Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 502
Filter
1.
Endocrinology ; 164(3)2023 01 09.
Article in English | MEDLINE | ID: mdl-36690339

ABSTRACT

Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid that regulates fundamental cellular processes such as proliferation, migration, apoptosis, and differentiation through 5 cognate G protein-coupled receptors (S1P1-S1P5). We previously demonstrated that blockade of S1P2 signaling in S1P2-deficient mice attenuates high-fat diet-induced adipocyte hypertrophy and glucose intolerance and an S1P2-specific antagonist JTE-013 inhibits, whereas an S1P1/S1P3 dual antagonist (VPC23019) activates, adipogenic differentiation of preadipocytes. Based on those observations, this study examined whether an S1P1-specific agonist, SEW-2871, VPC23019, or their combination acts on obesity and glucose intolerance in leptin-deficient ob/ob mice. The oral administration of SEW-2871 or JTE-013 induced significant reductions in body/epididymal fat weight gains and epididymal/inguinal fat adipocyte sizes and improved glucose intolerance and adipocyte inflammation in ob/ob mice but not in their control C57BL/6J mice. Both SEW-2871 and JTE-013 decreased messenger RNA levels of tumor necrosis factor-α and CD11c, whereas they increased those of CD206 and adiponectin in the epididymal fats isolated from ob/ob mice with no changes in the levels of peroxisome proliferator activated receptor γ and its regulated genes. By contrast, VPC23019 did not cause any such alterations but counteracted with all those SEW-2871 actions in these mice. In conclusion, the S1P1 agonist SEW-2871 acted like the S1P2 antagonist JTE-013 to reduce body/epididymal fats and improve glucose tolerance in obese mice. Therefore, this study raises the possibility that endogenous S1P could promote obesity/type 2 diabetes through the S1P2, whereas exogenous S1P could act against them through the S1P1.


Subject(s)
Diabetes Mellitus, Type 2 , Glucose Intolerance , Animals , Male , Mice , Glucose , Lysophospholipids/pharmacology , Lysophospholipids/physiology , Mice, Inbred C57BL , Mice, Obese , Obesity , Receptors, Lysosphingolipid/genetics , Sphingosine/pharmacology , Sphingosine/physiology , Sphingosine-1-Phosphate Receptors
2.
Article in English | MEDLINE | ID: mdl-35667710

ABSTRACT

Extracellular signals act on G-protein-coupled receptors (GPCRs) to regulate homeostasis and adapt to stress. This involves rapid intracellular post-translational responses and long-lasting gene-expression changes that ultimately determine cellular phenotype and fate changes. The lipid mediator sphingosine 1-phosphate (S1P) and its receptors (S1PRs) are examples of well-studied GPCR signaling axis essential for vascular development, homeostasis, and diseases. The biochemical cascades involved in rapid S1P signaling are well understood. However, gene-expression regulation by S1PRs are less understood. In this review, we focus our attention to how S1PRs regulate nuclear chromatin changes and gene transcription to modulate vascular and lymphatic endothelial phenotypic changes during embryonic development and adult homeostasis. Because S1PR-targeted drugs approved for use in the treatment of autoimmune diseases cause substantial vascular-related adverse events, these findings are critical not only for general understanding of stimulus-evoked gene regulation in the vascular endothelium, but also for therapeutic development of drugs for autoimmune and perhaps vascular diseases.


Subject(s)
Signal Transduction , Sphingosine , Signal Transduction/physiology , Sphingosine/physiology , Receptors, G-Protein-Coupled , Endothelium, Vascular
3.
FEBS Open Bio ; 11(11): 3081-3089, 2021 11.
Article in English | MEDLINE | ID: mdl-34535977

ABSTRACT

Free fatty acid receptor 4 (FFAR4)/GPR120 comprises a receptor for medium- and long-chain fatty acids. We previously identified phytosphingosine (PHS) as a novel ligand of FFAR4. Although many natural FFAR4 ligands have carboxyl groups, PHS does not, thus suggesting that binding to FFAR4 is driven by a completely different mechanism than other natural ligands such as α-linolenic acid (ALA). To test this hypothesis, we performed docking simulation analysis using a FFAR4 homology model based on a protein model derived from the crystal structure of activated turkey beta-1 adrenoceptor. The docking simulation revealed that the probable hydrogen bonds to FFAR4 differ between various ligands. In particular, binding was predicted between R264 of the FFAR4 and the oxygen of the carboxylate group in ALA, as well as between E249 of the FFAR4 and the oxygen of the hydroxy group at the C4-position in PHS. Alanine substitution at E249 (E249A) dramatically reduced PHS-induced FFAR4 activation but demonstrated a weaker effect on ALA-induced FFAR4 activation. Kinetic analysis and Km values clearly demonstrated that the E249A substitution resulted in reduced affinity for PHS but not for ALA. Additionally, we observed that sphingosine, lacking a hydroxyl group at C4-position, could not activate FFAR4. Our data show that E249 of the FFAR4 receptor is crucial for binding to the hydroxy group at the C4-position in PHS, and this is a completely different molecular mechanism of binding from ALA. Because GPR120 agonists have attracted attention as treatments for type 2 diabetes, our findings may provide new insights into their development.


