collected from broncho-alveolar lavage fluids (BALF), and S1pr2 deletion markedly diminished the stimulated expression of these genes. BALF cells from bleomycin–administered wild-type mice showed a marked increase in phosphorylation of STAT6, a transcription factor which is activated downstream of IL-13, compared with saline–administered wild-type mice. Interestingly, in bleomycin–administered S1pr2-/- mice, STAT6 phosphorylation in BALF cells was substantially diminished compared with wild-type mice. Finally, pharmacological S1PR2 blockade in S1pr2+/+ mice alleviated bleomycin–induced lung fibrosis. Thus, S1PR2 facilitates lung fibrosis through the mechanisms involving augmentation of IL-13 expression and its signaling in BALF cells, and represents a novel target for treating lung fibrosis.
Collapse
MESH Headings
- Animals
- Bleomycin/toxicity
- Bronchoalveolar Lavage Fluid/chemistry
- Bronchoalveolar Lavage Fluid/cytology
- Disease Models, Animal
- Idiopathic Pulmonary Fibrosis/etiology
- Idiopathic Pulmonary Fibrosis/metabolism
- Idiopathic Pulmonary Fibrosis/pathology
- Interleukin-13/genetics
- Interleukin-13/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/pathology
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Lysosphingolipid/deficiency
- Receptors, Lysosphingolipid/genetics
- Receptors, Lysosphingolipid/metabolism
- STAT6 Transcription Factor/metabolism
- Signal Transduction
- Sphingosine-1-Phosphate Receptors
- Transplantation Chimera/genetics
- Transplantation Chimera/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Juanjuan Zhao
- Department of Physiology, Kanazawa University School of Medicine, Ishikawa, Japan
| | - Yasuo Okamoto
- Department of Physiology, Kanazawa University School of Medicine, Ishikawa, Japan
| | - Yuya Asano
- Department of Physiology, Kanazawa University School of Medicine, Ishikawa, Japan
| | - Kazuhiro Ishimaru
- Department of Physiology, Kanazawa University School of Medicine, Ishikawa, Japan
| | - Sho Aki
- Department of Physiology, Kanazawa University School of Medicine, Ishikawa, Japan
| | - Kazuaki Yoshioka
- Department of Physiology, Kanazawa University School of Medicine, Ishikawa, Japan
| | - Noriko Takuwa
- Department of Physiology, Kanazawa University School of Medicine, Ishikawa, Japan
- Department of Health and Medical Sciences, Ishikawa Prefectural Nursing University, Ishikawa, Japan
| | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Kanazawa University School of Medicine, Ishikawa, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Kanagawa, Japan
| | - Chiaki Takahashi
- Division of Oncology and Molecular Biology, Cancer Research Institute, Kanazawa University, Ishikawa, Japan
| | - Takumi Nishiuchi
- Division of Functional Genomics, Advanced Science Research Center, Kanazawa University, Ishikawa, Japan
| | - Yoh Takuwa
- Department of Physiology, Kanazawa University School of Medicine, Ishikawa, Japan
- * E-mail:
| |
Collapse
|
29
|
Blanchard O, Stepanovska B, Starck M, Erhardt M, Römer I, Meyer Zu Heringdorf D, Pfeilschifter J, Zangemeister-Wittke U, Huwiler A. Downregulation of the S1P Transporter Spinster Homology Protein 2 (Spns2) Exerts an Anti-Fibrotic and Anti-Inflammatory Effect in Human Renal Proximal Tubular Epithelial Cells. Int J Mol Sci 2018; 19:ijms19051498. [PMID: 29772789 PMCID: PMC5983760 DOI: 10.3390/ijms19051498] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/14/2018] [Accepted: 05/15/2018] [Indexed: 01/09/2023] Open
Abstract
Sphingosine kinase (SK) catalyses the formation of sphingosine 1-phosphate (S1P), which acts as a key regulator of inflammatory and fibrotic reactions, mainly via S1P receptor activation. Here, we show that in the human renal proximal tubular epithelial cell line HK2, the profibrotic mediator transforming growth factor β (TGFβ) induces SK-1 mRNA and protein expression, and in parallel, it also upregulates the expression of the fibrotic markers connective tissue growth factor (CTGF) and fibronectin. Stable downregulation of SK-1 by RNAi resulted in the increased expression of CTGF, suggesting a suppressive effect of SK-1-derived intracellular S1P in the fibrotic process, which is lost when SK-1 is downregulated. In a further approach, the S1P transporter Spns2, which is known to export S1P and thereby reduces intracellular S1P levels, was stably downregulated in HK2 cells by RNAi. This treatment decreased TGFβ-induced CTGF and fibronectin expression, and it abolished the strong induction of the monocyte chemotactic protein 1 (MCP-1) by the pro-inflammatory cytokines tumor necrosis factor (TNF)α and interleukin (IL)-1β. Moreover, it enhanced the expression of aquaporin 1, which is an important water channel that is expressed in the proximal tubules, and reverted aquaporin 1 downregulation induced by IL-1β/TNFα. On the other hand, overexpression of a Spns2-GFP construct increased S1P secretion and it resulted in enhanced TGFβ-induced CTGF expression. In summary, our data demonstrate that in human renal proximal tubular epithelial cells, SK-1 downregulation accelerates an inflammatory and fibrotic reaction, whereas Spns2 downregulation has an opposite effect. We conclude that Spns2 represents a promising new target for the treatment of tubulointerstitial inflammation and fibrosis.
Collapse
Affiliation(s)
- Olivier Blanchard
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010 Bern, Switzerland.
| | - Bisera Stepanovska
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010 Bern, Switzerland.
| | - Manuel Starck
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010 Bern, Switzerland.
| | - Martin Erhardt
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010 Bern, Switzerland.
| | - Isolde Römer
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt am Main, Goethe-University, Theodor-Stern Kai 7, D-60590 Frankfurt am Main, Germany.
| | - Dagmar Meyer Zu Heringdorf
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt am Main, Goethe-University, Theodor-Stern Kai 7, D-60590 Frankfurt am Main, Germany.
| | - Josef Pfeilschifter
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt am Main, Goethe-University, Theodor-Stern Kai 7, D-60590 Frankfurt am Main, Germany.
| | - Uwe Zangemeister-Wittke
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010 Bern, Switzerland.
