ijms-logo

Journal Browser

Journal Browser

Sphingolipids: Signals and Disease

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Biochemistry".

Deadline for manuscript submissions: closed (31 October 2017) | Viewed by 132878

Special Issue Editor


E-Mail Website
Guest Editor
Department of Nutritional Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
Interests: sphingolipidomics; sphingosine 1-phosphate; S1P-receptors; insulin resistance; dendritic cells; epigenetics; nanotoxicology
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

It is now well appreciated that sphingolipids are not only ubiquitous membrane lipids in eukaryotes but also modulate a myriad of physiological and pathophysiological processes. The horizon on sphingolipid research has risen exponentially since bioactive sphingolipid derivatives were first described just over two decades ago. Indeed, in 2010 a first sphingolipid receptor modulator, fingolimod, was employed as a human therapeutic for the treatment of multiple sclerosis. Today it is well established that sphingolipid derivatives are critical players in immunology, inflammation, and cancer, as well as in cardiovascular and metabolic disorders. Sphingolipid research is of great complexity due to the diversity of distinct sphingolipid molelcules and the interconnected metabolic pathways. The levels of sphingolipids are tightly regulated by a multitude of enzymes which are involved in the biosynthesis and degradation of sphingolipids. New knowledge on the biology and metabolism of sphingolipids is anticipated to further understand the role of this lipid class in a variety of pathophysiological conditions.
In this Special Issue, we will welcome your contributions in the form of original research and review articles in all aspects of Sphingolipids.

Prof. Dr. Burkhard Kleuser
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • bioactive sphingolipids
  • ceramides
  • Sphingosine 1-phosphate
  • S1P-receptors
  • sphingolipid metabolism
  • sphingolipids and metabolic disorders
  • sphingolipids and inflammation
  • sphingolipids and infections
  • sphingolipids and cancer

Related Special Issue

Published Papers (24 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

3 pages, 154 KiB  
Editorial
The Enigma of Sphingolipids in Health and Disease
by Burkhard Kleuser
Int. J. Mol. Sci. 2018, 19(10), 3126; https://doi.org/10.3390/ijms19103126 - 12 Oct 2018
Cited by 2 | Viewed by 2100
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)

Research

Jump to: Editorial, Review

19 pages, 2944 KiB  
Article
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
by Olivier Blanchard, Bisera Stepanovska, Manuel Starck, Martin Erhardt, Isolde Römer, Dagmar Meyer zu Heringdorf, Josef Pfeilschifter, Uwe Zangemeister-Wittke and Andrea Huwiler
Int. J. Mol. Sci. 2018, 19(5), 1498; https://doi.org/10.3390/ijms19051498 - 17 May 2018
Cited by 21 | Viewed by 5282
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 [...] Read more.
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. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Figure 1

14 pages, 4355 KiB  
Article
Sphingosine-1-Phosphate Receptor 1 Is Involved in Non-Obese Diabetic Mouse Thymocyte Migration Disorders
by Julia P. Lemos, Salete Smaniotto, Carolina V. Messias, Otacilio C. Moreira, Vinicius Cotta-de-Almeida, Mireille Dardenne, Wilson Savino and Daniella A. Mendes-da-Cruz
Int. J. Mol. Sci. 2018, 19(5), 1446; https://doi.org/10.3390/ijms19051446 - 12 May 2018
Cited by 9 | Viewed by 4080
Abstract
NOD (non-obese diabetic) mice spontaneously develop type 1 diabetes following T cell-dependent destruction of pancreatic β cells. Several alterations are observed in the NOD thymus, including the presence of giant perivascular spaces (PVS) filled with single-positive (SP) CD4+ and CD8+ T [...] Read more.
NOD (non-obese diabetic) mice spontaneously develop type 1 diabetes following T cell-dependent destruction of pancreatic β cells. Several alterations are observed in the NOD thymus, including the presence of giant perivascular spaces (PVS) filled with single-positive (SP) CD4+ and CD8+ T cells that accumulate in the organ. These cells have a decreased expression of membrane CD49e (the α5 integrin chain of the fibronectin receptor VLA-5 (very late antigen-5). Herein, we observed lower sphingosine-1-phosphate receptor 1 (S1P1) expression in NOD mouse thymocytes when compared with controls, mainly in the mature SP CD4+CD62Lhi and CD8+CD62Lhi subpopulations bearing the CD49e phenotype. In contrast, differences in S1P1 expression were not observed in mature CD49e+ thymocytes. Functionally, NOD CD49e thymocytes had reduced S1P-driven migratory response, whereas CD49e+ cells were more responsive to S1P. We further noticed a decreased expression of the sphingosine-1-phosphate lyase (SGPL1) in NOD SP thymocytes, which can lead to a higher sphingosine-1-phosphate (S1P) expression around PVS and S1P1 internalization. In summary, our results indicate that the modulation of S1P1 expression and S1P/S1P1 interactions in NOD mouse thymocytes are part of the T-cell migratory disorder observed during the pathogenesis of type 1 diabetes. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

