Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 247
Filter
1.
Science ; 381(6663): 1182-1189, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37708259

ABSTRACT

Lysosomes critically rely on bis(monoacylglycero)phosphate (BMP) to stimulate lipid catabolism, cholesterol homeostasis, and lysosomal function. Alterations in BMP levels in monogenic and complex neurodegeneration suggest an essential function in human health. However, the site and mechanism responsible for BMP synthesis have been subject to debate for decades. Here, we report that the Batten disease gene product CLN5 is the elusive BMP synthase (BMPS). BMPS-deficient cells exhibited a massive accumulation of the BMP synthesis precursor lysophosphatidylglycerol (LPG), depletion of BMP species, and dysfunctional lipid metabolism. Mechanistically, we found that BMPS mediated synthesis through an energy-independent base exchange reaction between two LPG molecules with increased activity on BMP-laden vesicles. Our study elucidates BMP biosynthesis and reveals an anabolic function of late endosomes/lysosomes.


Subject(s)
Lysophospholipids , Lysosomal Membrane Proteins , Monoglycerides , Neuronal Ceroid-Lipofuscinoses , Humans , Lysosomal Membrane Proteins/genetics , Lysosomes , Monoglycerides/biosynthesis , Neuronal Ceroid-Lipofuscinoses/genetics , Nitric Oxide Synthase , Lysophospholipids/biosynthesis
2.
J Biol Chem ; 299(9): 105126, 2023 09.
Article in English | MEDLINE | ID: mdl-37543362

ABSTRACT

Oxidative stress triggered by aging, radiation, or inflammation impairs ovarian function by inducing granulosa cell (GC) apoptosis. However, the mechanism inducing GC apoptosis has not been characterized. Here, we found that ovarian GCs from aging patients showed increased oxidative stress, enhanced reactive oxygen species activity, and significantly decreased expression of the known antiapoptotic factor sphingosine-1-phosphate/sphingosine kinase 1 (SPHK1) in GCs. Interestingly, the expression of Krüppel-like factor 12 (KLF12) was significantly increased in the ovarian GCs of aging patients. Furthermore, we determined that KLF12 was significantly upregulated in hydrogen peroxide-treated GCs and a 3-nitropropionic acid-induced in vivo model of ovarian oxidative stress. This phenotype was further confirmed to result from inhibition of SPHK1 by KLF12. Interestingly, when endogenous KLF12 was knocked down, it rescued oxidative stress-induced apoptosis. Meanwhile, supplementation with SPHK1 partially reversed oxidative stress-induced apoptosis. However, this function was lost in SPHK1 with deletion of the binding region to the KLF12 promoter. SPHK1 reversed apoptosis caused by hydrogen peroxide-KLF12 overexpression, a result further confirmed in an in vitro ovarian culture model and an in vivo 3-nitropropionic acid-induced ovarian oxidative stress model. Overall, our study reveals that KLF12 is involved in regulating apoptosis induced by oxidative stress in aging ovarian GCs and that sphingosine-1-phosphate/SPHK1 can rescue GC apoptosis by interacting with KLF12 in negative feedback.


Subject(s)
Aging , Apoptosis , Granulosa Cells , Hydrogen Peroxide , Kruppel-Like Transcription Factors , Lysophospholipids , Phosphotransferases (Alcohol Group Acceptor) , Sphingosine , Female , Humans , Aging/metabolism , Feedback, Physiological , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Hydrogen Peroxide/pharmacology , In Vitro Techniques , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Lysophospholipids/biosynthesis , Lysophospholipids/metabolism , Organ Culture Techniques , Oxidative Stress/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Promoter Regions, Genetic , Sphingosine/biosynthesis , Sphingosine/metabolism , Reactive Oxygen Species/metabolism
3.
Int J Mol Sci ; 24(7)2023 Apr 01.
Article in English | MEDLINE | ID: mdl-37047587

ABSTRACT

Mast cells are responsible for IgE-dependent allergic responses, but they also produce various bioactive mediators and contribute to the pathogenesis of various cardiovascular diseases, including pulmonary hypertension (PH). The importance of lipid mediators in the pathogenesis of PH has become evident in recent years, as exemplified by prostaglandin I2, the most central therapeutic target in pulmonary arterial hypertension. New bioactive lipids other than eicosanoids have also been identified that are associated with the pathogenesis of PH. However, it remains largely unknown how mast cell-derived lipid mediators are involved in pulmonary vascular remodeling. Recently, it has been demonstrated that mast cells produce epoxidized n-3 fatty acid (n-3 epoxides) in a degranulation-independent manner, and that n-3 epoxides produced by mast cells regulate the abnormal activation of pulmonary fibroblasts and suppress the progression of pulmonary vascular remodeling. This review summarizes the role of mast cells and bioactive lipids in the pathogenesis of PH. In addition, we introduce the pathophysiological role and therapeutic potential of n-3 epoxides, a mast cell-derived novel lipid mediator, in the pulmonary vascular remodeling in PH. Further knowledge of mast cells and lipid mediators is expected to lead to the development of innovative therapies targeting pulmonary vascular remodeling.