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010 Bern, Switzerland.
| |
Collapse
|
30
|
Sukocheva OA. Expansion of Sphingosine Kinase and Sphingosine-1-Phosphate Receptor Function in Normal and Cancer Cells: From Membrane Restructuring to Mediation of Estrogen Signaling and Stem Cell Programming. Int J Mol Sci 2018; 19:ijms19020420. [PMID: 29385066 PMCID: PMC5855642 DOI: 10.3390/ijms19020420] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/21/2018] [Accepted: 01/24/2018] [Indexed: 02/05/2023] Open
Abstract
Sphingolipids, sphingolipid metabolizing enzymes, and their receptors network are being recognized as part of the signaling mechanisms, which govern breast cancer cell growth, migration, and survival during chemotherapy treatment. Approximately 70% of breast cancers are estrogen receptor (ER) positive and, thus, rely on estrogen signaling. Estrogen activates an intracellular network composed of many cytoplasmic and nuclear mediators. Some estrogen effects can be mediated by sphingolipids. Estrogen activates sphingosine kinase 1 (SphK1) and amplifies the intracellular concentration of sphingosine-1-phosphate (S1P) in breast cancer cells during stimulation of proliferation and survival. Specifically, Estrogen activates S1P receptors (S1PR) and induces growth factor receptor transactivation. SphK, S1P, and S1PR expression are causally associated with endocrine resistance and progression to advanced tumor stages in ER-positive breast cancers in vivo. Recently, the network of SphK/S1PR was shown to promote the development of ER-negative cancers and breast cancer stem cells, as well as stimulating angiogenesis. Novel findings confirm and broaden our knowledge about the cross-talk between sphingolipids and estrogen network in normal and malignant cells. Current S1PRs therapeutic inhibition was indicated as a promising chemotherapy approach in non-responsive and advanced malignancies. Considering that sphingolipid signaling has a prominent role in terminally differentiated cells, the impact should be considered when designing specific SphK/S1PR inhibitors. This study analyzes the dynamic of the transformation of sphingolipid axis during a transition from normal to pathological condition on the level of the whole organism. The sphingolipid-based mediation and facilitation of global effects of estrogen were critically accented as a bridging mechanism that should be explored in cancer prevention.
Collapse
Affiliation(s)
- Olga A Sukocheva
- College of Nursing and Health Sciences, Flinders University of South Australia, Bedford Park, SA 5042, Australia.
| |
Collapse
|
31
|
Wang YL, Xue P, Xu CY, Wang Z, Liu XS, Hua LL, Bai HY, Zeng ZL, Duan HF, Li JF. SPK1-transfected UCMSC has better therapeutic activity than UCMSC in the treatment of experimental autoimmune encephalomyelitis model of Multiple sclerosis. Sci Rep 2018; 8:1756. [PMID: 29379030 PMCID: PMC5788935 DOI: 10.1038/s41598-018-19703-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/04/2018] [Indexed: 12/11/2022] Open
Abstract
Multiple Sclerosis (MS), is a chronic inflammatory autoimmune disorder of the central nervous system that leads to chronic demyelination with axonal damage and neuronal loss. Mesenchymal stem cells (MSCs) represent a promising therapeutic approach for MS. In the current study, we investigated the effects of MSCs derived from the human umbilical cord (UCMSC) transfected by sphingosine kinase 1 (SPK1) gene. All the results showed that transplantation of UCMSCs gene modified by SPK1 (UCMSC-SPK1) dramatically reduce the severity of neurological deficits of the experimental autoimmune encephalomyelitis (EAE) mice, paralleling by reductions in demyelination, axonal loss, and astrogliosis. UCMSC-SPK1 transplantation also could inhibit the development of natural killer (NK) responses in the spleen of EAE mice, and increase the ratio of CD4+ CD25+ FoxP3+ (Treg) T cells. Furthermore, we described that a shift in the cytokine response from Th1/Th17 to Th2 was an underlying mechanism that suppressed CNS autoimmunity. UCMSCs transfected by SPK1 gene potentially offer a novel mode for the treatment of MS, and the specific mechanism of SPK1 in treating MS/EAE.
Collapse
Affiliation(s)
- Yun-Liang Wang
- Department of Neurology, the Second Affiliated Hospital of Zhengzhou University, No. 32 Nanyang Road, Zhengzhou, 450014, China.,Department of Neurology, the 148th Hospital of Chinese PLA, No. 20 North Road Zhoucun District, Zibo, 255300, China
| | - Peng Xue
- Department of Neurology, the Second Affiliated Hospital of Zhengzhou University, No. 32 Nanyang Road, Zhengzhou, 450014, China
| | - Chun-Yang Xu
- Department of Neurology, the Second Affiliated Hospital of Zhengzhou University, No. 32 Nanyang Road, Zhengzhou, 450014, China
| | - Zhen Wang
- Department of Neurology, the 148th Hospital of Chinese PLA, No. 20 North Road Zhoucun District, Zibo, 255300, China
| | - Xin-Shan Liu
- Electroencephalogram Room of Sanbo Brain Hospital, Capital Medical University, No. 50 Xiangshanyikesong Haidian District, Beijing, 100093, China
| | - Lin-Lin Hua
- Department of Neurology, the Second Affiliated Hospital of Zhengzhou University, No. 32 Nanyang Road, Zhengzhou, 450014, China
| | - Hong-Ying Bai
- Department of Neurology, the Second Affiliated Hospital of Zhengzhou University, No. 32 Nanyang Road, Zhengzhou, 450014, China
| | - Zhi-Lei Zeng
- Department of Neurology, the Second Affiliated Hospital of Zhengzhou University, No. 32 Nanyang Road, Zhengzhou, 450014, China
| | - Hai-Feng Duan
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Jin-Feng Li
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
32
|
Huwiler A, Pfeilschifter J. Sphingolipid signaling in renal fibrosis. Matrix Biol 2018; 68-69:230-247. [PMID: 29343457 DOI: 10.1016/j.matbio.2018.01.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 12/28/2022]
Abstract
Over the last decade, various sphingolipid subspecies have gained increasing attention as important signaling molecules that regulate a multitude of physiological and pathophysiological processes including inflammation and tissue remodeling. These mediators include ceramide, sphingosine 1-phosphate (S1P), the cerebroside glucosylceramide, lactosylceramide, and the gangliosides GM3 and Gb3. These lipids have been shown to accumulate in various chronic kidney diseases that typically end in renal fibrosis and ultimately renal failure. This review will summarize the effects and contributions of those enzymes that regulate the generation and interconversion of these lipids, notably the acid sphingomyelinase, the acid sphingomyelinase-like protein SMPDL3B, the sphingosine kinases, the S1P lyase, the glucosylceramide synthase, the GM3 synthase, and the α-galactosidase A, to renal fibrotic diseases. Strategies of manipulating these enzymes for therapeutic purposes and the impact of existing drugs on renal pathologies will be discussed.