15 pages, 1700 KiB  
Article
S1P4 Regulates Passive Systemic Anaphylaxis in Mice but Is Dispensable for Canonical IgE-Mediated Responses in Mast Cells
by Joseph M. Kulinski, Richard L. Proia, Elisabeth M. Larson, Dean D. Metcalfe and Ana Olivera
Int. J. Mol. Sci. 2018, 19(5), 1279; https://doi.org/10.3390/ijms19051279 - 25 Apr 2018
Cited by 12 | Viewed by 4520
Abstract
Mast cells are key players in the development of inflammatory allergic reactions. Cross-linking of the high-affinity receptor for IgE (FcεRI) on mast cells leads to the generation and secretion of the sphingolipid mediator, sphingosine-1-phosphate (S1P) which is able, in turn, to transactivate its [...] Read more.
Mast cells are key players in the development of inflammatory allergic reactions. Cross-linking of the high-affinity receptor for IgE (FcεRI) on mast cells leads to the generation and secretion of the sphingolipid mediator, sphingosine-1-phosphate (S1P) which is able, in turn, to transactivate its receptors on mast cells. Previous reports have identified the expression of two of the five receptors for S1P on mast cells, S1P1 and S1P2, with functions in FcεRI-mediated chemotaxis and degranulation, respectively. Here, we show that cultured mouse mast cells also express abundant message for S1P4. Genetic deletion of S1pr4 did not affect the differentiation of bone marrow progenitors into mast cells or the proliferation of mast cells in culture. A comprehensive characterization of IgE-mediated responses in S1P4-deficient bone marrow-derived and peritoneal mouse mast cells indicated that this receptor is dispensable for mast cell degranulation, cytokine/chemokine production and FcεRI-mediated chemotaxis in vitro. However, interleukin-33 (IL-33)-mediated enhancement of IgE-induced degranulation was reduced in S1P4-deficient peritoneal mast cells, revealing a potential negative regulatory role for S1P4 in an IL-33-rich environment. Surprisingly, genetic deletion of S1pr4 resulted in exacerbation of passive systemic anaphylaxis to IgE/anti-IgE in mice, a phenotype likely related to mast cell-extrinsic influences, such as the high circulating levels of IgE in these mice which increases FcεRI expression and consequently the extent of the response to FcεRI engagement. Thus, we provide evidence that S1P4 modulates anaphylaxis in an unexpected manner that does not involve regulation of mast cell responsiveness to IgE stimulation. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Figure 1

15 pages, 3864 KiB  
Article
Low Serum Levels of (Dihydro-)Ceramides Reflect Liver Graft Dysfunction in a Real-World Cohort of Patients Post Liver Transplantation
by Victoria Therese Mücke, Janis Gerharz, Katja Jakobi, Dominique Thomas, Nerea Ferreirós Bouzas, Marcus Maximilian Mücke, Sven Trötschler, Nina Weiler, Martin-Walter Welker, Stefan Zeuzem, Josef Pfeilschifter and Georgios Grammatikos
Int. J. Mol. Sci. 2018, 19(4), 991; https://doi.org/10.3390/ijms19040991 - 26 Mar 2018
Cited by 8 | Viewed by 4678
Abstract
Patients after orthopic liver transplantation (OLT) are at risk of developing graft dysfunction. Sphingolipids (SL’s) have been identified to play a pivotal role in the regulation of hepatocellular apoptosis, inflammation and immunity. We aimed to investigate the serum SL profile in a prospective [...] Read more.
Patients after orthopic liver transplantation (OLT) are at risk of developing graft dysfunction. Sphingolipids (SL’s) have been identified to play a pivotal role in the regulation of hepatocellular apoptosis, inflammation and immunity. We aimed to investigate the serum SL profile in a prospective real-world cohort of post-OLT patients. From October 2015 until July 2016, 149 well-characterized post-OLT patients were analyzed. SL’s were assessed in serum probes via Liquid Chromatography/Tandem Mass Spectrometry. Twenty-nine (20%) patients had a biopsy proven graft rejection with decreased C20-ceramide (Cer) (p = 0.042), C18-dihydroceramide (DHC) (p = 0.022) and C24DHC (p = 0.060) levels. Furthermore, C18DHC (p = 0.044) and C24DHC (p = 0.011) were significantly down-regulated in patients with ischemic type biliary lesions (ITBL; n = 15; 10%). One-hundred and thirty-three patients (89%) have so far received tacrolimus as the main immunosuppressive agent with observed elevations of C14Cer (p = 0.052), C18Cer (p = 0.049) and C18:1Cer (p = 0.024). Hepatocellular carcinoma (HCC) pre-OLT was associated with increases in C24:1Cer (p = 0.024) and C24:1DHC (p = 0.024). In this large prospective cross-sectional study of patients, post-OLT serum levels of (very-)long chain (dihydro-)ceramides associate with graft rejection, ITBL, tacrolimus intake and HCC pre-OLT. Hence, serum SL’s may be indicative of graft complications. Further research is necessary to identify their diverse mechanistic role in regulating immunity and inflammation in patients post-OLT. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Figure 1

13 pages, 1352 KiB  
Article
S1P Signalling Differentially Affects Migration of Peritoneal B Cell Populations In Vitro and Influences the Production of Intestinal IgA In Vivo
by Annabel Kleinwort, Felix Lührs, Claus-Dieter Heidecke, Martin Lipp and Tobias Schulze
Int. J. Mol. Sci. 2018, 19(2), 391; https://doi.org/10.3390/ijms19020391 - 29 Jan 2018
Cited by 21 | Viewed by 4761
Abstract
Introduction: Sphingosine-1-phosphate (S1P) regulates the migration of follicular B cells (B2 cells) and directs the positioning of Marginal zone B cells (MZ B cells) within the spleen. The function of S1P signalling in the third B cell lineage, B1 B cells, mainly present [...] Read more.
Introduction: Sphingosine-1-phosphate (S1P) regulates the migration of follicular B cells (B2 cells) and directs the positioning of Marginal zone B cells (MZ B cells) within the spleen. The function of S1P signalling in the third B cell lineage, B1 B cells, mainly present in the pleural and peritoneal cavity, has not yet been determined. Methods: S1P receptor expression was analysed in peritoneal B cells by real-time polymerase chain reaction (qPCR). The chemotactic response to S1P was studied in vitro. The role of S1P signalling was further explored in a s1p4−/− mouse strain. Results: Peritoneal B cells expressed considerable amounts of the S1P receptors 1 and 4 (S1P1 and S1P4, respectively). S1P1 showed differential expression between the distinct peritoneal B cell lineages. While B2 cells showed no chemotactic response to S1P, B1 B cells showed a migration response to S1P. s1p4−/− mice displayed significant alterations in the composition of peritoneal B cell populations, as well as a significant reduction of mucosal immunoglobulin A (IgA) in the gut. Discussion: S1P signalling influences peritoneal B1 B cell migration. S1P4 deficiency alters the composition of peritoneal B cell populations and reduces secretory IgA levels. These findings suggest that S1P signalling may be a target to modulate B cell function in inflammatory intestinal pathologies. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Figure 1