Subject(s)
Sphingosine/analogs & derivatives , Sphingosine/metabolism , Cell Communication , Fatty Acids , HEK293 Cells , Humans , Kinetics , Ligands , Molecular Docking Simulation/methods , Protein Binding , Receptors, G-Protein-Coupled , Sphingosine/physiology
4.
Int J Mol Sci ; 22(16)2021 Aug 20.
Article in English | MEDLINE | ID: mdl-34445695

ABSTRACT

Accumulating evidence indicates that the molecular pathways mediating wound healing induce cell migration and localization of cytokines to sites of injury. Macrophages are immune cells that sense and actively respond to disturbances in tissue homeostasis by initiating, and subsequently resolving, inflammation. Hypoxic conditions generated at a wound site also strongly recruit macrophages and affect their function. Hypoxia inducible factor (HIF)-1α is a transcription factor that contributes to both glycolysis and the induction of inflammatory genes, while also being critical for macrophage activation. For the latter, HIF-1α regulates sphingosine 1-phosphate (S1P) to affect the migration, activation, differentiation, and polarization of macrophages. Recently, S1P and HIF-1α have received much attention, and various studies have been performed to investigate their roles in initiating and resolving inflammation via macrophages. It is hypothesized that the HIF-1α/S1P/S1P receptor axis is an important determinant of macrophage function under inflammatory conditions and during disease pathogenesis. Therefore, in this review, biological regulation of monocytes/macrophages in response to circulating HIF-1α is summarized, including signaling by S1P/S1P receptors, which have essential roles in wound healing.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Wound Healing/physiology , Animals , Cell Differentiation/genetics , Cell Movement/physiology , Cytokines/metabolism , Gene Expression/genetics , Gene Expression Regulation/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Inflammation/metabolism , Lysophospholipids/physiology , Macrophage Activation/physiology , Macrophages/metabolism , Macrophages/physiology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Sphingosine/metabolism , Sphingosine/physiology
5.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34298977

ABSTRACT

For decades, lipids were confined to the field of structural biology and energetics as they were considered only structural constituents of cellular membranes and efficient sources of energy production. However, with advances in our understanding in lipidomics and improvements in the technological approaches, astounding discoveries have been made in exploring the role of lipids as signaling molecules, termed bioactive lipids. Among these bioactive lipids, sphingolipids have emerged as distinctive mediators of various cellular processes, ranging from cell growth and proliferation to cellular apoptosis, executing immune responses to regulating inflammation. Recent studies have made it clear that sphingolipids, their metabolic intermediates (ceramide, sphingosine-1-phosphate, and N-acetyl sphingosine), and enzyme systems (cyclooxygenases, sphingosine kinases, and sphingomyelinase) harbor diverse yet interconnected signaling pathways in the central nervous system (CNS), orchestrate CNS physiological processes, and participate in a plethora of neuroinflammatory and neurodegenerative disorders. Considering the unequivocal importance of sphingolipids in CNS, we review the recent discoveries detailing the major enzymes involved in sphingolipid metabolism (particularly sphingosine kinase 1), novel metabolic intermediates (N-acetyl sphingosine), and their complex interactions in CNS physiology, disruption of their functionality in neurodegenerative disorders, and therapeutic strategies targeting sphingolipids for improved drug approaches.


Subject(s)
Central Nervous System/physiopathology , Inflammation/physiopathology , Membrane Lipids/physiology , Models, Biological , Nerve Degeneration/physiopathology , Neurodegenerative Diseases/physiopathology , Sphingolipids/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Ceramides/physiology , Eicosanoids/physiology , Forecasting , Homeostasis , Humans , Inflammation/pathology , Lipoxygenase/physiology , Lysophospholipids/physiology , Nerve Degeneration/pathology , Neurodegenerative Diseases/pathology , Neuroglia/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Prostaglandin-Endoperoxide Synthases/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology
6.
J Diabetes Res ; 2021: 4932974, 2021.
Article in English | MEDLINE | ID: mdl-33791388