Collapse
Affiliation(s)
- Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland.
| | - Josef Pfeilschifter
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe- University, Frankfurt am Main, Germany
| |
Collapse
|
33
|
Schwalm S, Beyer S, Frey H, Haceni R, Grammatikos G, Thomas D, Geisslinger G, Schaefer L, Huwiler A, Pfeilschifter J. Sphingosine Kinase-2 Deficiency Ameliorates Kidney Fibrosis by Up-Regulating Smad7 in a Mouse Model of Unilateral Ureteral Obstruction. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2413-2429. [DOI: 10.1016/j.ajpath.2017.06.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/10/2017] [Accepted: 06/29/2017] [Indexed: 12/31/2022]
|
34
|
González-Fernández B, Sánchez DI, González-Gallego J, Tuñón MJ. Sphingosine 1-Phosphate Signaling as a Target in Hepatic Fibrosis Therapy. Front Pharmacol 2017; 8:579. [PMID: 28890699 PMCID: PMC5574909 DOI: 10.3389/fphar.2017.00579] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/10/2017] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is an excess production of extracellular matrix proteins as a result of chronic liver disease which leads to cell death and organ dysfunction. The key cells involved in fibrogenesis are resident hepatic stellate cells (HSCs) which are termed myofibroblasts after activation, acquiring contractile, proliferative, migratory and secretory capability. Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid with well-established effects on angiogenesis, carcinogenesis and immunity. Accumulating evidence demonstrates that this metabolite is involved in the profibrotic inflammatory process through the regulation of pleiotropic cell responses, such as vascular permeability, leukocyte infiltration, cell survival, migration, proliferation and HSCs differentiation to myofibroblasts. S1P is synthesized by sphingosine kinases (SphKs) and many of its actions are mediated by S1P specific cell surface receptors (S1P1-5), although different intracellular targets of S1P have been identified. Modulation of SphKs/S1P/S1P receptors signaling is known to result in beneficial effects on various in vivo and in vitro models of liver fibrosis. Thus, a better knowledge of the molecular mechanisms involved in the modulation of the S1P pathway could help to improve liver fibrosis therapy. In this review, we analyze the effects of the S1P axis on the fibrogenic process, and the involvement of a range of inhibitors or approaches targeting enzymes related to S1P in the abrogation of pathological fibrogenesis. All in all, targeting this pathway offers therapeutic potential in the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
| | | | - Javier González-Gallego
- Institute of Biomedicine, University of LeónLeón, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain
| | - María J Tuñón
- Institute of Biomedicine, University of LeónLeón, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain
| |
Collapse
|
35
|
Li S, Xu P, Han L, Mao W, Wang Y, Luo G, Yang N. Disease-syndrome combination modeling: metabolomic strategy for the pathogenesis of chronic kidney disease. Sci Rep 2017; 7:8830. [PMID: 28821830 PMCID: PMC5562836 DOI: 10.1038/s41598-017-09311-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/25/2017] [Indexed: 01/26/2023] Open
Abstract
Conventional disease animal models have limitations on the conformity to the actual clinical situation. Disease-syndrome combination (DS) modeling may provide a more efficient strategy for biomedicine research. Disease model and DS model of renal fibrosis in chronic kidney disease were established by ligating the left ureter and by ligating unilateral ureteral combined with exhaustive swimming, respectively. Serum metabolomics was conducted to evaluate disease model and DS model by using ultra performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. Potential endogenous biomarkers were identified by multivariate statistical analysis. There are no differences between two models regarding their clinical biochemistry and kidney histopathology, while metabolomics highlights their difference. It is found that abnormal sphingolipid metabolism is a common characteristic of both models, while arachidonic acid metabolism, linolenic acid metabolism and glycerophospholipid metabolism are highlighted in DS model. Metabolomics is a promising approach to evaluate experiment animal models. DS model are comparatively in more coincidence with clinical settings, and is superior to single disease model for the biomedicine research.
Collapse
Affiliation(s)
- Shasha Li
- Guangdong Provincial Hospital of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong, 510120, China
| | - Peng Xu
- Guangdong Provincial Hospital of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong, 510120, China
| | - Ling Han
- Guangdong Provincial Hospital of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong, 510120, China
| | - Wei Mao
- Guangdong Provincial Hospital of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong, 510120, China
| | - Yiming Wang
- Guangdong Provincial Hospital of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong, 510120, China
- Department of Chemistry, Tsinghua University, No. 30 Shuangqing Road in Haidian Distric, Beijing, 100084, China
| | - Guoan Luo
- Guangdong Provincial Hospital of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong, 510120, China.
- Department of Chemistry, Tsinghua University, No. 30 Shuangqing Road in Haidian Distric, Beijing, 100084, China.
| | - Nizhi Yang
- Guangdong Provincial Hospital of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
36
|
Zhao YD, Yin L, Archer S, Lu C, Zhao G, Yao Y, Wu L, Hsin M, Waddell TK, Keshavjee S, Granton J, de Perrot M. Metabolic heterogeneity of idiopathic pulmonary fibrosis: a metabolomic study. BMJ Open Respir Res 2017; 4:e000183. [PMID: 28883924 PMCID: PMC5531310 DOI: 10.1136/bmjresp-2017-000183] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 11/28/2022] Open
Abstract
Introduction Idiopathic pulmonary fibrosis (IPF) is a chronic and fatal disease of unknown cause characterised by progressive fibrotic formation in lung tissue. We hypothesise that disrupted metabolic pathways in IPF contribute to disease pathogenesis. Methods Metabolomics of human IPF was performed using mass spectroscopy (IPF lung=8; donor lung=8). Gene expression of key metabolic enzymes was measured using microarrays. Of the 108 metabolites whose levels were found altered, 48 were significantly increased, whereas 60 were significantly decreased in IPF samples compared with normal controls. Results Specific metabolic pathways mediating the IPF remodelling were found with a downregulated sphingolipid metabolic pathway but an upregulated arginine pathway in IPF. In addition, disrupted glycolysis, mitochondrial beta-oxidation and tricarboxylic acid cycle, altered bile acid, haem and glutamate/aspartate metabolism were found in IPF samples compared with control. Conclusions Our results show alterations in metabolic pathways for energy consumption during lung structural remodelling, which may contribute to IPF pathogenesis. We believe that this is the first report of simultaneously and systemically measuring changes of metabolites involving nine metabolic pathways in human severe IPF lungs. The measurement of the metabolites may serve in the future diagnosis and prognosis of IPF.