19 pages, 4005 KiB  
Article
Lysophosphatidic Acid Signaling Axis Mediates Ceramide 1-Phosphate-Induced Proliferation of C2C12 Myoblasts
by Caterina Bernacchioni, Francesca Cencetti, Alberto Ouro, Marina Bruno, Antonio Gomez-Muñoz, Chiara Donati and Paola Bruni
Int. J. Mol. Sci. 2018, 19(1), 139; https://doi.org/10.3390/ijms19010139 - 04 Jan 2018
Cited by 23 | Viewed by 4398
Abstract
Sphingolipids are not only crucial for membrane architecture but act as critical regulators of cell functions. The bioactive sphingolipid ceramide 1-phosphate (C1P), generated by the action of ceramide kinase, has been reported to stimulate cell proliferation, cell migration and to regulate inflammatory responses [...] Read more.
Sphingolipids are not only crucial for membrane architecture but act as critical regulators of cell functions. The bioactive sphingolipid ceramide 1-phosphate (C1P), generated by the action of ceramide kinase, has been reported to stimulate cell proliferation, cell migration and to regulate inflammatory responses via activation of different signaling pathways. We have previously shown that skeletal muscle is a tissue target for C1P since the phosphosphingolipid plays a positive role in myoblast proliferation implying a role in muscle regeneration. Skeletal muscle displays strong capacity of regeneration thanks to the presence of quiescent adult stem cells called satellite cells that upon trauma enter into the cell cycle and start proliferating. However, at present, the exact molecular mechanism by which C1P triggers its mitogenic effect in myoblasts is lacking. Here, we report for the first time that C1P stimulates C2C12 myoblast proliferation via lysophosphatidic acid (LPA) signaling axis. Indeed, C1P subsequently to phospholipase A2 activation leads to LPA1 and LPA3 engagement, which in turn drive Akt (protein kinase B) and ERK1/2 (extracellular signal-regulated kinases 1/2) activation, thus stimulating DNA synthesis. The present findings shed new light on the key role of bioactive sphingolipids in skeletal muscle and provide further support to the notion that these pleiotropic molecules might be useful therapeutic targets for skeletal muscle regeneration. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

17 pages, 9787 KiB  
Article
Sphingolipids in Ventilator Induced Lung Injury: Role of Sphingosine-1-Phosphate Lyase
by Vidyani Suryadevara, Panfeng Fu, David Lenin Ebenezer, Evgeny Berdyshev, Irina A. Bronova, Long Shuang Huang, Anantha Harijith and Viswanathan Natarajan
Int. J. Mol. Sci. 2018, 19(1), 114; https://doi.org/10.3390/ijms19010114 - 01 Jan 2018
Cited by 28 | Viewed by 5315
Abstract
Mechanical ventilation (MV) performed in respiratory failure patients to maintain lung function leads to ventilator-induced lung injury (VILI). This study investigates the role of sphingolipids and sphingolipid metabolizing enzymes in VILI using a rodent model of VILI and alveolar epithelial cells subjected to [...] Read more.
Mechanical ventilation (MV) performed in respiratory failure patients to maintain lung function leads to ventilator-induced lung injury (VILI). This study investigates the role of sphingolipids and sphingolipid metabolizing enzymes in VILI using a rodent model of VILI and alveolar epithelial cells subjected to cyclic stretch (CS). MV (0 PEEP (Positive End Expiratory Pressure), 30 mL/kg, 4 h) in mice enhanced sphingosine-1-phosphate lyase (S1PL) expression, and ceramide levels, and decreased S1P levels in lung tissue, thereby leading to lung inflammation, injury and apoptosis. Accumulation of S1P in cells is a balance between its synthesis catalyzed by sphingosine kinase (SphK) 1 and 2 and catabolism mediated by S1P phosphatases and S1PL. Thus, the role of S1PL and SphK1 in VILI was investigated using Sgpl1+/− and Sphk1−/− mice. Partial genetic deletion of Sgpl1 protected mice against VILI, whereas deletion of SphK1 accentuated VILI in mice. Alveolar epithelial MLE-12 cells subjected to pathophysiological 18% cyclic stretch (CS) exhibited increased S1PL protein expression and dysregulation of sphingoid bases levels as compared to physiological 5% CS. Pre-treatment of MLE-12 cells with S1PL inhibitor, 4-deoxypyridoxine, attenuated 18% CS-induced barrier dysfunction, minimized cell apoptosis and cytokine secretion. These results suggest that inhibition of S1PL that increases S1P levels may offer protection against VILI. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