ABSTRACT

OBJECTIVES: This study was designed to explore the intellectual landscape of research into the application of sphingosine 1 phosphate (S1P) in age-related diseases and to identify thematic development trends and research frontiers in this area. METHODS: Scientometric research was conducted by analyzing bibliographic records retrieved from the Web of Science (WOS) Sci-Expanded Database dated between 1900 and 2020. Countries, institutions, authors, keyword occurrence analysis, and cooperation network analysis were performed using the CiteSpace and VOSviewer software. RESULTS: A total of 348 valid records were included in the final dataset, and the number of publications and the frequency of citations have grown rapidly over the last ten years. The USA (n = 175), China (n = 42), and Germany (n = 37) were the three largest contributors to the global publications on S1P and aging, while the Medical University of South Carolina (n = 15), University of California, San Francisco (n = 13), and University of Toronto (n = 13) were the leading institutions in this field. Analysis showed that early studies primarily focused on the mechanism of S1P intervention in AD. While S1P and its relevant metabolites have remained a long-term active area of research, recent studies have focused more on interventions aimed at improving retinal degeneration, cardiomyopathy, multiple sclerosis, and diabetes, among others. CONCLUSIONS: It is worth mentioning that this manuscript is the first to describe any bibliometric analysis of S1P and its application in age-related interventions. This study includes a discussion of the (1) historical overview of the topic; (2) main contributors: journals, countries, institutes, funding agencies, and authors; (3) collaboration between institutes and authors; (4) research hot spots and zones; and 5) research trends and frontiers. This will enable scholars to understand the current status of S1P research in age-related diseases.


Subject(s)
Aging , Biomedical Research/trends , Lysophospholipids/physiology , Sphingosine/analogs & derivatives , Humans , Sphingosine/physiology
7.
Hum Cell ; 34(3): 734-744, 2021 May.
Article in English | MEDLINE | ID: mdl-33683655

ABSTRACT

In malaria-endemic countries, the burden of hypertension is on the rise. Although malaria and hypertension seem to have no direct link, several studies in recent years support their possible link. Three bioactive molecules such as angiotensin II (Ang II), bradykinin (BK) and sphingosine 1-phosphate (S1P) are crucial in regulating blood pressure. While the increased level of Ang II and S1P are responsible for inducing hypertension, BK is arthero-protective and anti-hypertensive. Therefore, in the present review, based on available literatures we highlight the present knowledge on the production and bioavailability of these molecules, the mechanism of their regulation of hypertension, and patho-physiological role in malaria. Further, a possible link between malaria and hypertension is hypothesized through various arguments based on experimental evidence. Understanding of their mechanisms of blood pressure regulation during malaria infection may open up avenues for drug therapeutics and management of malaria in co-morbidity with hypertension.


Subject(s)
Angiotensin II/physiology , Bradykinin/physiology , Hypertension/etiology , Lysophospholipids/physiology , Malaria/complications , Sphingosine/analogs & derivatives , Blood Pressure , Comorbidity , Female , Humans , Hypertension/epidemiology , Malaria/epidemiology , Male , Pregnancy , Sphingosine/physiology
8.
Neuromolecular Med ; 23(1): 47-67, 2021 03.
Article in English | MEDLINE | ID: mdl-33180310

ABSTRACT

Sphingosine 1-phosphates (S1Ps) are bioactive lipids that mediate a diverse range of effects through the activation of cognate receptors, S1P1-S1P5. Scrutiny of S1P-regulated pathways over the past three decades has identified important and occasionally counteracting functions in the brain and cerebrovascular system. For example, while S1P1 and S1P3 mediate proinflammatory effects on glial cells and directly promote endothelial cell barrier integrity, S1P2 is anti-inflammatory but disrupts barrier integrity. Cumulatively, there is significant preclinical evidence implicating critical roles for this pathway in regulating processes that drive cerebrovascular disease and vascular dementia, both being part of the continuum of vascular cognitive impairment (VCI). This is supported by clinical studies that have identified correlations between alterations of S1P and cognitive deficits. We review studies which proposed and evaluated potential mechanisms by which such alterations contribute to pathological S1P signaling that leads to VCI-associated chronic neuroinflammation and neurodegeneration. Notably, S1P receptors have divergent but overlapping expression patterns and demonstrate complex interactions. Therefore, the net effect produced by S1P represents the cumulative contributions of S1P receptors acting additively, synergistically, or antagonistically on the neural, vascular, and immune cells of the brain. Ultimately, an optimized therapeutic strategy that targets S1P signaling will have to consider these complex interactions.