Collapse
Affiliation(s)
- Yidan D Zhao
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Li Yin
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Stephen Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Catherine Lu
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - George Zhao
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Yan Yao
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Licun Wu
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Michael Hsin
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Thomas K Waddell
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| | - John Granton
- Division of Respirology, University Health Network, Toronto, Ontario, Canada
| | - Marc de Perrot
- Department of Thoracic Surgery, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
37
|
Vestri A, Pierucci F, Frati A, Monaco L, Meacci E. Sphingosine 1-Phosphate Receptors: Do They Have a Therapeutic Potential in Cardiac Fibrosis? Front Pharmacol 2017. [PMID: 28626422 PMCID: PMC5454082 DOI: 10.3389/fphar.2017.00296] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid that is characterized by a peculiar mechanism of action. In fact, S1P, which is produced inside the cell, can act as an intracellular mediator, whereas after its export outside the cell, it can act as ligand of specific G-protein coupled receptors, which were initially named endothelial differentiation gene (Edg) and eventually renamed sphingosine 1-phosphate receptors (S1PRs). Among the five S1PR subtypes, S1PR1, S1PR2 and S1PR3 isoforms show broad tissue gene expression, while S1PR4 is primarily expressed in immune system cells, and S1PR5 is expressed in the central nervous system. There is accumulating evidence for the important role of S1P as a mediator of many processes, such as angiogenesis, carcinogenesis and immunity, and, ultimately, fibrosis. After a tissue injury, the imbalance between the production of extracellular matrix (ECM) and its degradation, which occurs due to chronic inflammatory conditions, leads to an accumulation of ECM and, consequential, organ dysfunction. In these pathological conditions, many factors have been described to act as pro- and anti-fibrotic agents, including S1P. This bioactive lipid exhibits both pro- and anti-fibrotic effects, depending on its site of action. In this review, after a brief description of sphingolipid metabolism and signaling, we emphasize the involvement of the S1P/S1PR axis and the downstream signaling pathways in the development of fibrosis. The current knowledge of the therapeutic potential of S1PR subtype modulators in the treatment of the cardiac functions and fibrinogenesis are also examined.
Collapse
Affiliation(s)
- Ambra Vestri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy
| | - Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy.,Interuniversity Institutes of MyologyFirenze, Italy
| | - Alessia Frati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy
| | - Lucia Monaco
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of RomeRome, Italy
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy.,Interuniversity Institutes of MyologyFirenze, Italy
| |
Collapse
|
38
|
Childress ES, Kharel Y, Brown AM, Bevan DR, Lynch KR, Santos WL. Transforming Sphingosine Kinase 1 Inhibitors into Dual and Sphingosine Kinase 2 Selective Inhibitors: Design, Synthesis, and in Vivo Activity. J Med Chem 2017; 60:3933-3957. [PMID: 28406646 PMCID: PMC6047346 DOI: 10.1021/acs.jmedchem.7b00233] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a pleiotropic signaling molecule that interacts with its five G-protein coupled receptors (S1P1-5) to regulate cell growth and survival and has been implicated in a variety of diseases including cancer and sickle cell disease. As the key mediators in the synthesis of S1P, sphingosine kinase (SphK) isoforms 1 and 2 have attracted attention as viable targets for pharmaceutical inhibition. In this article, we describe the design, synthesis, and biological evaluation of aminothiazole-based guanidine inhibitors of SphK. Surprisingly, combining features of reported SphK1 inhibitors generated SphK1/2 dual inhibitor 20l (SLC4011540) (hSphK1 Ki = 120 nM, hSphK2 Ki = 90 nM) and SphK2 inhibitor 20dd (SLC4101431) (Ki = 90 nM, 100-fold SphK2 selectivity). These compounds effectively decrease S1P levels in vitro. In vivo administration of 20dd validated that inhibition of SphK2 increases blood S1P levels.
Collapse
Affiliation(s)
- Elizabeth S. Childress
- Department of Chemistry and VT Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Anne M. Brown
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - David R. Bevan
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Kevin R. Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Webster L. Santos
- Department of Chemistry and VT Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
39
|
Gendron DR, Lemay AM, Lecours PB, Perreault-Vallières V, Huppé CA, Bossé Y, Blanchet MR, Dion G, Marsolais D. FTY720 promotes pulmonary fibrosis when administered during the remodelling phase following a bleomycin-induced lung injury. Pulm Pharmacol Ther 2017; 44:50-56. [PMID: 28323056 DOI: 10.1016/j.pupt.2017.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 12/22/2016] [Accepted: 03/13/2017] [Indexed: 12/28/2022]
Abstract
Fibrosis complicates numerous pathologies including interstitial lung diseases. Sphingosine analogs such as FTY720 can alleviate lung injury-induced fibrosis in murine models. Contradictorily, FTY720 also promotes in vitro processes normally leading to fibrosis and high doses in vivo foster lung fibrosis by enhancing vascular leakage into the lung. The goal of this study was to determine the effect of low doses of FTY720 on lung fibrosis triggered by an acute injury in mice. We first defined the time-boundaries delimiting the inflammatory and remodelling phases of an injury elicited by bleomycin based on neutrophil counts, total lung capacity and lung stiffness. Thereafter, FTY720 (0.1 mg/kg) was delivered during either the inflammatory or the remodelling phases of bleomycin-induced injury. While FTY720 decreased fibrosis by 60% and lung stiffness by 28% when administered during the inflammatory phase, it increased fibrosis (2.1-fold) and lung stiffness (1.7-fold) when administered during the remodelling phase. FTY720-induced worsening of fibrosis was associated with an increased expression of connective tissue growth factor, but not with vascular leakage into the lung. Thus, the timing of FTY720 delivery following a bleomycin-induced lung injury determines pro-vs anti-fibrotic outcomes.