3135 KiB  
Article
Apolipoprotein M Inhibits Angiogenic and Inflammatory Response by Sphingosine 1-Phosphate on Retinal Pigment Epithelium Cells
by Ryo Terao, Megumi Honjo and Makoto Aihara
Int. J. Mol. Sci. 2018, 19(1), 112; https://doi.org/10.3390/ijms19010112 - 31 Dec 2017
Cited by 24 | Viewed by 4561
Abstract
Sphingosine 1-phosphate (S1P) is a potent lipid mediator that modulates inflammatory responses and proangiogenic factors. It has been suggested that S1P upregulates choroidal neovascularization (CNV) and may be deeply involved in the pathogenesis of exudative age-related macular degeneration (AMD). Recent studies have suggested [...] Read more.
Sphingosine 1-phosphate (S1P) is a potent lipid mediator that modulates inflammatory responses and proangiogenic factors. It has been suggested that S1P upregulates choroidal neovascularization (CNV) and may be deeply involved in the pathogenesis of exudative age-related macular degeneration (AMD). Recent studies have suggested that apolipoprotein M (ApoM), a carrier protein for S1P, modulates the biological properties of S1P in the pathogenesis of atherosclerosis. However, the role of ApoM/S1P in AMD has not been explored. We investigated the effect of S1P on proangiogenic factors in human retinal pigment epithelium (RPE) cell lines in vitro. S1P promoted the expression of vascular endothelial growth factor in RPE cells. Hypoxia inducible factor-1α expression was also upregulated. These S1P-induced enhancements in growth factors and chemotactic cytokines in RPE cells were significantly inhibited by ApoM treatment. Additionally, in vivo experiments using a laser-induced CNV murine model demonstrated that intravitreal ApoM injection significantly reduced the progression of CNV formation. Although the detailed mechanisms remain to be elucidated, the present results provide a novel potential therapeutic target for AMD, and demonstrate a suppressive role for ApoM and S1P in the pathology of CNV progression. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Figure 1

5712 KiB  
Article
Sphingosine-1-Phosphate Receptor 1, Expressed in Myeloid Cells, Slows Diet-Induced Atherosclerosis and Protects against Macrophage Apoptosis in Ldlr KO Mice
by Leticia Gonzalez, Alexander S. Qian, Usama Tahir, Pei Yu and Bernardo L. Trigatti
Int. J. Mol. Sci. 2017, 18(12), 2721; https://doi.org/10.3390/ijms18122721 - 15 Dec 2017
Cited by 24 | Viewed by 4699
Abstract
We generated myeloid specific sphingosine-1-phosphate receptor 1 (S1pr1) deficient mice by crossing mice that had myeloid specific expression of Cre recombinase (lyzMCre) with mice having the S1pr1 gene flanked by loxP recombination sites. We transplanted bone marrow from [...] Read more.
We generated myeloid specific sphingosine-1-phosphate receptor 1 (S1pr1) deficient mice by crossing mice that had myeloid specific expression of Cre recombinase (lyzMCre) with mice having the S1pr1 gene flanked by loxP recombination sites. We transplanted bone marrow from these mice and control lyzMCre mice with intact macrophage S1pr1 gene expression into low-density lipoprotein (LDL) receptor gene (Ldlr) deficient mice. The resulting chimeras were fed a high fat atherogenic diet for nine or twelve weeks and evaluated for atherosclerosis development in the aortic sinus. Selective S1pr1 deficiency in bone marrow-derived myeloid cells resulted in accelerated development of atherosclerosis, necrotic core formation and the appearance of apoptotic cells within atherosclerotic plaques of Ldlr knockout mice in response to a high fat diet. Examination of macrophages in culture revealed that the sphingosine-1-phosphate receptor 1 selective agonist, SEW2871 or high density lipoprotein (HDL), protected macrophages against apoptosis induced by endoplasmic reticulum (ER) stress or oxidized LDL, through activation of phosphatidylinositol-3-kinase/Akt signaling. Targeted S1pr1-deletion prevented Akt activation and protection against apoptosis by either SEW2871 or HDL. Our data suggests that sphingosine-1-phosphate receptor 1 in macrophages plays an important role in protecting them against apoptosis in vitro and in atherosclerotic plaques in vivo, and delays diet induced atherosclerosis development in Ldlr deficient mice. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

1543 KiB  
Article
Pharmacokinetics, Pharmacodynamics, Tolerability, and Food Effect of Cenerimod, a Selective S1P1 Receptor Modulator in Healthy Subjects
by Pierre-Eric Juif, Daniela Baldoni, Maribel Reyes, Darren Wilbraham, Salvatore Febbraro, Andrea Vaclavkova, Matthias Hoch and Jasper Dingemanse
Int. J. Mol. Sci. 2017, 18(12), 2636; https://doi.org/10.3390/ijms18122636 - 06 Dec 2017
Cited by 14 | Viewed by 6436
Abstract
The pharmacokinetics, pharmacodynamics, tolerability, and food effect of cenerimod, a potent sphingosine-1-phosphate subtype 1 receptor modulator, were investigated in three sub-studies. Two double-blind, placebo-controlled, randomised studies in healthy male subjects were performed. Cenerimod was administered either as single dose (1, 3, 10 or [...] Read more.
The pharmacokinetics, pharmacodynamics, tolerability, and food effect of cenerimod, a potent sphingosine-1-phosphate subtype 1 receptor modulator, were investigated in three sub-studies. Two double-blind, placebo-controlled, randomised studies in healthy male subjects were performed. Cenerimod was administered either as single dose (1, 3, 10 or 25 mg; Study 1) or once daily for 35 days (0.5, 1, 2 or 4 mg; Study 2). A two-period cross-over, open-label study was performed to assess the food effect (1 mg, Study 3). The pharmacokinetic profile of cenerimod was characterised by a tmax of 5.0–6.2 h. Terminal half-life after single and multiple doses ranged from 170 to 199 h and 283 to 539 h, respectively. Food had no relevant effect on the pharmacokinetics of cenerimod. A dose-dependent decrease in lymphocyte count was observed after initiation of cenerimod and reached a plateau (maximum change from baseline: −64%) after 20–23 days of treatment. Lymphocyte counts returned to baseline values at end-of-study examination. One serious adverse event of circulatory collapse (25 mg dose group, maximum tolerated dose: 10 mg) and adverse events of mild-to-moderate intensity were reported. Treatment initiation was associated with transient decreases in heart rate and blood pressure at doses >1 and ≥10 mg, respectively. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Figure 1