Subject(s)
Dementia, Vascular/physiopathology , Lysophospholipids/physiology , Sphingosine-1-Phosphate Receptors/physiology , Sphingosine/analogs & derivatives , Aldehyde-Lyases/antagonists & inhibitors , Aldehyde-Lyases/physiology , Alzheimer Disease/physiopathology , Animals , Cerebrovascular Disorders/physiopathology , Clinical Trials as Topic , Drug Delivery Systems , Drug Evaluation, Preclinical , Fingolimod Hydrochloride/therapeutic use , Humans , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/physiopathology , Inflammation , Ischemic Stroke/drug therapy , Ischemic Stroke/physiopathology , Mice , Mice, Knockout , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/physiology , Signal Transduction , Sphingosine/physiology , Sphingosine-1-Phosphate Receptors/drug effects
9.
Neuromolecular Med ; 23(1): 211-223, 2021 03.
Article in English | MEDLINE | ID: mdl-32914259

ABSTRACT

Sphingosine 1-phosphate (S1P) is an important lipid biomolecule that exerts pleiotropic cellular actions as it binds to and activates its five G-protein-coupled receptors, S1P1-5. Through these receptors, S1P can mediate diverse biological activities in both healthy and diseased conditions. S1P is produced by S1P-producing enzymes, sphingosine kinases (SphK1 and SphK2), and is abundantly present in different organs, including the brain. The medically important roles of receptor-mediated S1P signaling are well characterized in multiple sclerosis because FTY720 (Gilenya™, Novartis), a non-selective S1P receptor modulator, is currently used as a treatment for this disease. In cerebral ischemia, its role is also notable because of FTY720's efficacy in both rodent models and human patients with cerebral ischemia. In particular, some of the S1P receptors, including S1P1, S1P2, and S1P3, have been identified as pathogenic players in cerebral ischemia. Other than these receptors, S1P itself and S1P-producing enzymes have been shown to play certain roles in cerebral ischemia. This review aims to compile the current updates and overviews about the roles of S1P signaling, along with a focus on S1P receptors in cerebral ischemia, based on recent studies that used in vivo rodent models of cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , Lysophospholipids/physiology , Nerve Tissue Proteins/physiology , Sphingosine-1-Phosphate Receptors/physiology , Sphingosine/analogs & derivatives , Animals , Brain Damage, Chronic/etiology , Brain Damage, Chronic/metabolism , Brain Ischemia/complications , Clinical Trials as Topic , Disease Models, Animal , Drug Evaluation, Preclinical , Fingolimod Hydrochloride/therapeutic use , Humans , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Inflammation , Ischemic Stroke/drug therapy , Neovascularization, Physiologic/drug effects , Neuroprotective Agents/therapeutic use , Phosphotransferases (Alcohol Group Acceptor)/physiology , Rats , Signal Transduction/physiology , Sphingosine/physiology
10.
Blood ; 137(5): 690-701, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33232973

ABSTRACT

Transfusion-related acute lung injury (TRALI) is a hazardous transfusion complication with an associated mortality of 5% to 15%. We previously showed that stored (5 days) but not fresh platelets (1 day) cause TRALI via ceramide-mediated endothelial barrier dysfunction. As biological ceramides are hydrophobic, extracellular vesicles (EVs) may be required to shuttle these sphingolipids from platelets to endothelial cells. Adding to complexity, EV formation in turn requires ceramide. We hypothesized that ceramide-dependent EV formation from stored platelets and EV-dependent sphingolipid shuttling induces TRALI. EVs formed during storage of murine platelets were enumerated, characterized for sphingolipids, and applied in a murine TRALI model in vivo and for endothelial barrier assessment in vitro. Five-day EVs were more abundant, had higher long-chain ceramide (C16:0, C18:0, C20:0), and lower sphingosine-1-phosphate (S1P) content than 1-day EVs. Transfusion of 5-day, but not 1-day, EVs induced characteristic signs of lung injury in vivo and endothelial barrier disruption in vitro. Inhibition or supplementation of ceramide-forming sphingomyelinase reduced or enhanced the formation of EVs, respectively, but did not alter the injuriousness per individual EV. Barrier failure was attenuated when EVs were abundant in or supplemented with S1P. Stored human platelet 4-day EVs were more numerous compared with 2-day EVs, contained more long-chain ceramide and less S1P, and caused more endothelial cell barrier leak. Hence, platelet-derived EVs become more numerous and more injurious (more long-chain ceramide, less S1P) during storage. Blockade of sphingomyelinase, EV elimination, or supplementation of S1P during platelet storage may present promising strategies for TRALI prevention.


Subject(s)
Extracellular Vesicles/physiology , Platelet Transfusion/adverse effects , Sphingolipids/metabolism , Transfusion-Related Acute Lung Injury/etiology , Animals , Blood Platelets/ultrastructure , Blood Preservation , Ceramides/metabolism , Endothelial Cells/physiology , Endotoxins/toxicity , Humans , Lysophospholipids/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology , Transfusion-Related Acute Lung Injury/metabolism , Transfusion-Related Acute Lung Injury/prevention & control
11.
Cell Death Dis ; 11(10): 892, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33093451