Collapse
Affiliation(s)
- David R Gendron
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec City, Québec, Canada
| | - Anne-Marie Lemay
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec City, Québec, Canada
| | - Pascale Blais Lecours
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec City, Québec, Canada
| | - Valérie Perreault-Vallières
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec City, Québec, Canada
| | - Carole-Ann Huppé
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec City, Québec, Canada
| | - Ynuk Bossé
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec City, Québec, Canada; Département de Médecine, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Marie-Renée Blanchet
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec City, Québec, Canada; Département de Médecine, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Geneviève Dion
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec City, Québec, Canada; Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), Québec City, Québec, Canada
| | - David Marsolais
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec City, Québec, Canada; Département de Médecine, Faculté de Médecine, Université Laval, Québec City, Québec, Canada.
| |
Collapse
|
40
|
Hsieh LTH, Nastase MV, Roedig H, Zeng-Brouwers J, Poluzzi C, Schwalm S, Fork C, Tredup C, Brandes RP, Wygrecka M, Huwiler A, Pfeilschifter J, Schaefer L. Biglycan- and Sphingosine Kinase-1 Signaling Crosstalk Regulates the Synthesis of Macrophage Chemoattractants. Int J Mol Sci 2017; 18:ijms18030595. [PMID: 28282921 PMCID: PMC5372611 DOI: 10.3390/ijms18030595] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 02/27/2017] [Accepted: 03/06/2017] [Indexed: 12/14/2022] Open
Abstract
In its soluble form, the extracellular matrix proteoglycan biglycan triggers the synthesis of the macrophage chemoattractants, chemokine (C-C motif) ligand CCL2 and CCL5 through selective utilization of Toll-like receptors (TLRs) and their adaptor molecules. However, the respective downstream signaling events resulting in biglycan-induced CCL2 and CCL5 production have not yet been defined. Here, we show that biglycan stimulates the production and activation of sphingosine kinase 1 (SphK1) in a TLR4- and Toll/interleukin (IL)-1R domain-containing adaptor inducing interferon (IFN)-β (TRIF)-dependent manner in murine primary macrophages. We provide genetic and pharmacological proof that SphK1 is a crucial downstream mediator of biglycan-triggered CCL2 and CCL5 mRNA and protein expression. This is selectively driven by biglycan/SphK1-dependent phosphorylation of the nuclear factor NF-κB p65 subunit, extracellular signal-regulated kinase (Erk)1/2 and p38 mitogen-activated protein kinases. Importantly, in vivo overexpression of soluble biglycan causes Sphk1-dependent enhancement of renal CCL2 and CCL5 and macrophage recruitment into the kidney. Our findings describe the crosstalk between biglycan- and SphK1-driven extracellular matrix- and lipid-signaling. Thus, SphK1 may represent a new target for therapeutic intervention in biglycan-evoked inflammatory conditions.
Collapse
Affiliation(s)
- Louise Tzung-Harn Hsieh
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Madalina-Viviana Nastase
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
- National Institute for Chemical-Pharmaceutical Research and Development, 112 Vitan Avenue, Bucharest 031299, Romania.
| | - Heiko Roedig
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Jinyang Zeng-Brouwers
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Chiara Poluzzi
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Stephanie Schwalm
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Christian Fork
- Institut für Kardiovaskulare Physiologie, Klinikum der Goethe-Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Claudia Tredup
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Ralf P Brandes
- Institut für Kardiovaskulare Physiologie, Klinikum der Goethe-Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Friedrichstrasse 24, Giessen 35392, Germany.
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital INO-F, Bern CH-3010, Switzerland.
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Liliana Schaefer
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| |
Collapse
|
41
|
Characterization of cholesterol homeostasis in sphingosine-1-phosphate lyase-deficient fibroblasts reveals a Niemann-Pick disease type C-like phenotype with enhanced lysosomal Ca 2+ storage. Sci Rep 2017; 7:43575. [PMID: 28262793 PMCID: PMC5337937 DOI: 10.1038/srep43575] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/25/2017] [Indexed: 02/08/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) lyase irreversibly cleaves S1P, thereby catalysing the ultimate step of sphingolipid degradation. We show here that embryonic fibroblasts from S1P lyase-deficient mice (Sgpl1−/−-MEFs), in which S1P and sphingosine accumulate, have features of Niemann-Pick disease type C (NPC) cells. In the presence of serum, overall cholesterol content was elevated in Sgpl1−/−-MEFs, due to upregulation of the LDL receptor and enhanced cholesterol uptake. Despite this, activation of sterol regulatory element-binding protein-2 was increased in Sgpl1−/−-MEFs, indicating a local lack of cholesterol at the ER. Indeed, free cholesterol was retained in NPC1-containing vesicles, which is a hallmark of NPC. Furthermore, upregulation of amyloid precursor protein in Sgpl1−/−-MEFs was mimicked by an NPC1 inhibitor in Sgpl1+/+-MEFs and reduced by overexpression of NPC1. Lysosomal pH was not altered by S1P lyase deficiency, similar to NPC. Interestingly, lysosomal Ca2+ content and bafilomycin A1-induced [Ca2+]i increases were enhanced in Sgpl1−/−-MEFs, contrary to NPC. These results show that both a primary defect in cholesterol trafficking and S1P lyase deficiency cause overlapping phenotypic alterations, and challenge the present view on the role of sphingosine in lysosomal Ca2+ homeostasis.
Collapse
|
42
|
González-Fernández B, Sánchez DI, Crespo I, San-Miguel B, Álvarez M, Tuñón MJ, González-Gallego J. Inhibition of the SphK1/S1P signaling pathway by melatonin in mice with liver fibrosis and human hepatic stellate cells. Biofactors 2017; 43:272-282. [PMID: 27801960 DOI: 10.1002/biof.1342] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 09/07/2016] [Accepted: 10/03/2016] [Indexed: 01/02/2023]
Abstract
The sphingosine kinase 1/sphingosine 1-phosphate (SphK1/S1P) system is involved in different pathological processes, including fibrogenesis. Melatonin abrogates activation of hepatic stellate cells (HSCs) and attenuates different profibrogenic pathways in animal models of fibrosis, but it is unknown if protection associates with its inhibitory effect on the SphK1/S1P axis. Mice in treatment groups received carbon tetrachloride (CCl4 ) 5 μL g-1 body wt i.p. twice a week for 4 or 6 weeks. Melatonin was given at 5 or 10 mg kg-1 day-1 i.p, beginning 2 weeks after the start of CCl4 administration. At both 4 and 6 weeks following CCl4 treatment, liver mRNA levels, protein concentration and immunohistochemical labelling for SphK1 increased significantly. S1P production, and expression of S1P receptor (S1PR)1, S1PR3 and acid sphingomyelinase (ASMase) were significantly elevated. However, there was a decreased expression of S1PR2 and S1P lyase (S1PL). Melatonin attenuated liver fibrosis, as shown by a significant inhibition of the expression of α-smooth muscle actin (α-SMA), transforming growth factor (TGF)-β and collagen (Col) Ι. Furthermore, melatonin inhibited S1P production, lowered expression of SphK1, S1PR1, SP1R3, and ASMase, and increased expression of S1PL. Melatonin induced a reversal of activated human HSCs cell line LX2, as evidenced by a reduction in α-SMA, TGF-β, and Col I expression. Melatonin-treated cells also exhibited an inhibition of the SphK1/S1P axis. Antifibrogenic effect of SphK1 inhibition was confirmed by treatment of LX2 cells with PF543. Abrogation of the lipid signaling pathway by the indole reveals novel molecular pathways that may account for the protective effect of melatonin in liver fibrogenesis. © 2016 BioFactors, 43(2):272-282, 2017.
Collapse
Affiliation(s)
| | - Diana I Sánchez
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | - Irene Crespo
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), León, Spain
| | | | | | - María J Tuñón
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), León, Spain
| | - Javier González-Gallego
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), León, Spain
| |
Collapse
|
43
|
Serum lipid alterations identified in chronic hepatitis B, hepatitis B virus-associated cirrhosis and carcinoma patients. Sci Rep 2017; 7:42710. [PMID: 28198443 PMCID: PMC5309878 DOI: 10.1038/srep42710] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/12/2017] [Indexed: 01/03/2023] Open
Abstract
The incidences of chronic hepatitis B (CHB), Hepatitis B virus (HBV)-associated cirrhosis and HBV-associated carcinoma are high and increasing. This study was designed to evaluate serum lipid metabolite changes that are associated with the progression from CHB to HBV-associated cirrhosis and ultimately to HBV-associated HCC. A targeted metabolomic assay was performed in fasting sera from 136 CHB patients, 104 HBV-associated cirrhosis, and 95 HBV-associated HCC using ultra-performance liquid chromatography triple quadrupole mass spectrometry. A total of 140 metabolites were identified. Clear separations between each two groups were obtained using the partial least squares discriminate analysis of 9 lipid metabolites. Progressively lower levels of long-chain lysophosphatidylcholines (lysoPC a C18:2, lysoPC a C20:3, lysoPC a C20:4) were observed from CHB to cirrhosis to carcinoma; lower levels of lysoPC a C20:4 were found in patients with higher model for end-stage liver disease in the same disease group; and lysoPC a C20:3 levels were lower in Child-Pugh Class C than in Class A and Class B in HBV-associated cirrhosis and HBV-associated HCC groups. The octadecadienyl carnitine level was higher in HBV-associated cirrhosis group than in other two groups. Serum levels of selected long-chain lysoPCs are promising markers for the progression of HBV-associated liver diseases.
Collapse
|
44
|
Luo J, Zhang M, Huang H, Wang Y, Yuan X, Ma S, Liu J, Zhou S, Zhang S. Matrilin-2 regulates proliferation, apoptosis and cell cycle during radiation-induced injury in HPAEpiC cell. Biochem Biophys Res Commun 2016; 485:577-583. [PMID: 27923659 DOI: 10.1016/j.bbrc.2016.12.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 12/03/2016] [Indexed: 12/14/2022]
Abstract
Radiation pulmonary injury is related to the accumulation of extracellular matrix proteins in the alveolar interstitial space. Matrilin-2 as a component of extracellular filamentous networks, present higher level in the lung tissue from irradiated mice and irradiated pulmonary epithelial cell line, HPAEpiC cells. Knockdown of endogenous matrilin-2 prevents the apoptosis of HPAEpiC cell induced by the irradiation injury. Consistently, over-expression of matrilin-2 reduced the proliferation and induced apoptosis of HPAEpiC cells. Matrilin-2 promotes the expression of p21 via increasing the transcriptional activity of p53, by which induces the G1 phase arresting in HPAEpiC cells. In summary, matrilin-2, increased by irradiation, reduced the proliferation and induces apoptosis of pulmonary epithelial cells via p53/p21 pathway.
Collapse
Affiliation(s)
- Junming Luo
- Department of Pathology, Qinghai People's Provincial Hospital, Xining 810007, Qinghai Province, People's Republic of China
| | - Menglan Zhang
- Department of Pathology, Qinghai People's Provincial Hospital, Xining 810007, Qinghai Province, People's Republic of China
| | - Hong Huang
- Department of Pathology, Qinghai People's Provincial Hospital, Xining 810007, Qinghai Province, People's Republic of China
| | - Yichun Wang
- Department of Anesthesiology, Hunan Provincial Tumor Hospital, Central South University, Changsha 410013, Hunan Province, People's Republic of China
| | - Xin Yuan
- Department of Internal Medicine, Qinghai People's Provincial Hospital, Xining 810007, Qinghai Province, People's Republic of China
| | - Siqing Ma
- Department of Critical Care Medicine, Qinghai People's Provincial Hospital, Xining 810007, Qinghai Province, People's Republic of China
| | - Jingshi Liu
- Department of Anesthesiology, Hunan Provincial Tumor Hospital, Central South University, Changsha 410013, Hunan Province, People's Republic of China
| | - Shiying Zhou
- Department of Pathology, Qinghai People's Provincial Hospital, Xining 810007, Qinghai Province, People's Republic of China
| | - Shukun Zhang
- Department of Pathology, Qinghai People's Provincial Hospital, Xining 810007, Qinghai Province, People's Republic of China.
| |
Collapse
|
45
|
Bajwa A, Huang L, Kurmaeva E, Ye H, Dondeti KR, Chroscicki P, Foley LS, Balogun ZA, Alexander KJ, Park H, Lynch KR, Rosin DL, Okusa MD. Sphingosine Kinase 2 Deficiency Attenuates Kidney Fibrosis via IFN- γ. J Am Soc Nephrol 2016; 28:1145-1161. [PMID: 27799486 DOI: 10.1681/asn.2016030306] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 09/11/2016] [Indexed: 12/17/2022] Open
Abstract
Maladaptive repair after AKI may lead to progressive fibrosis and decline in kidney function. Sphingosine 1-phosphate has an important role in kidney injury and pleiotropic effects in fibrosis. We investigated the involvement of sphingosine kinase 1 and 2 (SphK1 and SphK2), which phosphorylate sphingosine to produce sphingosine 1-phosphate, in kidney fibrosis induced by folic acid (FA) or unilateral ischemia-reperfusion injury. Analysis of Masson trichrome staining and fibrotic marker protein and mRNA expression 14 days after AKI revealed that wild-type (WT) and Sphk1-/- mice exhibited more kidney fibrosis than Sphk2-/- mice. Furthermore, kidneys of FA-treated WT and Sphk1-/- mice had greater immune cell infiltration and expression of fibrotic and inflammatory markers than kidneys of FA-treated Sphk2-/- mice. In contrast, kidneys of Sphk2-/- mice exhibited greater expression of Ifng and IFN-γ-responsive genes (Cxcl9 and Cxcl10) than kidneys of WT or Sphk1-/- mice did at this time point. Splenic T cells from untreated Sphk2-/- mice were hyperproliferative and produced more IFN-γ than did those of WT or Sphk1-/- mice. IFN-γ blocking antibody administered to Sphk2-/- mice or deletion of Ifng (Sphk2-/-Ifng-/- mice) blocked the protective effect of SphK2 deficiency in fibrosis. Moreover, adoptive transfer of Sphk2-/- (but not Sphk2-/-Ifng-/- ) CD4 T cells into WT mice blocked FA-induced fibrosis. Finally, a selective SphK2 inhibitor blocked FA-induced kidney fibrosis in WT mice. These studies demonstrate that SphK2 inhibition may serve as a novel therapeutic approach for attenuating kidney fibrosis.
Collapse
Affiliation(s)
- Amandeep Bajwa
- Division of Nephrology, .,Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and
| | - Liping Huang
- Division of Nephrology.,Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and
| | - Elvira Kurmaeva
- Division of Nephrology.,Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and
| | - Hong Ye
- Division of Nephrology.,Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and
| | - Krishna R Dondeti
- Division of Nephrology.,Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and
| | - Piotr Chroscicki
- Division of Nephrology.,Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and
| | - Leah S Foley
- Division of Nephrology.,Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and
| | - Z Ayoade Balogun
- Division of Nephrology.,Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and
| | - Kyle J Alexander
- Division of Nephrology.,Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and
| | - Hojung Park
- Division of Nephrology.,Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and
| | - Kevin R Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Diane L Rosin
- Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and .,Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Mark D Okusa
- Division of Nephrology.,Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, and
| |
Collapse
|
46
|
Chang N, Ge J, Xiu L, Zhao Z, Duan X, Tian L, Xie J, Yang L, Li L. HuR mediates motility of human bone marrow-derived mesenchymal stem cells triggered by sphingosine 1-phosphate in liver fibrosis. J Mol Med (Berl) 2016; 95:69-82. [PMID: 27543493 DOI: 10.1007/s00109-016-1460-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/14/2016] [Accepted: 08/10/2016] [Indexed: 12/12/2022]
Abstract
Sphingosine 1-phosphate (S1P) participates in migration of bone marrow (BM)-derived mesenchymal stem cells (BMSCs) toward damaged liver via upregulation of S1P receptor 3 (S1PR3) during mouse liver fibrogenesis. But, the molecular mechanism is still unclear. HuR, as an RNA-binding protein, regulates tumor cell motility. Here, we examined the role of HuR in migration of human BMSCs (hBMSCs) in liver fibrosis. Results showed that HuR messenger RNA (mRNA) level was increased in human or mouse fibrotic livers, and correlated with S1PR3 mRNA expression. Using immunofluorescence, we found that HuR mainly localized in the nuclei of hepatocytes and non-parenchymal cells in normal livers. However, in fibrotic livers, we detected an increased HuR cytoplasmic localization in non-parenchymal cells. In chimeric mice of BM cell-labeled by EGFP, significant numbers of EGFP-positive cells (BM origin) were positive for HuR in fibrotic areas. Meanwhile, HuR-positive cells were also positive for α-SMA (myofibroblasts). In vitro, S1P induced hBMSCs migration via S1PR3 upregulation. HuR involved in S1P-induced hBMSCs migration and increased stabilization of S1PR3 mRNA via competing with miR-30e. RNA immunoprecipitation showed that HuR interacted with S1PR3 mRNA 3'UTR. Moreover, S1P resulted in phosphorylation and cytoplasmic translocation of HuR via S1PR3 and p38MAPK. Furthermore, we transplanted EGFP+ BMSCs with or without HuR small interfering RNA (siRNA) into carbon tetrachloride-treated mice and found that knockdown of HuR inhibited the migration of BMSCs toward injured livers by flow cytometric analysis in vivo. We identified a positive feedback regulation mechanism between HuR and S1PR3 in S1P-induced BMSCs migration. HuR participates in upregulation of S1PR3 induced by S1P. S1P results in phosphorylation and translocation of HuR via S1PR3. Our results provide a new regulatory manner to the mechanism of liver fibrogenesis. KEY MESSAGE HuR expression and cytoplasmic localization were increased in fibrotic livers. S1P induced migration of human bone marrow Mesenchymal Stem Cells via S1PR3 and HuR. HuR regulated S1PR3 mRNA expression by binding with S1PR3 mRNA 3'UTR. S1P induced HuR phosphorylation and cytoplasmic translocation via S1PR3. HuR regulated S1PR3 expression by competing with miR-30e.
Collapse
Affiliation(s)
- Na Chang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Jingjing Ge
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Lei Xiu
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Zhongxin Zhao
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Xianghui Duan
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Lei Tian
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Jieshi Xie
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Lin Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Liying Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China.
| |
Collapse
|
47
|
Blankenbach KV, Schwalm S, Pfeilschifter J, Meyer Zu Heringdorf D. Sphingosine-1-Phosphate Receptor-2 Antagonists: Therapeutic Potential and Potential Risks. Front Pharmacol 2016; 7:167. [PMID: 27445808 PMCID: PMC4914510 DOI: 10.3389/fphar.2016.00167] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/03/2016] [Indexed: 12/26/2022] Open
Abstract
The sphingosine-1-phosphate (S1P) signaling system with its specific G-protein-coupled S1P receptors, the enzymes of S1P metabolism and the S1P transporters, offers a multitude of promising targets for drug development. Until today, drug development in this area has nearly exclusively focused on (functional) antagonists at the S1P1 receptor, which cause a unique phenotype of immunomodulation. Accordingly, the first-in class S1P1 receptor modulator, fingolimod, has been approved for the treatment of relapsing-remitting multiple sclerosis, and novel S1P1 receptor (functional) antagonists are being developed for autoimmune and inflammatory diseases such as psoriasis, inflammatory bowel disease, lupus erythematodes, or polymyositis. Besides the S1P1 receptor, also S1P2 and S1P3 are widely expressed and regulate many diverse functions throughout the body. The S1P2 receptor, in particular, often exerts cellular functions which are opposed to the functions of the S1P1 receptor. As a consequence, antagonists at the S1P2 receptor have the potential to be useful in a contrasting context and different areas of indication compared to S1P1 antagonists. The present review will focus on the therapeutic potential of S1P2 receptor antagonists and discuss their opportunities as well as their potential risks. Open questions and areas which require further investigations will be emphasized in particular.