Subject(s)
Airway Remodeling , Fatty Acids, Unsaturated , Hypertension, Pulmonary , Lysophospholipids , Mast Cells , Pulmonary Artery , Mast Cells/metabolism , Airway Remodeling/immunology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/pathology , Pulmonary Artery/immunology , Pulmonary Artery/pathology , Lysophospholipids/biosynthesis , Fatty Acids, Unsaturated/biosynthesis , Humans , Animals
4.
J Am Heart Assoc ; 10(14): e021261, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34240614

ABSTRACT

Background Most of the circulating sphingosine-1-phosphate (S1P) is bound to ApoM (apolipoprotein M) of high-density lipoprotein (HDL) and mediates many beneficial effects of HDL on the vasculature via G protein-coupled S1P receptors. HDL-bound S1P is decreased in atherosclerosis, myocardial infarction, and diabetes mellitus. In addition to being the target, the endothelium is a source of S1P, which is transported outside of the cells by Spinster-2, contributing to circulating S1P as well as to local signaling. Mice lacking endothelial S1P receptor 1 are hypertensive, suggesting a vasculoprotective role of S1P signaling. This study investigates the role of endothelial-derived S1P and ApoM-bound S1P in regulating vascular tone and blood pressure. Methods and Results ApoM knockout (ApoM KO) mice and mice lacking endothelial Spinster-2 (ECKO-Spns2) were infused with angiotensin II for 28 days. Blood pressure, measured by telemetry and tail-cuff, was significantly increased in both ECKO-Spns2 and ApoM KO versus control mice, at baseline and following angiotensin II. Notably, ECKO-Spns2 presented an impaired vasodilation to flow and blood pressure dipping, which is clinically associated with increased risk for cardiovascular events. In hypertension, both groups presented reduced flow-mediated vasodilation and some degree of impairment in endothelial NO production, which was more evident in ECKO-Spns2. Increased hypertension in ECKO-Spns2 and ApoM KO mice correlated with worsened cardiac hypertrophy versus controls. Conclusions Our study identifies an important role for Spinster-2 and ApoM-HDL in blood pressure homeostasis via S1P-NO signaling and dissects the pathophysiological impact of endothelial-derived S1P and ApoM of HDL-bound S1P in hypertension and cardiac hypertrophy.


Subject(s)
Anion Transport Proteins/genetics , Apolipoproteins M/genetics , Endothelium, Vascular/physiopathology , Gene Expression Regulation , Hypertension/genetics , Lysophospholipids/genetics , Sphingosine/analogs & derivatives , Vascular Stiffness/physiology , Animals , Anion Transport Proteins/biosynthesis , Apolipoproteins M/biosynthesis , Disease Models, Animal , Endothelium, Vascular/metabolism , Hypertension/metabolism , Hypertension/physiopathology , Lysophospholipids/biosynthesis , Male , Mice , Mice, Knockout , RNA/genetics , Sphingosine/biosynthesis , Sphingosine/genetics
5.
Cardiovasc Drugs Ther ; 35(1): 167-183, 2021 02.
Article in English | MEDLINE | ID: mdl-32519066

ABSTRACT

BACKGROUND: Despite advances in the development of lipid-lowering therapies, clinical trials have shown that a significant residual risk of cardiovascular disease persists. Specifically, new drugs are needed for non-responding or statin-intolerant subjects or patients considered at very high risk for cardiovascular events even though are already on treatment with the best standard of care. RESULTS AND CONCLUSIONS: Besides, genetic and epidemiological studies and Mendelian randomization analyses have strengthened the linear correlation between the concentration of low-density lipoprotein cholesterol (LDL-C) and the incidence of cardiovascular events and highlighted various novel therapeutic targets. This review describes the novel strategies to reduce the levels of LDL-C, non-HDL-C, triglyceride, apolipoprotein B, and Lp(a), focusing on those developed using biotechnology-based strategies.