ABSTRACT

Breast cancer is one of the most common female malignant cancers. Biorhythm disorder largely increases the risk of breast cancer. We aimed to investigate the biological functions and molecular mechanisms of circadian gene TIMELESS circadian regulator (TIM) in estrogen receptor (ER)-positive breast cancer and provide a new therapeutic target for breast cancer patients. Here, we explored that the expression of TIM was elevated in breast cancer, and high expression of TIM in cancer tissues was associated with poor prognosis, especially in the ER-positive breast cancer patients. In addition, we found that TIM promoted cell proliferation and enhanced mitochondrial respiration. TIM interacted with specificity protein 1 (Sp1) which contributes to upregulate the expression of alkaline ceramidase 2 (ACER2). Moreover, ACER2 is responsible for TIM-mediated promotive effects of cell growth and mitochondrial respiration. Collectively, our research unveiled a novel function of TIM in sphingolipid metabolism through interaction with Sp1. It provides a new theoretical explanation for the pathogenesis of breast cancer, and targeting TIM may serve as a potential therapeutic target for ER-positive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Sphingolipids/metabolism , Alkaline Ceramidase/physiology , Animals , Biopsy , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lysophospholipids/physiology , Mice , Mice, Nude , Mitochondria/drug effects , Sp1 Transcription Factor/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology , Xenograft Model Antitumor Assays
12.
Environ Toxicol ; 35(9): 991-997, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32401414

ABSTRACT

Rheumatoid arthritis (RA) is a systemic autoimmune inflammatory disease, in which the immune system attacks synovial joint tissues. Interleukin (IL)-1ß is a critical proinflammatory cytokine in RA progression. Sphingosine-1-phosphate (S1P), a platelet-derived lysophospholipid mediator, reportedly regulates osteoimmunology. Here, we investigated how S1P mediates IL-1ß expression in osteoblasts. Our analysis of records from the Gene Expression Omnibus (GEO) database demonstrate higher levels of IL-1ß in patients with RA compared with those with osteoarthritis. Stimulation of osteoblasts with S1P concentration dependently increased mRNA and protein expression of IL-1ß. Elevations in IL-1ß mRNA expression induced by S1P were reduced by the small interfering RNA (siRNA) against the S1P1 receptor. S1P also augmented JAK and STAT3 molecular cascades. We also found that JAK and STAT3 inhibitors and their siRNAs antagonized S1P-promoted IL-1ß expression. Our results indicate that S1P promotes the expression of IL-1ß in osteoblasts via the S1P1 receptor and the JAK and STAT3 signaling pathways.


Subject(s)
Interleukin-1beta/genetics , Janus Kinases/metabolism , Lysophospholipids/physiology , Osteoblasts/metabolism , STAT3 Transcription Factor/metabolism , Sphingosine/analogs & derivatives , Arthritis, Rheumatoid/metabolism , Cells, Cultured , Humans , Lysophospholipids/pharmacology , Male , Osteoarthritis/metabolism , Osteoblasts/drug effects , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Sphingosine/pharmacology , Sphingosine/physiology , Sphingosine-1-Phosphate Receptors/genetics
13.
Chin Med J (Engl) ; 133(3): 292-300, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31904729

ABSTRACT

BACKGROUND: Baicalein has been shown to have anti-inflammatory and anti-tumor activities. However, the mechanisms underlying its anti-inflammatory effect on colitis remain unclear. METHODS: A dextran sodium sulfate (DSS)-induced model of acute colitis was established in BALB/c mice (6-8 weeks old, weighing 18-22 g). Six groups of mice received: (1) water for 10 days (control), n = 6; (2) DSS 4% solution in the drinking water for 7 days, followed by normal water for 3 days, n = 7; (3), (4), and (5) as for group 2 plus baicalein (10, 20, 40 mg/kg) administered once daily starting on day 1, n = 6; and (6) as for (2) plus 5-aminosalicylic acid (50 mg/kg) administered once daily starting on day 1, n = 6. Body weights, stool consistency, and hematochezia were recorded, and the severity of colitis was evaluated using a disease activity index. On day 11, the mice were euthanized, and organs and blood were collected for analysis. Serum inflammatory factors were detected by enzyme-linked immunosorbent assay; CD11b-positive cells were analyzed by immunofluorescence microscopy; expression of retinoic-acid-receptor-related orphan nuclear receptor gamma, sphingosine kinase 1 (SPHK1), and phosphorylated signal transducer and activator of transcription 3 (p-STAT3) was detected by immunohistochemistry; and expression of nucleotide-binding oligomerization domain 2 (NOD2), SPHK1, sphingosine 1-phosphate receptor 1 (S1PR1), total STAT3, and p-STAT3 were detected by western blotting analysis. Inter-group differences were compared using Student's t test. RESULTS: Baicalein treatment dose-dependently reduced DSS-induced weight loss (P < 0.01 or P < 0.05), splenomegaly (P < 0.01), and colonic damage, as reflected by amelioration of diarrhea, rectal bleeding, and colonic ulceration, congestion, edema (shown as colon length, P < 0.05 or P < 0.01), and inflammatory cell infiltration. Baicalein also significantly decreased the levels of inflammatory mediators in the serum (P < 0.01) and colon, and significantly inhibited expression of NOD2 SPHK1, S1PR1, and p-STAT3 in the colon (P < 0.05). CONCLUSIONS: Baicalein treatment ameliorated colitis in mice by inhibiting S1P-STAT3 signaling, suggesting that this flavonoid might be beneficial in the treatment of colitis.