1031 KiB  
Article
Plasma Sphingolipids in Acute Pancreatitis
by Tomasz Konończuk, Bartłomiej Łukaszuk, Małgorzata Żendzian-Piotrowska, Andrzej Dąbrowski, Michalina Krzyżak, Lucyna Ostrowska and Krzysztof Kurek
Int. J. Mol. Sci. 2017, 18(12), 2606; https://doi.org/10.3390/ijms18122606 - 04 Dec 2017
Cited by 17 | Viewed by 3688
Abstract
Acute pancreatitis (AP) is a prevalent gastrointestinal disorder associated with systemic inflammatory response syndrome and, in the case of severe AP, a mortality rate ranging from 36% to 50%. Standard clinical treatment of AP includes intensive hydration, analgesia, and management of complications. Unfortunately, [...] Read more.
Acute pancreatitis (AP) is a prevalent gastrointestinal disorder associated with systemic inflammatory response syndrome and, in the case of severe AP, a mortality rate ranging from 36% to 50%. Standard clinical treatment of AP includes intensive hydration, analgesia, and management of complications. Unfortunately, the direct treatment of AP at the level of its molecular pathomechanism has not yet been established. Recent studies indicate that the sphingolipid signaling pathway may be one of the important factors contributing to the development of inflammation in pancreatic diseases. In the current study, we sought to investigate this promising route. We examined the plasma sphingolipid profile of 44 patients with acute pancreatitis, dividing them into three groups: mild, moderate and severe AP. Samples were collected from these groups at days 1, 3 and 7 following their hospital admission. We demonstrated significant changes in blood plasma sphingolipids in relation to the time course of AP. We also found an inhibition of de novo ceramide synthesis in mild and moderate AP. However, the most important and novel finding was a significant elevation in sphingosine-1-phosphate (S1P) (a downstream metabolite of ceramide) in mild AP, as well as a dramatic reduction in the lipid molecule content in the early stage (days 1 and 3) of severe AP. This strongly indicates that plasma S1P could serve as a prognostic marker of AP severity. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

2506 KiB  
Article
Altered Leukocyte Sphingolipid Pathway in Breast Cancer
by Larissa P. Maia, Paula S. Santos, Patrícia T. Alves, Cláudia M. Rodrigues, Thaíse G. Araújo, Yara Cristina P. Maia, Alinne Tatiane F. Câmara, Donizeti W. Santos and Luiz Ricardo Goulart
Int. J. Mol. Sci. 2017, 18(12), 2521; https://doi.org/10.3390/ijms18122521 - 24 Nov 2017
Cited by 5 | Viewed by 4355
Abstract
Sphingolipid metabolism pathway is essential in membrane homeostasis, and its dysfunction has been associated with favorable tumor microenvironment, disease progression, and chemotherapy resistance. Its major components have key functions on survival and proliferation, with opposing effects. We have profiled the components of the [...] Read more.
Sphingolipid metabolism pathway is essential in membrane homeostasis, and its dysfunction has been associated with favorable tumor microenvironment, disease progression, and chemotherapy resistance. Its major components have key functions on survival and proliferation, with opposing effects. We have profiled the components of the sphingolipid pathway on leukocytes of breast cancer (BC) patients undergoing chemotherapy treatment and without, including the five sphingosine 1-phosphate (S1P) receptors, the major functional genes, and cytokines, in order to better understand the S1P signaling in the immune cells of these patients. To the best of our knowledge, this is the first characterization of the sphingolipid pathway in whole blood of BC patients. Skewed gene profiles favoring high SPHK1 expression toward S1P production during BC development was observed, which was reversed by chemotherapy treatment, and reached similar levels to those found in healthy donors. Such levels were also correlated with high levels of TNF-α. Our data revealed an important role of the sphingolipid pathway in immune cells in BC with skewed signaling of S1P receptors, which favored cancer development even under chemotherapy, and may probably be a trigger of cancer resistance. Thus, these molecules must be considered as a target pathway for combined BC therapeutics. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

2056 KiB  
Article
Development of 1,2,3-Triazole-Based Sphingosine Kinase Inhibitors and Their Evaluation as Antiproliferative Agents
by Angela Corvino, Roberta Rosa, Giuseppina Maria Incisivo, Ferdinando Fiorino, Francesco Frecentese, Elisa Magli, Elisa Perissutti, Irene Saccone, Vincenzo Santagada, Giuseppe Cirino, Maria Antonietta Riemma, Piero A. Temussi, Paola Ciciola, Roberto Bianco, Giuseppe Caliendo, Fiorentina Roviezzo and Beatrice Severino
Int. J. Mol. Sci. 2017, 18(11), 2332; https://doi.org/10.3390/ijms18112332 - 05 Nov 2017
Cited by 5 | Viewed by 4355
Abstract
Two series of N-(aryl)-1-(hydroxyalkyl)pyrrolidine-2-carboxamides (2a2g and 3a3g) and 1,4-disubstituted 1,2,3-triazoles (5a5h and 8a8h) were synthesized. All the compounds, containing a lipophilic tail and a polar headgroup, were evaluated as sphingosine [...] Read more.
Two series of N-(aryl)-1-(hydroxyalkyl)pyrrolidine-2-carboxamides (2a2g and 3a3g) and 1,4-disubstituted 1,2,3-triazoles (5a5h and 8a8h) were synthesized. All the compounds, containing a lipophilic tail and a polar headgroup, were evaluated as sphingosine kinase (SphK) inhibitors by assessing their ability to interfere with the acetylcholine (Ach) induced relaxation of aortic rings pre-contracted with phenylephrine. Moreover, their antiproliferative activity was tested on several cell lines expressing both SphK1 and SphK2. Compounds 5h and 8f, identified as the most efficient antiproliferative agents, showed a different selectivity profile, with 8f being selective for SphK1. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

Review

Jump to: Editorial, Research

18 pages, 881 KiB  
Review
Divergent Role of Sphingosine 1-Phosphate in Liver Health and Disease
by Burkhard Kleuser
Int. J. Mol. Sci. 2018, 19(3), 722; https://doi.org/10.3390/ijms19030722 - 03 Mar 2018
Cited by 35 | Viewed by 5548
Abstract
Two decades ago, sphingosine 1-phosphate (S1P) was discovered as a novel bioactive molecule that regulates a variety of cellular functions. The plethora of S1P-mediated effects is due to the fact that the sphingolipid not only modulates intracellular functions but also acts as a [...] Read more.
Two decades ago, sphingosine 1-phosphate (S1P) was discovered as a novel bioactive molecule that regulates a variety of cellular functions. The plethora of S1P-mediated effects is due to the fact that the sphingolipid not only modulates intracellular functions but also acts as a ligand of G protein-coupled receptors after secretion into the extracellular environment. In the plasma, S1P is found in high concentrations, modulating immune cell trafficking and vascular endothelial integrity. The liver is engaged in modulating the plasma S1P content, as it produces apolipoprotein M, which is a chaperone for the S1P transport. Moreover, the liver plays a substantial role in glucose and lipid homeostasis. A dysfunction of glucose and lipid metabolism is connected with the development of liver diseases such as hepatic insulin resistance, non-alcoholic fatty liver disease, or liver fibrosis. Recent studies indicate that S1P is involved in liver pathophysiology and contributes to the development of liver diseases. In this review, the current state of knowledge about S1P and its signaling in the liver is summarized with a specific focus on the dysregulation of S1P signaling in obesity-mediated liver diseases. Thus, the modulation of S1P signaling can be considered as a potential therapeutic target for the treatment of hepatic diseases. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Figure 1

28 pages, 1026 KiB  
Review
Abnormal Sphingolipid World in Inflammation Specific for Lysosomal Storage Diseases and Skin Disorders
by Marta Moskot, Katarzyna Bocheńska, Joanna Jakóbkiewicz-Banecka, Bogdan Banecki and Magdalena Gabig-Cimińska
Int. J. Mol. Sci. 2018, 19(1), 247; https://doi.org/10.3390/ijms19010247 - 15 Jan 2018
Cited by 27 | Viewed by 7504
Abstract
Research in recent years has shown that sphingolipids are essential signalling molecules for the proper biological and structural functioning of cells. Long-term studies on the metabolism of sphingolipids have provided evidence for their role in the pathogenesis of a number of diseases. As [...] Read more.
Research in recent years has shown that sphingolipids are essential signalling molecules for the proper biological and structural functioning of cells. Long-term studies on the metabolism of sphingolipids have provided evidence for their role in the pathogenesis of a number of diseases. As many inflammatory diseases, such as lysosomal storage disorders and some dermatologic diseases, including psoriasis, atopic dermatitis and ichthyoses, are associated with the altered composition and metabolism of sphingolipids, more studies precisely determining the responsibilities of these compounds for disease states are required to develop novel pharmacological treatment opportunities. It is worth emphasizing that knowledge from the study of inflammatory metabolic diseases and especially the possibility of their treatment may lead to insight into related metabolic pathways, including those involved in the formation of the epidermal barrier and providing new approaches towards workable therapies. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

1307 KiB  
Review
Translational Aspects of Sphingolipid Metabolism in Renal Disorders
by Alaa Abou Daher, Tatiana El Jalkh, Assaad A. Eid, Alessia Fornoni, Brian Marples and Youssef H. Zeidan
Int. J. Mol. Sci. 2017, 18(12), 2528; https://doi.org/10.3390/ijms18122528 - 25 Nov 2017
Cited by 22 | Viewed by 7132
Abstract
Sphingolipids, long thought to be passive components of biological membranes with merely a structural role, have proved throughout the past decade to be major players in the pathogenesis of many human diseases. The study and characterization of several genetic disorders like Fabry’s and [...] Read more.
Sphingolipids, long thought to be passive components of biological membranes with merely a structural role, have proved throughout the past decade to be major players in the pathogenesis of many human diseases. The study and characterization of several genetic disorders like Fabry’s and Tay Sachs, where sphingolipid metabolism is disrupted, leading to a systemic array of clinical symptoms, have indeed helped elucidate and appreciate the importance of sphingolipids and their metabolites as active signaling molecules. In addition to being involved in dynamic cellular processes like apoptosis, senescence and differentiation, sphingolipids are implicated in critical physiological functions such as immune responses and pathophysiological conditions like inflammation and insulin resistance. Interestingly, the kidneys are among the most sensitive organ systems to sphingolipid alterations, rendering these molecules and the enzymes involved in their metabolism, promising therapeutic targets for numerous nephropathic complications that stand behind podocyte injury and renal failure. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Figure 1