Subject(s)
Dyslipidemias/drug therapy , Hypolipidemic Agents/therapeutic use , Antibodies, Monoclonal, Humanized , Apolipoproteins B/drug effects , Cholesterol, LDL/drug effects , Clinical Trials as Topic , Genetic Therapy/methods , Humans , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/adverse effects , Lysophospholipids/biosynthesis , Oligonucleotides, Antisense/therapeutic use , RNA, Small Interfering/therapeutic use , Triglycerides/biosynthesis
6.
Environ Microbiol ; 22(12): 5300-5308, 2020 12.
Article in English | MEDLINE | ID: mdl-32929857

ABSTRACT

Acinetobacter baumannii is an opportunistic pathogen, which has become a rising threat in healthcare facilities worldwide due to increasing antibiotic resistances and optimal adaptation to clinical environments and the human host. We reported in a former publication on the identification of three phopholipases of the phospholipase D (PLD) superfamily in A. baumannii ATCC 19606T acting in concerted manner as virulence factors in Galleria mellonella infection and lung epithelial cell invasion. This study focussed on the function of the three PLDs. A Δpld1-3 mutant was defect in biosynthesis of the phospholipids cardiolipin (CL) and monolysocardiolipin (MLCL), whereas the deletion of pld2 and pld3 abolished the production of MLCL. Complementation of the Δpld1-3 mutant with pld1 restored CL biosynthesis demonstrating that the PLD1 is implicated in CL biosynthesis. Complementation of the Δpld1-3 mutant with either pld2 or pld3 restored MLCL and CL production leading to the conclusion that PLD2 and PLD3 are implicated in CL and MLCL production. Mutant studies revealed that two catalytic motifs are essential for the PLD3-mediated biosynthesis of CL and MLCL. The Δpld1-3 mutant exhibited a decreased colistin and polymyxin B resistance indicating a role of CL in cationic antimicrobial peptides (CAMPs) resistance.


Subject(s)
Acinetobacter baumannii/metabolism , Antimicrobial Cationic Peptides/metabolism , Cardiolipins/biosynthesis , Drug Resistance, Bacterial , Phospholipase D/metabolism , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Lysophospholipids/biosynthesis , Mutation , Phospholipase D/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
7.
J Cell Mol Med ; 24(18): 10290-10301, 2020 09.
Article in English | MEDLINE | ID: mdl-32803879

ABSTRACT

There are many different types of cardiovascular diseases, which impose a huge economic burden due to their extremely high mortality rates, so it is necessary to explore the underlying mechanisms to achieve better supportive and curative care outcomes. Sphingosine 1-phosphate (S1P) is a bioactive lipid mediator with paracrine and autocrine activities that acts through its cell surface S1P receptors (S1PRs) and intracellular signals. In the circulatory system, S1P is indispensable for both normal and disease conditions; however, there are very different views on its diverse roles, and its specific relevance to cardiovascular pathogenesis remains elusive. Here, we review the synthesis, release and functions of S1P, specifically detail the roles of S1P and S1PRs in some common cardiovascular diseases, and then address several controversial points, finally, we focus on the development of S1P-based therapeutic approaches in cardiovascular diseases, such as the selective S1PR1 modulator amiselimod (MT-1303) and the non-selective S1PR1 and S1PR3 agonist fingolimod, which may provide valuable insights into potential therapeutic strategies for cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/metabolism , Lysophospholipids/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine/analogs & derivatives , Animals , Humans , Lysophospholipids/biosynthesis , Models, Biological , Signal Transduction , Sphingosine/biosynthesis , Sphingosine/metabolism
8.
Essays Biochem ; 64(3): 579-589, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32579188

ABSTRACT

Sphingolipids are a class of complex lipids containing a backbone of sphingoid bases, namely the organic aliphatic amino alcohol sphingosine (Sph), that are essential constituents of eukaryotic cells. They were first described as major components of cell membrane architecture, but it is now well established that some sphingolipids are bioactive and can regulate key biological functions. These include cell growth and survival, cell differentiation, angiogenesis, autophagy, cell migration, or organogenesis. Furthermore, some bioactive sphingolipids are implicated in pathological processes including inflammation-associated illnesses such as atherosclerosis, rheumatoid arthritis, inflammatory bowel disease (namely Crohn's disease and ulcerative colitis), type II diabetes, obesity, and cancer. A major sphingolipid metabolite is ceramide, which is the core of sphingolipid metabolism and can act as second messenger, especially when it is produced at the plasma membrane of cells. Ceramides promote cell cycle arrest and apoptosis. However, ceramide 1-phosphate (C1P), the product of ceramide kinase (CerK), and Sph 1-phosphate (S1P), which is generated by the action of Sph kinases (SphK), stimulate cell proliferation and inhibit apoptosis. Recently, C1P has been implicated in the spontaneous migration of cells from some types of cancer, and can enhance cell migration/invasion of malignant cells through interaction with a Gi protein-coupled receptor. In addition, CerK and SphK are implicated in inflammatory responses, some of which are associated with cancer progression and metastasis. Hence, targeting these sphingolipid kinases to inhibit C1P or S1P production, or blockade of their receptors might contribute to the development of novel therapeutic strategies to reduce metabolic alterations and disease.


Subject(s)
Cell Movement , Ceramides/biosynthesis , Lysophospholipids/biosynthesis , Neoplasms/metabolism , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , Animals , Humans , Inflammation/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction , Sphingosine/biosynthesis
9.
Biochem Biophys Res Commun ; 528(3): 473-477, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32507278

ABSTRACT

Cyanobacteria and chloroplasts are believed to share a common ancestor, but synthetic pathways for membrane lipids are different. Lyso-phosphatidic acid (lyso-PA) is the precursor for the synthesis of all membrane lipids and synthesized by an acyl-ACP dependent glycerol-3-phosphate acyltransferase (GPAT) in chloroplasts. In cyanobacteria, GPAT genes are not found and, instead, genes coding for enzymes in the acyl-phosphate dependent lyso-PA synthetic pathway (plsX and plsY) are conserved. We report that the PlsX/Y dependent lyso-PA synthetic pathway is essential in cyanobacteria, but can be replaced by acyl-ACP dependent GPAT from Escherichia coli (plsB) and Arabidopsis thaliana (ATS1). Cyanobacteria thus display the capacity to accept enzymes from other organisms to synthesize essential components. This ability may have enabled them to evolve into current chloroplasts from their ancestral origins.


Subject(s)
Chloroplasts/metabolism , Membrane Lipids/biosynthesis , Synechocystis/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Biosynthetic Pathways , Chloroplasts/genetics , Endophytes/genetics , Endophytes/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Evolution, Molecular , Gene Knockout Techniques , Genes, Bacterial , Genes, Essential , Glycerol-3-Phosphate O-Acyltransferase/genetics , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Lysophospholipids/biosynthesis , Symbiosis , Synechocystis/genetics
10.
J Lipid Res ; 61(7): 1075-1086, 2020 07.
Article in English | MEDLINE | ID: mdl-32430316

ABSTRACT

The glycerol phosphate pathway produces more than 90% of the liver triacylglycerol (TAG). LysoPA, an intermediate in this pathway, is produced by glycerol-3-phosphate acyltransferase. Glycerophosphodiester phosphodiesterase domain containing 3 (GDPD3), whose gene was recently cloned, contains lysophospholipase D activity, which produces LysoPA from lysophospholipids. Whether human GDPD3 plays a role in hepatic TAG homeostasis is unknown. We hypothesized that human GDPD3 increases LysoPA production and availability in the glycerol phosphate pathway, promoting TAG biosynthesis. To test our hypothesis, we infected C57BL/6J mice with adeno-associated virus encoding a hepatocyte-specific albumin promoter that drives GFP (control) or FLAG-tagged human GDPD3 overexpression and fed the mice chow or a Western diet to induce hepatosteatosis. Hepatic human GDPD3 overexpression induced LysoPA production and increased FA uptake and incorporation into TAG in mouse hepatocytes and livers, ultimately exacerbating Western diet-induced liver steatosis. Our results also showed that individuals with hepatic steatosis have increased GDPD3 mRNA levels compared with individuals without steatosis. Collectively, these findings indicate that upregulation of GDPD3 expression may play a key role in hepatic TAG accumulation and may represent a molecular target for managing hepatic steatosis.


Subject(s)
Fatty Acids/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Liver/metabolism , Lysophospholipids/biosynthesis , Phosphoric Diester Hydrolases/genetics , Animals , Biological Transport/genetics , Gene Expression , Humans , Mice
11.
PLoS One ; 15(4): e0226050, 2020.
Article in English | MEDLINE | ID: mdl-32240164

ABSTRACT

Autotaxin (ATX) is a secreted lysophospholipase D catalyzing the extracellular production of lysophosphatidic acid (LPA), a growth factor-like signaling lysophospholipid. ATX and LPA signaling have been incriminated in the pathogenesis of different chronic inflammatory diseases and various types of cancer. In this report, deregulated ATX and LPA levels were detected in the spinal cord and plasma of mice during the development of experimental autoimmune encephalomyelitis (EAE). Among the different sources of ATX expression in the inflamed spinal cord, F4/80+ CD11b+ cells, mostly activated macrophages and microglia, were found to express ATX, further suggesting an autocrine role for ATX/LPA in their activation, an EAE hallmark. Accordingly, ATX genetic deletion from CD11b+ cells attenuated the severity of EAE, thus proposing a pathogenic role for the ATX/LPA axis in neuroinflammatory disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Lysophospholipids/genetics , Multiple Sclerosis/genetics , Phosphoric Diester Hydrolases/genetics , Animals , CD11b Antigen/genetics , Central Nervous System/metabolism , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gene Deletion , Gene Expression/genetics , Humans , Lysophospholipids/biosynthesis , Macrophages/metabolism , Macrophages/pathology , Mice , Microglia/metabolism , Microglia/pathology , Multiple Sclerosis/blood , Multiple Sclerosis/physiopathology , Signal Transduction/genetics , Spinal Cord/metabolism , Spinal Cord/physiopathology
12.
CNS Neurosci Ther ; 26(5): 538-548, 2020 05.
Article in English | MEDLINE | ID: mdl-31814336

ABSTRACT

AIMS: Sphingosine kinase 1 (Sphk1) and the signaling molecule sphingosine-1-phosphate (S1P) are known to be key regulators of a variety of important biological processes, such as neovascularization. Nitric oxide (NO) is also known to play a role in vasoactive properties, whether Sphk1/S1P signaling is able to alter angiogenesis in the context of cerebral ischemia-reperfusion injury (IRI), and whether such activity is linked with NO production, however, remains uncertain. METHODS: We used immunofluorescence to detect the expression of Sphk1 and NOS in cerebral epithelial cells (EC) after IR or oxygen-glucose deprivation (OGDR). Western blotting was used to detect the Sphk1 and NOS protein levels in brain tissues or HBMECs. Adenovirus transfection was used to inhibit Sphk1 and NOS. An NO kit was used to detect NO contents in brain tissues and epithelial cells. Tube formation assays were conducted to measure angiogenesis. RESULTS: We determined that EC used in a model of cerebral IRI expressed Sphk1, and that inhibiting this expression led to decreased expression of two isoforms of NO synthase (eNOS and iNOS), as well as to decrease neovascularization density and NO production following injury. In HBMECs, knocking down Sphk1 markedly reduced NO production owing to reduced eNOS activity, and inhibiting eNOS directly similarly decreased NO production in a manner which could be reversed via exogenously treating cells with S1P. We further found that knocking down Sphk1 reduced HBMEC eNOS expression, in addition to decreasing the adhesion, migration, and tube formation abilities of these cells under OGDR conditions. CONCLUSIONS: Based on these results, we therefore postulate that Sphk1/S1P signaling is able to mediate angiogenesis following cerebral IRI via the regulation of eNOS activity and NO production. As such, targeting these pathways may potentially represent a novel means of improving patient prognosis in those suffering from cerebral IRI.


Subject(s)
Brain Ischemia/metabolism , Lysophospholipids/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Reperfusion Injury/metabolism , Sphingosine/analogs & derivatives , Animals , Brain Ischemia/pathology , Cells, Cultured , Humans , Male , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Rats , Rats, Wistar , Reperfusion Injury/pathology , Sphingosine/biosynthesis
13.
Handb Exp Pharmacol ; 259: 49-76, 2020.
Article in English | MEDLINE | ID: mdl-29460151

ABSTRACT

There is substantial evidence that the enzymes, sphingosine kinase 1 and 2, which catalyse the formation of the bioactive lipid sphingosine 1-phosphate, are involved in pathophysiological processes. In this chapter, we appraise the evidence that both enzymes are druggable and describe how isoform-specific inhibitors can be developed based on the plasticity of the sphingosine-binding site. This is contextualised with the effect of sphingosine kinase inhibitors in cancer, pulmonary hypertension, neurodegeneration, inflammation and sickling.


Subject(s)
Enzyme Inhibitors/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Anemia, Sickle Cell , Binding Sites , Humans , Hypertension, Pulmonary , Inflammation , Lysophospholipids/biosynthesis , Neoplasms , Neurodegenerative Diseases , Sphingosine/analogs & derivatives , Sphingosine/biosynthesis
14.
J Bone Miner Res ; 34(12): 2264-2276, 2019 12.
Article in English | MEDLINE | ID: mdl-31373726

ABSTRACT

Spondyloarthritis (SpA) is a common rheumatic disease characterized by enthesis inflammation (enthesitis) and ectopic ossification (enthesophytes). The current pathogenesis model suggests that inflammation and mechanical stress are both strongly involved in SpA pathophysiology. We have previously observed that the levels of sphingosine 1-phosphate (S1P), a bone anabolic molecule, were particularly high in SpA patients' serum compared to healthy donors. Therefore, we wondered how this deregulation was related to SpA molecular mechanisms. Mouse primary osteoblasts, chondrocytes, and tenocytes were used as cell culture models. The sphingosine kinase 1 (Sphk1) gene expression and S1P secretion were significantly enhanced by cyclic stretch in osteoblasts and chondrocytes. Further, TNF-α and IL-17, cytokines implicated in enthesitis, increased Sphk1 mRNA in chondrocytes in an additive manner when combined to stretch. The immunochemistry on mouse ankles showed that sphingosine kinase 1 (SK1) was localized in some chondrocytes; the addition of a pro-inflammatory cocktail augmented Sphk1 expression in cultured ankles. Subsequently, fingolimod was used to block S1P metabolism in cell cultures. It inhibited S1P receptors (S1PRs) signaling and SK1 and SK2 activity in both osteoblasts and chondrocytes. Fingolimod also reduced S1PR-induced activation by SpA patients' synovial fluid (SF), demonstrating that the stimulation of chondrocytes by SFs from SpA patients involves S1P. In addition, when the osteogenic culture medium was supplemented with fingolimod, alkaline phosphatase activity, matrix mineralization, and bone formation markers were significantly reduced in osteoblasts and hypertrophic chondrocytes. Osteogenic differentiation was accompanied by an increase in S1prs mRNA, especially S1P1/3 , but their contribution to S1P-impact on mineralization seemed limited. Our results suggest that S1P might be overproduced in SpA enthesis in response to cytokines and mechanical stress, most likely by chondrocytes. Moreover, S1P could locally favor the abnormal ossification of the enthesis; therefore, blocking the S1P metabolic pathway could be a potential therapeutic approach for the treatment of SpA. © 2019 American Society for Bone and Mineral Research.


Subject(s)
Cytokines/pharmacology , Lysophospholipids/biosynthesis , Osteogenesis , Sphingosine/analogs & derivatives , Spondylarthritis/pathology , Spondylarthritis/physiopathology , Stress, Mechanical , Adolescent , Adult , Aged , Animals , Calcification, Physiologic/drug effects , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Female , Fingolimod Hydrochloride/pharmacology , Humans , Male , Metabolic Networks and Pathways/drug effects , Mice , Middle Aged , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/biosynthesis , Synovial Fluid/metabolism , Tenocytes/drug effects , Tenocytes/metabolism , Up-Regulation/drug effects , Young Adult
15.
Int J Mol Sci ; 20(15)2019 Jul 26.
Article in English | MEDLINE | ID: mdl-31357484

ABSTRACT

Sphingosine 1-phosphate (S1P) is a potent lipid mediator that modulates inflammation and angiogenesis. In this study, we investigated the possible involvement of S1P in the pathology of light-induced retinal degeneration in vivo and in vitro. The intracellular S1P and sphingosine kinase (SphK) activity in a photoreceptor cell line (661W cells) was significantly increased by exposure to light. The enhancement of SphK1 expression was dependent on illumination, and all-trans-retinal significantly promoted SphK1 expression. S1P treatment reduced protein kinase B (Akt) phosphorylation and increased the protein expression of cleaved caspase-3, and induced photoreceptor cell apoptosis. In vivo, light exposure enhanced the expression of SphK1 in the outer segments of photoreceptors. Intravitreal injection of a SphK inhibitor significantly suppressed the thinning of the outer nuclear layer and ameliorated the attenuation of the amplitudes of a-waves and b-waves of electroretinograms during light-induced retinal degeneration. These findings imply that light exposure induces the synthesis of S1P in photoreceptors by upregulating SphK1, which is facilitated by all-trans-retinal, causing retinal degeneration. Inhibition of this enhancement may be a therapeutic target of outer retinal degeneration, including age-related macular degeneration.


Subject(s)
Light , Lysophospholipids/biosynthesis , Photoreceptor Cells/metabolism , Photoreceptor Cells/radiation effects , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Sphingosine/analogs & derivatives , Stress, Physiological/radiation effects , Animals , Apoptosis , Cell Line , Disease Models, Animal , Disease Susceptibility , Electroretinography , Humans , Light/adverse effects , Macular Degeneration/etiology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mice , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Photoreceptor Cells/pathology , Retina/metabolism , Retina/pathology , Retina/radiation effects , Retinal Degeneration/diagnostic imaging , Retinal Degeneration/pathology , Sphingosine/biosynthesis , Tomography, Optical Coherence
16.
Cell Cycle ; 18(14): 1646-1659, 2019 07.
Article in English | MEDLINE | ID: mdl-31203714

ABSTRACT

Mitosis has been traditionally considered a metabolically inactive phase. We have previously shown, however, that extensive alterations in lipids occur as the cells traverse mitosis, including increased de novo fatty acid (FA) and phosphatidylcholine (PtdCho) synthesis and decreased lysophospholipid content. Given the diverse structural and functional properties of these lipids, we sought to study their metabolic fate and their importance for cell cycle completion. Here we show that FA and PtdCho synthesized at the mitotic exit are destined to the nuclear envelope. Importantly, FA and PtdCho synthesis, but not the decrease in lysophospholipid content, are necessary for cell cycle completion beyond G2/M. Moreover, the presence of alternative pathways for PtdCho synthesis renders the cells less sensitive to its inhibition than to the impairment of FA synthesis. FA synthesis, thus, represents a cell cycle-related metabolic vulnerability that could be exploited for combined chemotherapy. We explored the combination of fatty acid synthase (FASN) inhibition with agents that act at different phases of the cell cycle. Our results show that the effect of FASN inhibition may be enhanced under some drug combinations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acids/biosynthesis , G2 Phase Cell Cycle Checkpoints/drug effects , Lipogenesis/drug effects , Mitosis/drug effects , Nuclear Envelope/metabolism , Phosphatidylcholines/biosynthesis , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Endoplasmic Reticulum/metabolism , Etoposide/pharmacology , Fatty Acid Synthases/metabolism , HeLa Cells , Humans , Lipogenesis/physiology , Lysophospholipids/biosynthesis , Lysophospholipids/chemistry , Mitosis/physiology , Nuclear Envelope/drug effects , Nuclear Envelope/enzymology
17.
Prostaglandins Other Lipid Mediat ; 143: 106335, 2019 08.
Article in English | MEDLINE | ID: mdl-31054330

ABSTRACT

Lysophosphatidic acid (LPA) is a simple phospholipid that exerts pleiotropic effects on numerous cell types by activating its family of cognate G protein-coupled receptors (GPCRs) and participates in many biological processes, including organismal development, wound healing, and carcinogenesis. Bone cells, such as bone marrow mesenchymal stromal (stem) cells (BMSCs), osteoblasts, osteocytes and osteoclasts play essential roles in bone homeostasis and repair. Previous studies have identified the presence of specific LPA receptors in these bone cells. In recent years, an increasing number of cellular effects of LPA, such as the induction of cell proliferation, survival, migration, differentiation and cytokine secretion, have been found in different bone cells. Moreover, some biomaterials containing LPA have shown the ability to enhance osteogenesis. This review will focus on findings associated with LPA functions in these bone cells and present current studies related to the application of LPA in bone regenerative medicine. Further understanding this information will help us develop better strategies for bone healing.


Subject(s)
Bone Regeneration , Bone and Bones/cytology , Bone and Bones/physiology , Lysophospholipids/metabolism , Animals , Bone Regeneration/drug effects , Bone and Bones/drug effects , Humans , Lysophospholipids/biosynthesis , Lysophospholipids/pharmacology , Receptors, Lysophosphatidic Acid/metabolism
18.
IUBMB Life ; 71(9): 1284-1292, 2019 09.
Article in English | MEDLINE | ID: mdl-30889317

ABSTRACT

This study was designed to explore whether exosomal sphingosine 1-phosphate (S1P) from mesenchymal stem cells (MSCs) regulate the Treg/Th17 balance in aplastic anemia (AA) patients and to validate the underlying mechanism. To address this, exosomes from human bone marrow MSCs (MSCs-Exos) were co-cultured with CD4+ T cells from AA patients (AA CD4+ T cells), which were transfected with si-S1PR1, si-S1PR3, or not. The proportion of Th17 and Treg was evaluated by flow cytometry. The levels of Th17-associated interleukin-17 (IL-17), Treg-associated IL-10, and transforming growth factor-ß were determined by ELISA. S1P content in MSCs-Exos isolated from control, si-SphK1, or si-SphK2 transfected MSCs was examined by LC-MS/MS. Hematoxylin and eosin staining of bone marrow tissues was performed to evaluate the effect of MSCs-Exos in AA mice. Our results showed that MSCs-Exos reversed the increased Th17/Treg in AA through SphK1-mediated exosomal S1P enrichment. Furthermore, the promotion of Treg differentiation by exosomal S1P from MSCs was mediated through the receptor S1PR1 expressed on CD4+ T cells. Further in vivo experiments showed that MSCs-Exos reversed the increased Th17/Treg and alleviated AA progression in AA mice. In summary, SphK1-mediated enrichment of exosomal S1P secreted by MSCs reversed the increased Treg/Th17 ratio via the receptor S1PR1 in AA patients. © 2019 IUBMB Life, 71(9):1284-1292, 2019.


Subject(s)
Anemia, Aplastic/immunology , Lysophospholipids/immunology , Sphingosine/analogs & derivatives , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Anemia, Aplastic/genetics , Anemia, Aplastic/pathology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Chromatography, Liquid , Coculture Techniques , Exosomes/metabolism , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lysophospholipids/biosynthesis , Lysophospholipids/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Sphingosine/biosynthesis , Sphingosine/immunology , Sphingosine/metabolism , Tandem Mass Spectrometry , Transforming Growth Factor beta/genetics
19.
Acta Derm Venereol ; 99(6): 594-601, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30834454

ABSTRACT

Sphingosine-1-phosphate (S1P) is a signalling sphingolipid metabolite that regulates important cell processes, including cell proliferation and apoptosis. Circulating S1P levels have been reported to be increased in patients with psoriasis relative to healthy patients. The aim of this study was to examine the potency of S1P inhibition using an imiquimod-induced psoriasis mouse model. Both topical ceramidase and sphingosine kinase 1/2 inhibition, which blocks S1P generation, alleviated imiquimod-induced skin lesions and reduced the serum interleukin 17-A levels induced by application of imiquimod. These treatments also normalized skin mRNA levels of genes associated with inflammation and keratinocyte differentiation. Inhibition of sphingosine kinase 2, but not sphingosine kinase 1, diminished levels of suppressor of cytokine signalling 1 and blocked T helper type 17 differentiation of naïve CD4+ T cells; imiquimod-induced psoriasis-like skin symptoms were also ameliorated. These results indicate the distinct effects of sphingosine kinase 1 and sphingosine kinase 2 inhibition on T helper type 17 generation and suggest molecules that inhibit S1P formation, including ceramidase and sphingosine kinase 2 inhibitors, as novel therapeutic candidates for psoriasis.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Enzyme Inhibitors/pharmacology , Lysophospholipids/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Psoriasis/drug therapy , Sphingosine/analogs & derivatives , Administration, Topical , Animals , Cell Differentiation/drug effects , Ceramidases/antagonists & inhibitors , Disease Models, Animal , Gene Expression/drug effects , Imiquimod , Immunity/drug effects , Inflammation/genetics , Interleukin-17/blood , Male , Mice , Psoriasis/chemically induced , Psoriasis/pathology , Quinolones/pharmacology , RNA, Messenger/metabolism , Sphingosine/biosynthesis , Suppressor of Cytokine Signaling 1 Protein , Th17 Cells
20.
Mol Oncol ; 13(2): 185-201, 2019 02.
Article in English | MEDLINE | ID: mdl-30353652

ABSTRACT

The peritoneal fluid of ovarian carcinoma patients promotes cancer cell invasion and metastatic spread with lysophosphatidic acid (LPA) as a potentially crucial mediator. However, the origin of LPA in ascites and the clinical relevance of individual LPA species have not been addressed. Here, we show that the levels of multiple acyl-LPA species are strongly elevated in ascites versus plasma and are associated with short relapse-free survival. Data derived from transcriptome and secretome analyses of primary ascite-derived cells indicate that (a) the major route of LPA synthesis is the consecutive action of a secretory phospholipase A2 (PLA2 ) and autotaxin, (b) that the components of this pathway are coordinately upregulated in ascites, and (c) that CD163+CD206+ tumor-associated macrophages play an essential role as main producers of PLA2 G7 and autotaxin. The latter conclusion is consistent with mass spectrometry-based metabolomic analyses of conditioned medium from ascites cells, which showed that tumor-associated macrophages, but not tumor cells, are able to produce 20:4 acyl-LPA in lipid-free medium. Furthermore, our transcriptomic data revealed that LPA receptor (LPAR) genes are expressed in a clearly cell type-selective manner: While tumor cells express predominantly LPAR1-3, macrophages and T cells also express LPAR5 and LPAR6 at high levels, pointing to cell type-selective LPA signaling pathways. RNA profiling identified cytokines linked to cell motility and migration as the most conspicuous class of LPA-induced genes in macrophages, suggesting that LPA exerts protumorigenic properties at least in part via the tumor secretome.


Subject(s)
Lysophospholipids/biosynthesis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Signal Transduction , Tumor Microenvironment , Ascites/metabolism , Cell Line, Tumor , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Macrophages/metabolism , Macrophages/pathology , Metabolome , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/genetics , Receptors, Lysophosphatidic Acid/metabolism , Treatment Outcome , Tumor Microenvironment/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...