Subject(s)
Colitis/prevention & control , Flavanones/therapeutic use , Lysophospholipids/physiology , STAT3 Transcription Factor/physiology , Sphingosine/analogs & derivatives , Animals , Colitis/chemically induced , Dextran Sulfate/pharmacology , Female , Flavanones/pharmacology , Mice , Mice, Inbred BALB C , Nod2 Signaling Adaptor Protein/analysis , Phosphotransferases (Alcohol Group Acceptor)/analysis , STAT3 Transcription Factor/analysis , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/physiology , Sphingosine-1-Phosphate Receptors/analysis
14.
Front Immunol ; 11: 620348, 2020.
Article in English | MEDLINE | ID: mdl-33633739

ABSTRACT

Alzheimer's disease (AD) is the leading cause of dementia worldwide giving rise to devastating forms of cognitive decline, which impacts patients' lives and that of their proxies. Pathologically, AD is characterized by extracellular amyloid deposition, neurofibrillary tangles and chronic neuroinflammation. To date, there is no cure that prevents progression of AD. In this review, we elaborate on how bioactive lipids, including sphingolipids (SL) and specialized pro-resolving lipid mediators (SPM), affect ongoing neuroinflammatory processes during AD and how we may exploit them for the development of new biomarker panels and/or therapies. In particular, we here describe how SPM and SL metabolism, ranging from ω-3/6 polyunsaturated fatty acids and their metabolites to ceramides and sphingosine-1-phosphate, initiates pro- and anti-inflammatory signaling cascades in the central nervous system (CNS) and what changes occur therein during AD pathology. Finally, we discuss novel therapeutic approaches to resolve chronic neuroinflammation in AD by modulating the SPM and SL pathways.


Subject(s)
Alzheimer Disease/metabolism , Fatty Acids, Omega-3/physiology , Fatty Acids, Omega-6/physiology , Sphingolipids/physiology , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Central Nervous System/metabolism , Ceramides/antagonists & inhibitors , Ceramides/physiology , Disease Models, Animal , Fatty Acids, Unsaturated/metabolism , Forecasting , Humans , Inflammation , Lipoxygenases/metabolism , Lysophospholipids/physiology , Mice , Microglia/pathology , Models, Biological , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Pattern Recognition/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology , Sphingosine 1 Phosphate Receptor Modulators/therapeutic use
15.
Ann Endocrinol (Paris) ; 80(5-6): 263-272, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31455516

ABSTRACT

Sphingosine-1-phoshate (S1P) is a membrane sphingolipid involved in several physiological processes, including cell proliferation, tissue growth, cell survival and migration, inflammation, vasculogenesis, and angiogenesis. Herein, we review the most critical effects of S1P on ovarian function, including its physiological and pathophysiological effects. Based on the available evidence, S1P plays an important role in ovarian physiology, participating as an essential stimulator of follicular development in both the preantral and antral phases, as well as in ovulation and corpus luteum development. Moreover, S1P may be a good cytoprotective agent against cancer treatment side-effects (chemotherapy with or without radiation therapy). In the future, this compound may be given for fertility preservation to women undergoing cancer treatment. However, further studies are required to confirm its efficacy in ovarian protection and also its safety in terms of cancer prognosis, given the biological action of the compound. Under- or over-production of S1P may be related to ovarian pathologies.


Subject(s)
Lysophospholipids/physiology , Ovarian Diseases/physiopathology , Ovary/physiopathology , Sphingosine/analogs & derivatives , Animals , Cell Proliferation , Corpus Luteum/growth & development , Female , Fertility Preservation , Humans , Ovarian Diseases/pathology , Ovarian Follicle/growth & development , Ovarian Neoplasms/pathology , Ovarian Neoplasms/physiopathology , Ovary/pathology , Sphingosine/physiology , Sphingosine-1-Phosphate Receptors/physiology
16.
FEBS J ; 286(19): 3909-3925, 2019 10.
Article in English | MEDLINE | ID: mdl-31162791