5198 KiB  
Review
Sphingosine 1-Phosphate (S1P) Signaling in Glioblastoma Multiforme—A Systematic Review
by Shailaja Mahajan-Thakur, Sandra Bien-Möller, Sascha Marx, Henry Schroeder and Bernhard H. Rauch
Int. J. Mol. Sci. 2017, 18(11), 2448; https://doi.org/10.3390/ijms18112448 - 17 Nov 2017
Cited by 43 | Viewed by 10661
Abstract
The multifunctional sphingosine-1-phosphate (S1P) is a lipid signaling molecule and central regulator in the development of several cancer types. In recent years, intriguing information has become available regarding the role of S1P in the progression of Glioblastoma multiforme (GBM), the most aggressive and [...] Read more.
The multifunctional sphingosine-1-phosphate (S1P) is a lipid signaling molecule and central regulator in the development of several cancer types. In recent years, intriguing information has become available regarding the role of S1P in the progression of Glioblastoma multiforme (GBM), the most aggressive and common brain tumor in adults. S1P modulates numerous cellular processes in GBM, such as oncogenesis, proliferation and survival, invasion, migration, metastasis and stem cell behavior. These processes are regulated via a family of five G-protein-coupled S1P receptors (S1PR1-5) and may involve mainly unknown intracellular targets. Distinct expression patterns and multiple intracellular signaling pathways of each S1PR subtype enable S1P to exert its pleiotropic cellular actions. Several studies have demonstrated alterations in S1P levels, the involvement of S1PRs and S1P metabolizing enzymes in GBM pathophysiology. While the tumorigenic actions of S1P involve the activation of several kinases and transcription factors, the specific G-protein (Gi, Gq, and G12/13)-coupled signaling pathways and downstream mediated effects in GBM remain to be elucidated in detail. This review summarizes the recent findings concerning the role of S1P and its receptors in GBM. We further highlight the current insights into the signaling pathways considered fundamental for regulating the cellular processes in GMB and ultimately patient prognosis. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

737 KiB  
Review
Sphingosine Kinase 1 and Sphingosine-1-Phosphate Signaling in Colorectal Cancer
by Yonghua Bao, Yongchen Guo, Chenglan Zhang, Fenghua Fan and Wancai Yang
Int. J. Mol. Sci. 2017, 18(10), 2109; https://doi.org/10.3390/ijms18102109 - 08 Oct 2017
Cited by 35 | Viewed by 6636
Abstract
Sphingosine kinase 1 (Sphk1) is a highly conserved lipid kinase that phosphorylates sphingosine to form sphingosine-1-phosphate (S1P). Growing studies have demonstrated that Sphk1 is overexpressed in various types of solid cancers and can be induced by growth factors, cytokines, and carcinogens, leading to [...] Read more.
Sphingosine kinase 1 (Sphk1) is a highly conserved lipid kinase that phosphorylates sphingosine to form sphingosine-1-phosphate (S1P). Growing studies have demonstrated that Sphk1 is overexpressed in various types of solid cancers and can be induced by growth factors, cytokines, and carcinogens, leading to the increase of S1P production. Subsequently, the increased Sphk1/S1P facilitates cancer cell proliferation, mobility, angiogenesis, invasion, and metastasis. Therefore, Sphk1/S1P signaling plays oncogenic roles. This review summarizes the features of Sphk1/S1P signaling and their functions in colorectal cancer cell growth, tumorigenesis, and metastasis, as well as the possible underlying mechanisms. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

886 KiB  
Review
Sphingosine 1-Phosphate Signaling and Its Pharmacological Modulation in Allogeneic Hematopoietic Stem Cell Transplantation
by Philip Smith, Catherine O’Sullivan and Peter Gergely
Int. J. Mol. Sci. 2017, 18(10), 2027; https://doi.org/10.3390/ijms18102027 - 21 Sep 2017
Cited by 6 | Viewed by 5351
Abstract
Allogeneic haemopoietic stem cell transplantation (HSCT) is increasingly used to treat haematological malignant diseases via the graft-versus-leukaemia (GvL) or graft-versus-tumour effects. Although improvements in infectious disease prophylaxis, immunosuppressive treatments, supportive care, and molecular based tissue typing have contributed to enhanced outcomes, acute graft-versus-host [...] Read more.
Allogeneic haemopoietic stem cell transplantation (HSCT) is increasingly used to treat haematological malignant diseases via the graft-versus-leukaemia (GvL) or graft-versus-tumour effects. Although improvements in infectious disease prophylaxis, immunosuppressive treatments, supportive care, and molecular based tissue typing have contributed to enhanced outcomes, acute graft-versus-host disease and other transplant related complications still contribute to high mortality and significantly limit the more widespread use of HSCT. Sphingosine 1-phosphate (S1P) is a zwitterionic lysophospholipid that has been implicated as a crucial signaling regulator in many physiological and pathophysiological processes including multiple cell types such as macrophages, dendritic cells, T cells, T regulatory cells and endothelial cells. Recent data suggested important roles for S1P signaling in engraftment, graft-versus-host disease (GvHD), GvL and other processes that occur during and after HSCT. Based on such data, pharmacological intervention via S1P modulation may have the potential to improve patient outcome by regulating GvHD and enhancing engraftment while permitting effective GvL. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Figure 1

1718 KiB  
Review
“Dicing and Splicing” Sphingosine Kinase and Relevance to Cancer
by Nahal Haddadi, Yiguang Lin, Ann M. Simpson, Najah T. Nassif and Eileen M. McGowan
Int. J. Mol. Sci. 2017, 18(9), 1891; https://doi.org/10.3390/ijms18091891 - 02 Sep 2017
Cited by 30 | Viewed by 6527
Abstract
Sphingosine kinase (SphK) is a lipid enzyme that maintains cellular lipid homeostasis. Two SphK isozymes, SphK1 and SphK2, are expressed from different chromosomes and several variant isoforms are expressed from each of the isozymes, allowing for the multi-faceted biological diversity of SphK activity. [...] Read more.
Sphingosine kinase (SphK) is a lipid enzyme that maintains cellular lipid homeostasis. Two SphK isozymes, SphK1 and SphK2, are expressed from different chromosomes and several variant isoforms are expressed from each of the isozymes, allowing for the multi-faceted biological diversity of SphK activity. Historically, SphK1 is mainly associated with oncogenicity, however in reality, both SphK1 and SphK2 isozymes possess oncogenic properties and are recognized therapeutic targets. The absence of mutations of SphK in various cancer types has led to the theory that cancer cells develop a dependency on SphK signaling (hyper-SphK signaling) or “non-oncogenic addiction”. Here we discuss additional theories of SphK cellular mislocation and aberrant “dicing and splicing” as contributors to cancer cell biology and as key determinants of the success or failure of SphK/S1P (sphingosine 1 phosphate) based therapeutics. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

797 KiB  
Review
The Role of Sphingolipids on Innate Immunity to Intestinal Salmonella Infection
by Fu-Chen Huang
Int. J. Mol. Sci. 2017, 18(8), 1720; https://doi.org/10.3390/ijms18081720 - 07 Aug 2017
Cited by 13 | Viewed by 5014
Abstract
Salmonella spp. remains a major public health problem for the whole world. To reduce the use of antimicrobial agents and drug-resistant Salmonella, a better strategy is to explore alternative therapy rather than to discover another antibiotic. Sphingolipid- and cholesterol-enriched lipid microdomains attract [...] Read more.
Salmonella spp. remains a major public health problem for the whole world. To reduce the use of antimicrobial agents and drug-resistant Salmonella, a better strategy is to explore alternative therapy rather than to discover another antibiotic. Sphingolipid- and cholesterol-enriched lipid microdomains attract signaling proteins and orchestrate them toward cell signaling and membrane trafficking pathways. Recent studies have highlighted the crucial role of sphingolipids in the innate immunity against infecting pathogens. It is therefore mandatory to exploit the role of the membrane sphingolipids in the innate immunity of intestinal epithelia infected by this pathogen. In the present review, we focus on the role of sphingolipids in the innate immunity of intestinal epithelia against Salmonella infection, including adhesion, autophagy, bactericidal effect, barrier function, membrane trafficking, cytokine and antimicrobial peptide expression. The intervention of sphingolipid-enhanced foods to make our life healthy or pharmacological agents regulating sphingolipids is provided at the end. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Figure 1

1168 KiB  
Review
A Novel Perspective on the ApoM-S1P Axis, Highlighting the Metabolism of ApoM and Its Role in Liver Fibrosis and Neuroinflammation
by Stefan Hajny and Christina Christoffersen
Int. J. Mol. Sci. 2017, 18(8), 1636; https://doi.org/10.3390/ijms18081636 - 27 Jul 2017
Cited by 22 | Viewed by 7215
Abstract
Hepatocytes, renal proximal tubule cells as well as the highly specialized endothelium of the blood brain barrier (BBB) express and secrete apolipoprotein M (apoM). ApoM is a typical lipocalin containing a hydrophobic binding pocket predominantly carrying Sphingosine-1-Phosphate (S1P). The small signaling molecule S1P [...] Read more.
Hepatocytes, renal proximal tubule cells as well as the highly specialized endothelium of the blood brain barrier (BBB) express and secrete apolipoprotein M (apoM). ApoM is a typical lipocalin containing a hydrophobic binding pocket predominantly carrying Sphingosine-1-Phosphate (S1P). The small signaling molecule S1P is associated with several physiological as well as pathological pathways whereas the role of apoM is less explored. Hepatic apoM acts as a chaperone to transport S1P through the circulation and kidney derived apoM seems to play a role in S1P recovery to prevent urinal loss. Finally, polarized endothelial cells constituting the lining of the BBB express apoM and secrete the protein to the brain as well as to the blood compartment. The review will provide novel insights on apoM and S1P, and its role in hepatic fibrosis, neuroinflammation and BBB integrity. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

1165 KiB  
Review
Sphingosine-1-Phosphate Metabolism and Its Role in the Development of Inflammatory Bowel Disease
by Tomasz Wollny, Marzena Wątek, Bonita Durnaś, Katarzyna Niemirowicz, Ewelina Piktel, Małgorzata Żendzian-Piotrowska, Stanisław Góźdź and Robert Bucki
Int. J. Mol. Sci. 2017, 18(4), 741; https://doi.org/10.3390/ijms18040741 - 31 Mar 2017
Cited by 29 | Viewed by 7052
Abstract
Beyond their role as structural molecules, sphingolipids are involved in many important cellular processes including cell proliferation, apoptosis, inflammation, and migration. Altered sphingolipid metabolism is observed in many pathological conditions including gastrointestinal diseases. Inflammatory bowel disease (IBD) represents a state of complex, unpredictable, [...] Read more.
Beyond their role as structural molecules, sphingolipids are involved in many important cellular processes including cell proliferation, apoptosis, inflammation, and migration. Altered sphingolipid metabolism is observed in many pathological conditions including gastrointestinal diseases. Inflammatory bowel disease (IBD) represents a state of complex, unpredictable, and destructive inflammation of unknown origin within the gastrointestinal tract. The mechanisms explaining the pathophysiology of IBD involve signal transduction pathways regulating gastro-intestinal system’s immunity. Progressive intestinal tissue destruction observed in chronic inflammation may be associated with an increased risk of colon cancer. Sphingosine-1-phosphate (S1P), a sphingolipid metabolite, functions as a cofactor in inflammatory signaling and becomes a target in the treatment of IBD, which might prevent its conversion to cancer. This paper summarizes new findings indicating the impact of (S1P) on IBD development and IBD-associated carcinogenesis. Full article
(This article belongs to the Special Issue Sphingolipids: Signals and Disease)
Show Figures

Graphical abstract

Back to TopTop