Collapse
Affiliation(s)
- Kira V Blankenbach
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Stephanie Schwalm
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Dagmar Meyer Zu Heringdorf
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| |
Collapse
|
48
|
Meshcheryakova A, Svoboda M, Tahir A, Köfeler HC, Triebl A, Mungenast F, Heinze G, Gerner C, Zimmermann P, Jaritz M, Mechtcheriakova D. Exploring the role of sphingolipid machinery during the epithelial to mesenchymal transition program using an integrative approach. Oncotarget 2016; 7:22295-323. [PMID: 26967245 PMCID: PMC5008362 DOI: 10.18632/oncotarget.7947] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 02/20/2016] [Indexed: 12/30/2022] Open
Abstract
The epithelial to mesenchymal transition (EMT) program is activated in epithelial cancer cells and facilitates their ability to metastasize based on enhanced migratory, proliferative, anti-apoptotic, and pluripotent capacities. Given the fundamental impact of sphingolipid machinery to each individual process, the sphingolipid-related mechanisms might be considered among the most prominent drivers/players of EMT; yet, there is still limited knowledge. Given the complexity of the interconnected sphingolipid system, which includes distinct sphingolipid mediators, their synthesizing enzymes, receptors and transporters, we herein apply an integrative approach for assessment of the sphingolipid-associated mechanisms underlying EMT program. We created the sphingolipid-/EMT-relevant 41-gene/23-gene signatures which were applied to denote transcriptional events in a lung cancer cell-based EMT model. Based on defined 35-gene sphingolipid/EMT-attributed signature of regulated genes, we show close associations between EMT markers, genes comprising the sphingolipid network at multiple levels and encoding sphingosine 1-phosphate (S1P)-/ceramide-metabolizing enzymes, S1P and lysophosphatidic acid (LPA) receptors and S1P transporters, pluripotency genes and inflammation-related molecules, and demonstrate the underlying biological pathways and regulators. Mass spectrometry-based sphingolipid analysis revealed an EMT-attributed shift towards increased S1P and LPA accompanied by reduced ceramide levels. Notably, using transcriptomics data across various cell-based perturbations and neoplastic tissues (24193 arrays), we identified the sphingolipid/EMT signature primarily in lung adenocarcinoma tissues; besides, bladder, colorectal and prostate cancers were among the top-ranked. The findings also highlight novel regulatory associations between influenza virus and the sphingolipid/EMT-associated mechanisms. In sum, data propose the multidimensional contribution of sphingolipid machinery to pathological EMT and may yield new biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Anastasia Meshcheryakova
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Martin Svoboda
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Ammar Tahir
- Institute of Analytical Chemistry, University of Vienna, Vienna, Austria
- Mass Spectrometry Center, University of Vienna, Vienna, Austria
| | - Harald C. Köfeler
- Core Facility for Mass Spectrometry, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Alexander Triebl
- Core Facility for Mass Spectrometry, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Felicitas Mungenast
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Georg Heinze
- Section for Clinical Biometrics, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University Vienna, Vienna, Austria
| | - Christopher Gerner
- Institute of Analytical Chemistry, University of Vienna, Vienna, Austria
- Mass Spectrometry Center, University of Vienna, Vienna, Austria
| | | | - Markus Jaritz
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Diana Mechtcheriakova
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
49
|
Frati A, Ricci B, Pierucci F, Nistri S, Bani D, Meacci E. Role of sphingosine kinase/S1P axis in ECM remodeling of cardiac cells elicited by relaxin. Mol Endocrinol 2016; 29:53-67. [PMID: 25415609 DOI: 10.1210/me.2014-1201] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The initiation and progression of heart failure is linked to adverse cardiac remodeling of the extracellular matrix (ECM) during disease mainly through the deregulation of myocardial metalloproteinases (MMPs). Relaxin (RLX), a peptide hormone acting as a physiological cardiac effector, is a key regulator of ECM remodeling in reproductive and nonreproductive tissues. Studying primary cultures of mouse cardiac muscle cells and rat H9c2 cardiomyoblasts, we have obtained evidence for a new signaling pathway activated by RLX to induce ECM remodeling that involves the bioactive sphingolipids sphingosine-1-phosphate (S1P) and ceramide. In both cell populations, recombinant human RLX increased sphingosine kinase activity and S1P formation, whereas sphingomyelin and ceramide content were decreased in [(3)H]serine-labeled cells. According to the literature, RLX promoted MMP-2 and MMP-9 expression/release. Pharmacological inhibition of sphingolipid metabolism and silencing of sphingosine kinase 1, the enzyme responsible for S1P formation, were able to prevent MMP expression/release elicited by the hormone and induce the expression of tissue inhibitor of MMPs. In addition, we found that sphingolipid signaling is required for the regulation of connective tissue growth factor, a member of the CCN 1-3 family of genes that are involved in cell proliferation and differentiation. Finally, the induction of cardiomyoblast maturation induced by RLX was also found to be counteracted by inhibition of S1P formation. In conclusion, these findings provide a novel mechanism by which RLX acts on cardiac ECM remodeling and cardiac cell differentiation and offer interesting therapeutic options to prevent heart fibrosis and to favor myocardial regeneration.
Collapse
Affiliation(s)
- Alessia Frati
- Department of Biomedical, Experimental, and Clinical Sciences (A.F., B.R., F.P., E.M.), Research Unit of Biochemistry, and Department of Experimental and Clinical Medicine (S.N., D.B.), Research Unit of Histology and Embryology, University of Florence, 50134 Florence, Italy
| | | | | | | | | | | |
Collapse
|
50
|
Proia RL, Hla T. Emerging biology of sphingosine-1-phosphate: its role in pathogenesis and therapy. J Clin Invest 2015; 125:1379-87. [PMID: 25831442 DOI: 10.1172/jci76369] [Citation(s) in RCA: 388] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Membrane sphingolipids are metabolized to sphingosine-1-phosphate (S1P), a bioactive lipid mediator that regulates many processes in vertebrate development, physiology, and pathology. Once exported out of cells by cell-specific transporters, chaperone-bound S1P is spatially compartmentalized in the circulatory system. Extracellular S1P interacts with five GPCRs that are widely expressed and transduce intracellular signals to regulate cellular behavior, such as migration, adhesion, survival, and proliferation. While many organ systems are affected, S1P signaling is essential for vascular development, neurogenesis, and lymphocyte trafficking. Recently, a pharmacological S1P receptor antagonist has won approval to control autoimmune neuroinflammation in multiple sclerosis. The availability of pharmacological tools as well as mouse genetic models has revealed several physiological actions of S1P and begun to shed light on its pathological roles. The unique mode of signaling of this lysophospholipid mediator is providing novel opportunities for therapeutic intervention, with possibilities to target not only GPCRs but also transporters, metabolic enzymes, and chaperones.
Collapse
|