ABSTRACT

Trypanosoma cruzi is the causative agent of Chagas disease. The only two drugs accepted for the treatment of this infection are benznidazole and nifurtimox, which are of limited use in the predominant chronic phase. On the search for new drugs, the intracellular Ca2+ regulation has been postulated as a possible target, due to differences found between host cells and the parasite. The mechanisms involved in the intracellular Ca2+ regulation of T. cruzi have been partially elucidated. However, nothing is known about a putative channel responsible for the Ca2+ entry into this parasite. In contrast, in Leishmania spp., a closely related hemoflagelate, a sphingosine-activated plasma membrane Ca2+ channel has been recently described. The latter resembles the L-type voltage-gated Ca2+ channel present in humans, but with distinct characteristics. This channel is one of the main targets concerning the mechanism of action of miltefosine, the unique oral drug approved against leishmaniasis. In the present work, we describe for the first time, the electrophysiological characterization of a sphingosine-activated Ca2+ channel of T. cruzi by reconstituting plasma membrane vesicles into giant liposomes and patch clamp. This channel shares some characteristic as activation by Bay K8644 and inhibition by channel blockers such as nifedipine. However, the T. cruzi channel differs from the L-type VGCC in its activation by sphingosine and/or miltefosine. Albeit the conductance for each, Ba2+ , Ca2+ and Sr2+ was similar, the parasite channel appears not to be voltage dependent. A gene that presents homology in critical amino acids with its human ortholog Ca2+ channel was identified.


Subject(s)
Calcium Channels/physiology , Sphingosine/physiology , Trypanosoma cruzi/physiology , Animals , Antiprotozoal Agents/pharmacology , Calcium/metabolism , Calcium Channels/drug effects , Ion Transport , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology
17.
Arterioscler Thromb Vasc Biol ; 39(8): 1550-1564, 2019 08.
Article in English | MEDLINE | ID: mdl-31189429

ABSTRACT

Despite decades of therapeutic advances, myocardial infarction remains a leading cause of death worldwide. Recent studies have identified HDLs (high-density lipoproteins) as a potential candidate for mitigating coronary ischemia/reperfusion injury via a broad spectrum of signaling pathways. HDL ligands, such as S1P (sphingosine-1-phosphate), Apo (apolipoprotein) A-I, clusterin, and miRNA, may influence the opening of the mitochondrial channel, insulin sensitivity, and production of vascular autacoids, such as NO, prostacyclin, and endothelin-1. In parallel, antioxidant activity and sequestration of oxidized molecules provided by HDL can attenuate the oxidative stress that triggers ischemia/reperfusion. Nevertheless, during myocardial infarction, oxidation and the capture of oxidized and proinflammatory molecules generate large phenotypic and functional changes in HDL, potentially limiting its beneficial properties. In this review, new findings from cellular and animal models, as well as from clinical studies, will be discussed to describe the cardioprotective benefits of HDL on myocardial infarction. Furthermore, mechanisms by which HDL modulates cardiac function and potential strategies to mitigate postmyocardial infarction risk damage by HDL will be detailed throughout the review.


Subject(s)
Lipoproteins, HDL/physiology , Myocardial Infarction/prevention & control , Animals , Cholesterol/metabolism , Endothelial Cells/physiology , Glucose/metabolism , Homeostasis , Humans , Lipoproteins, HDL/blood , Lysophospholipids/physiology , Oxidative Stress , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology
18.
Front Immunol ; 10: 596, 2019.
Article in English | MEDLINE | ID: mdl-30972080

ABSTRACT

Bone is a highly dynamic organ that is continuously being remodeled by the reciprocal interactions between bone and immune cells. We have originally established an advanced imaging system for visualizing the in vivo behavior of osteoclasts and their precursors in the bone marrow cavity using two-photon microscopy. Using this system, we found that the blood-enriched lipid mediator, sphingosine-1-phosphate, controlled the migratory behavior of osteoclast precursors. We also developed pH-sensing chemical fluorescent probes to detect localized acidification by bone-resorbing osteoclasts on the bone surface in vivo, and identified two distinct functional states of differentiated osteoclasts, "bone-resorptive" and "non-resorptive." Here, we summarize our studies on the dynamics and functions of bone and immune cells within the bone marrow. We further discuss how our intravital imaging techniques can be applied to evaluate the mechanisms of action of biological agents in inflammatory bone destruction. Our intravital imaging techniques would be beneficial for studying the cellular dynamics in arthritic inflammation and bone destruction in vivo and would also be useful for evaluating novel therapies in animal models of bone-destroying diseases.


Subject(s)
Bone Resorption/etiology , Bone and Bones/diagnostic imaging , Cell Communication , Osteoclasts/physiology , T-Lymphocytes/physiology , Animals , Lysophospholipids/physiology , Macrophages/physiology , Mice , Microscopy, Fluorescence, Multiphoton , Osteoblasts/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology
19.
Oncogene ; 38(24): 4788-4803, 2019 06.
Article in English | MEDLINE | ID: mdl-30816345

ABSTRACT

A role of sphingolipids for inflammatory bowel disease and cancer is evident. However, the relative and separate contribution of sphingolipid deterioration in inflammation versus carcinogenesis for the pathophysiology of colitis-associated colon cancer (CAC) was unknown and therefore examined in this study. We performed isogenic bone marrow transplantation of inducible sphingosine-1-phosphate (S1P) lyase knockout mice to specifically modulate sphingolipids and associated genes and proteins in a compartment-specific way in a DSS/AOM mediated CAC model. 3D organoid cultures were used in vitro. S1P lyase (SGPL1) knockout in either immune cells or tissue, caused local sphingolipid accumulation leading to a dichotomic development of CAC: Immune cell SGPL1 knockout (I-SGPL-/-) augmented massive immune cell infiltration initiating colitis with lesions and calprotectin increase. Pathological crypt remodeling plus extracellular S1P-signaling caused delayed tumor formation characterized by S1P receptor 1, STAT3 mRNA increase, as well as programmed cell death ligand 1 expression, accompanied by a putatively counter regulatory STAT1S727 phosphorylation. In contrast, tissue SGPL1 knockout (T-SGPL-/-) provoked immediate occurrence of epithelial-driven tumors with upregulated sphingosine kinase 1, S1P receptor 2 and epidermal growth factor receptor. Here, progressing carcinogenesis was accompanied by an IL-12 to IL-23 shift with a consecutive development of a Th2/GATA3-driven, tumor-favoring microenvironment. Moreover, the knockout models showed distinct lymphopenia and neutrophilia, different from the full SGPL1 knockout. This study shows that depending on the initiating cellular S1P source, the pathophysiology of inflammation-induced cancer versus cancer-induced inflammation develops through separate, discernible molecular steps.


Subject(s)
Aldehyde-Lyases/physiology , Carcinogenesis , Colitis/etiology , Colonic Neoplasms/complications , Inflammation/etiology , Aldehyde-Lyases/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cells, Cultured , Colitis/genetics , Colitis/pathology , Colon/metabolism , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Inflammation/genetics , Lysophospholipids/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology , Tumor Microenvironment/physiology
20.
FASEB J ; 33(6): 7180-7191, 2019 06.
Article in English | MEDLINE | ID: mdl-30844311

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a critical process implicated in the initial stage of cancer metastasis, which is the major cause of tumor recurrence and mortality. Although key transcription factors that regulate EMT, such as snail family transcriptional repressor 2 (SNAI2), are well characterized, the upstream signaling pathways controlling these transcriptional mediators are largely unknown, which limits therapeutic strategies. Sphingosine 1-phosphate (S1P) is a bioactive lipid mediator, generated by sphingosine kinases (SPHK1 and SPHK2), that mainly exerts its effects by binding to the following 5 GPCRs: S1P1 to S1P5. S1P signaling has been reported to regulate different aspects of cancer progression including cell proliferation, apoptosis, and migration; nevertheless, its role in cancer metastasis, specifically via EMT, is not established. Here we show that SPHK1 expression correlates significantly with EMT score in breast cancer cell lines, and with SNAI2 in patient-derived breast tumors. Cell-based assays demonstrate that S1P can rapidly up-regulate the expression of SNAI2 in breast cancer cells via the activation of cognate receptors S1P2 and S1P3. Knockdown studies suggest that S1P2 and S1P3 mediate this effect by activating myocardin-related transcription factor A (MRTF-A) and yes-associated protein (YAP), respectively. Michigan Cancer Foundation 7 cells stably overexpressing S1P2 or S1P3 exhibit a more invasive phenotype, when compared to control cells. Taken together, our findings suggest that S1P produced by SPHK1 induces SNAI2 expression via S1P2-YAP and S1P3-MRTF-A pathways, leading to enhanced cell invasion. Cumulatively, this study reveals a novel mechanism by which S1P activates parallel pathways that regulate the expression of SNAI2, a master regulator of EMT, and provides new insights into druggable therapeutic targets that may limit cancer metastasis. Wang, W., Hind, T., Lam, B. W. S., Herr, D. R. Sphingosine 1-phosphate signaling induces SNAI2 expression to promote cell invasion in breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/physiology , Lysophospholipids/physiology , Neoplasm Invasiveness/pathology , Neoplasm Proteins/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/physiology , Snail Family Transcription Factors/biosynthesis , Sphingosine/analogs & derivatives , Adaptor Proteins, Signal Transducing/physiology , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Lysophospholipids/pharmacology , MCF-7 Cells , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , RNA Interference , RNA Stability , RNA, Small Interfering/pharmacology , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/physiology , Sphingosine/pharmacology , Sphingosine/physiology , Sphingosine-1-Phosphate Receptors/physiology , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors/physiology , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL