Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Nat Commun ; 14(1): 2404, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37100836

ABSTRACT

Antiplatelet medication is standard of care in acute myocardial infarction (AMI). However, it may have obscured beneficial properties of the activated platelet secretome. We identify platelets as major source of a sphingosine-1-phosphate (S1P) burst during AMI, and find its magnitude to favorably associate with cardiovascular mortality and infarct size in STEMI patients over 12 months. Experimentally, administration of supernatant from activated platelets reduces infarct size in murine AMI, which is blunted in platelets deficient for S1P export (Mfsd2b) or production (Sphk1) and in mice deficient for cardiomyocyte S1P receptor 1 (S1P1). Our study reveals an exploitable therapeutic window in antiplatelet therapy in AMI as the GPIIb/IIIa antagonist tirofiban preserves S1P release and cardioprotection, whereas the P2Y12 antagonist cangrelor does not. Here, we report that platelet-mediated intrinsic cardioprotection is an exciting therapeutic paradigm reaching beyond AMI, the benefits of which may need to be considered in all antiplatelet therapies.


Subject(s)
Blood Platelets , Myocardial Infarction , Humans , Mice , Animals , Myocardial Infarction/drug therapy , Sphingosine , Lysophospholipids/therapeutic use , Myocytes, Cardiac
2.
Nat Rev Gastroenterol Hepatol ; 19(6): 351-366, 2022 06.
Article in English | MEDLINE | ID: mdl-35165437

ABSTRACT

Immune cell trafficking is a critical element of the intestinal immune response, both in homeostasis and in pathological conditions associated with inflammatory bowel disease (IBD). This process involves adhesion molecules, chemoattractants and receptors expressed on immune cell surfaces, blood vessels and stromal intestinal tissue as well as signalling pathways, including those modulated by sphingosine 1-phosphate (S1P). The complex biological processes of leukocyte recruitment, activation, adhesion and migration have been targeted by various monoclonal antibodies (vedolizumab, etrolizumab, ontamalimab). Promising preclinical and clinical data with several oral S1P modulators suggest that inhibition of lymphocyte egress from the lymph nodes to the bloodstream might be a safe and efficacious alternative mechanism for reducing inflammation in immune-mediated disorders, including Crohn's disease and ulcerative colitis. Although various questions remain, including the potential positioning of S1P modulators in treatment algorithms and their long-term safety, this novel class of compounds holds great promise. This Review summarizes the critical mediators and mechanisms involved in immune cell trafficking in IBD and the available evidence for efficacy, safety and pharmacokinetics of S1P receptor modulators in IBD and other immune-mediated disorders. Further, it discusses potential future approaches to incorporate S1P modulators into the treatment of IBD.


Subject(s)
Colitis, Ulcerative , Inflammatory Bowel Diseases , Colitis, Ulcerative/drug therapy , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Lysophospholipids/metabolism , Lysophospholipids/therapeutic use , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine/therapeutic use
3.
BMJ Open Respir Res ; 8(1)2021 Dec.
Article in English | MEDLINE | ID: mdl-34969771

ABSTRACT

INTRODUCTION: Idiopathic pulmonary fibrosis (IPF) and non-IPF, progressive fibrotic interstitial lung diseases (PF-ILD), are associated with a progressive loss of lung function and a poor prognosis. Treatment with antifibrotic agents can slow, but not halt, disease progression, and treatment discontinuation because of adverse events is common. Fibrotic diseases such as these can be mediated by lysophosphatidic acid (LPA), which signals via six LPA receptors (LPA1-6). Signalling via LPA1 appears to be fundamental in the pathogenesis of fibrotic diseases. BMS-986278, a second-generation LPA1 antagonist, is currently in phase 2 development as a therapy for IPF and PF-ILD. METHODS AND ANALYSIS: This phase 2, randomised, double-blind, placebo-controlled, parallel-group, international trial will include adults with IPF or PF-ILD. The trial will consist of a 42-day screening period, a 26-week placebo-controlled treatment period, an optional 26-week active-treatment extension period, and a 28-day post-treatment follow-up. Patients in both the IPF (n=240) and PF-ILD (n=120) cohorts will be randomised 1:1:1 to receive 30 mg or 60 mg BMS-986278, or placebo, administered orally two times per day for 26 weeks in the placebo-controlled treatment period. The primary endpoint is rate of change in per cent predicted forced vital capacity from baseline to week 26 in the IPF cohort. ETHICS AND DISSEMINATION: This study will be conducted in accordance with Good Clinical Practice guidelines, Declaration of Helsinki principles, and local ethical and legal requirements. Results will be reported in a peer-reviewed publication. TRIAL REGISTRATION NUMBER: NCT04308681.


Subject(s)
Idiopathic Pulmonary Fibrosis , Receptors, Lysophosphatidic Acid , Adult , Humans , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/drug therapy , Lysophospholipids/therapeutic use , Receptors, Lysophosphatidic Acid/therapeutic use , Vital Capacity
4.
JCI Insight ; 6(16)2021 08 23.
Article in English | MEDLINE | ID: mdl-34197342

ABSTRACT

Functional loss of myosin Vb (MYO5B) induces a variety of deficits in intestinal epithelial cell function and causes a congenital diarrheal disorder, microvillus inclusion disease (MVID). The impact of MYO5B loss on differentiated cell lineage choice has not been investigated. We quantified the populations of differentiated epithelial cells in tamoxifen-induced, epithelial cell-specific MYO5B-knockout (VilCreERT2 Myo5bfl/fl) mice utilizing digital image analysis. Consistent with our RNA-sequencing data, MYO5B loss induced a reduction in tuft cells in vivo and in organoid cultures. Paneth cells were significantly increased by MYO5B deficiency along with expansion of the progenitor cell zone. We further investigated the effect of lysophosphatidic acid (LPA) signaling on epithelial cell differentiation. Intraperitoneal LPA significantly increased tuft cell populations in both control and MYO5B-knockout mice. Transcripts for Wnt ligands were significantly downregulated by MYO5B loss in intestinal epithelial cells, whereas Notch signaling molecules were unchanged. Additionally, treatment with the Notch inhibitor dibenzazepine (DBZ) restored the populations of secretory cells, suggesting that the Notch pathway is maintained in MYO5B-deficient intestine. MYO5B loss likely impairs progenitor cell differentiation in the small intestine in vivo and in vitro, partially mediated by Wnt/Notch imbalance. Notch inhibition and/or LPA treatment may represent an effective therapeutic approach for treatment of MVID.


Subject(s)
Malabsorption Syndromes/genetics , Microvilli/pathology , Mucolipidoses/genetics , Myosin Type V/deficiency , Receptors, Notch/metabolism , Wnt Signaling Pathway/genetics , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Dibenzazepines/pharmacology , Disease Models, Animal , Enterocytes/drug effects , Enterocytes/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Jejunum/cytology , Jejunum/drug effects , Jejunum/pathology , Lysophospholipids/pharmacology , Lysophospholipids/therapeutic use , Malabsorption Syndromes/drug therapy , Malabsorption Syndromes/pathology , Mice , Mice, Knockout , Microvilli/genetics , Mucolipidoses/drug therapy , Mucolipidoses/pathology , Myosin Type V/genetics , Organoids , Primary Cell Culture , Receptors, Notch/antagonists & inhibitors , Stem Cells/physiology , Wnt Signaling Pathway/drug effects
5.
Thromb Haemost ; 121(3): 341-350, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33011963

ABSTRACT

The high mortality seen in sepsis is caused by a systemic hypotension in part owing to a drastic increase in vascular permeability accompanied by a loss of pericytes. As has been shown previously, pericyte retention in the perivascular niche during sepsis can enhance the integrity of the vasculature and promote survival via recruitment of adhesion proteins such as VE-cadherin and N-cadherin. Sphingosine-1-phosphate (S1P) represents a lipid mediator regulating the deposition of the crucial adhesion molecule VE-cadherin at sites of interendothelial adherens junctions and of N-cadherin at endothelial-pericyte adherens junctions. Furthermore, in septic patients, S1P plasma levels are decreased and correlate with mortality in an indirectly proportional way. In the present study, we investigated the potential of S1P to ameliorate a lipopolysaccharide-induced septic hypercirculation in mice. Here we establish S1P as an antagonist of pericyte loss, vascular hyperpermeability, and systemic hypotension, resulting in an increased survival in mice. During sepsis S1P preserved VE-cadherin and N-cadherin deposition, mediated by a reduction of Src and cadherin phosphorylation. At least in part, this effect is mediated by a reduction of globular actin and a subsequent increase in nuclear translocation of MRTF-A (myocardin-related transcription factor A). These findings indicate that S1P may counteract pericyte loss and microvessel disassembly during sepsis and additionally emphasize the importance of pericyte-endothelial interactions to stabilize the vasculature.


Subject(s)
Lysophospholipids/therapeutic use , Pericytes/drug effects , Sepsis/drug therapy , Sphingosine/analogs & derivatives , Trans-Activators/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/adverse effects , Mice, Inbred C57BL , Pericytes/metabolism , Pericytes/pathology , Sepsis/chemically induced , Sepsis/metabolism , Sepsis/pathology , Sphingosine/therapeutic use
6.
Exp Physiol ; 105(11): 1895-1906, 2020 11.
Article in English | MEDLINE | ID: mdl-32897592

ABSTRACT

NEW FINDINGS: What is the central question of the study? What are the consequences of reducing circulating sphingosine-1-phosphate (S1P) for muscle physiology in the murine model of Duchenne muscular dystrophy (DMD)? What is the main result and its importance? Reduction of the circulating S1P level in mdx mice aggravates the dystrophic phenotype, as seen by an increase in fibre atrophy, fibrosis and loss of specific force, suggesting that S1P signalling is a potential therapeutic target in DMD. Although further studies are needed, plasma S1P levels have the intriguing possibility of being used as a biomarker for disease severity, an important issue in DMD. ABSTRACT: Sphingosine-1-phosphate (S1P) is an important regulator of skeletal muscle properties. The dystrophin-deficient mdx mouse possesses low levels of S1P (∼50%) compared with wild type. Increased S1P availability was demonstrated to ameliorate the dystrophic phenotype in Drosophila and in mdx mice. Here, we analysed the effects produced by further reduction of S1P availability on the mass, force and regenerative capacity of dystrophic mdx soleus. Circulating S1P was neutralized by a specific anti-S1P antibody (S1P-Ab) known to lower the extracellular concentration of this signalling lipid. The S1P-Ab was administered intraperitoneally in adult mdx mice every 2 days for the duration of experiments. Soleus muscle properties were analysed 7 or 14 days after the first injection. The decreased availability of circulating S1P after the 14 day treatment reduced mdx soleus fibre cross-sectional area (-16%, P < 0.05), an effect that was associated with an increase in markers of proteolytic (MuRF1 and atrogin-1) and autophagic (p62 and LC3-II/LC3-I ratio) pathways. Moreover, an increase of fibrosis was also observed (+26%, P < 0.05). Notably, the treatment also caused a reduction of specific tetanic tension (-29%, P < 0.05). The mdx soleus regenerative capacity was only slightly influenced by reduced S1P. In conclusion, neutralization of circulating S1P reduces the mass and specific force and increases fibrosis of mdx soleus muscle, thus worsening the dystrophic phenotype. The results confirm that active, functional S1P signalling might counteract the progression of soleus mdx pathology and validate the pathway as a potential therapeutic target for muscular dystrophies.


Subject(s)
Dystrophin , Muscular Dystrophy, Duchenne , Animals , Disease Models, Animal , Dystrophin/metabolism , Lysophospholipids/metabolism , Lysophospholipids/therapeutic use , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Phenotype , Sphingosine/analogs & derivatives
7.
Gastroenterology ; 159(4): 1390-1405.e20, 2020 10.
Article in English | MEDLINE | ID: mdl-32534933

ABSTRACT

BACKGROUND & AIM: Myosin VB (MYO5B) is an essential trafficking protein for membrane recycling in gastrointestinal epithelial cells. The inactivating mutations of MYO5B cause the congenital diarrheal disease, microvillus inclusion disease (MVID). MYO5B deficiency in mice causes mislocalization of SGLT1 and NHE3, but retained apical function of CFTR, resulting in malabsorption and secretory diarrhea. Activation of lysophosphatidic acid (LPA) receptors can improve diarrhea, but the effect of LPA on MVID symptoms is unclear. We investigated whether LPA administration can reduce the epithelial deficits in MYO5B-knockout mice. METHODS: Studies were conducted with tamoxifen-induced, intestine-specific knockout of MYO5B (VilCreERT2;Myo5bflox/flox) and littermate controls. Mice were given LPA, an LPAR2 agonist (GRI977143), or vehicle for 4 days after a single injection of tamoxifen. Apical SGLT1 and CFTR activities were measured in Üssing chambers. Intestinal tissues were collected, and localization of membrane transporters was evaluated by immunofluorescence analysis in tissue sections and enteroids. RNA sequencing and enrichment analysis were performed with isolated jejunal epithelial cells. RESULTS: Daily administration of LPA reduced villus blunting, frequency of multivesicular bodies, and levels of cathepsins in intestinal tissues of MYO5B-knockout mice compared with vehicle administration. LPA partially restored the brush border height and the localization of SGLT1 and NHE3 in small intestine of MYO5B-knockout mice and enteroids. The SGLT1-dependent short-circuit current was increased and abnormal CFTR activities were decreased in jejunum from MYO5B-knockout mice given LPA compared with vehicle. CONCLUSIONS: LPA may regulate a MYO5B-independent trafficking mechanism and brush border maturation, and therefore be developed for treatment of MVID.


Subject(s)
Lysophospholipids/therapeutic use , Malabsorption Syndromes/drug therapy , Malabsorption Syndromes/pathology , Microvilli/pathology , Mucolipidoses/drug therapy , Mucolipidoses/pathology , Myosin Type V/deficiency , Sodium-Glucose Transporter 1/metabolism , Animals , Disease Models, Animal , Enterocytes/pathology , Malabsorption Syndromes/etiology , Mice , Mice, Knockout , Mucolipidoses/etiology
8.
Article in English | MEDLINE | ID: mdl-32179099

ABSTRACT

We previously detected a submicromolar concentration of lysophosphatidic acid (LPA) in human saliva. Here, we compare LPA concentrations in human gingival crevicular fluid (GCF) from patients with periodontitis and healthy controls, and examine how the local LPA levels are regulated enzymatically. The concentrations of LPA and its precursor lysophospholipids in GCF was measured by liquid chromatography-tandem mass spectrometry. The LPA-producing and LPA-degrading enzymatic activities were measured by quantifying the liberated choline and free fatty acid, respectively. The concentration of LPA in GCF of periodontitis patients was lower than that of healthy controls, due to higher soluble lysophospholipase activity toward LPA. LPA was found to prevent survival of Sa3, a human gingival epithelium-derived tumor cell line, activate Sa3 through Ca2+ mobilization, and release interleukin 6 from Sa3 in vitro. Furthermore, local injection of LPA into the gingiva attenuated ligature-induced experimental alveolar bone loss induced by oral bacteria inoculation in a rat model of periodontitis in vivo. A high concentration of LPA in human GCF is necessary to maintain normal gingival epithelial integrity and function, protecting the progression of periodontitis.


Subject(s)
Alveolar Bone Loss/metabolism , Gingival Crevicular Fluid/metabolism , Lysophospholipase/metabolism , Lysophospholipids/metabolism , Periodontitis/metabolism , Adult , Aged , Alveolar Bone Loss/etiology , Alveolar Bone Loss/prevention & control , Animals , Cells, Cultured , Female , Humans , Lysophospholipids/therapeutic use , Male , Middle Aged , Periodontitis/complications , Periodontitis/drug therapy , Rats , Rats, Wistar
9.
Pharmacol Ther ; 204: 107404, 2019 12.
Article in English | MEDLINE | ID: mdl-31472182

ABSTRACT

Lysophosphatidic acid (LPA) is a collective name for a set of bioactive lipid species. Via six widely distributed G protein-coupled receptors (GPCRs), LPA elicits a plethora of biological responses, contributing to inflammation, thrombosis and atherosclerosis. There have recently been considerable advances in GPCR signaling especially recognition of the extended role for GPCR transactivation of tyrosine and serine/threonine kinase growth factor receptors. This review covers LPA signaling pathways in the light of new information. The use of transgenic and gene knockout animals, gene manipulated cells, pharmacological LPA receptor agonists and antagonists have provided many insights into the biological significance of LPA and individual LPA receptors in the progression of atherosclerosis and vascular diseases. This review provides a comprehensive presentation of LPA with the highlight of the distinct role of its receptors in cell and animal models that relate to atherosclerosis and vascular diseases, and therefore provides new opportunities to reduce the burden of cardiovascular diseases. The recent drug development strategies that target LPA signaling pathways are also included in this review.


Subject(s)
Atherosclerosis/drug therapy , Lysophospholipids/pharmacology , Lysophospholipids/therapeutic use , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Vascular Diseases/drug therapy , Animals , Atherosclerosis/physiopathology , Humans , Receptors, Lysophosphatidic Acid/physiology , Signal Transduction/physiology , Vascular Diseases/physiopathology
10.
Oxid Med Cell Longev ; 2019: 9137654, 2019.
Article in English | MEDLINE | ID: mdl-31341538

ABSTRACT

Astragaloside IV is one of the main active ingredients isolated from Astragalus membranaceus. Here we confirmed its protective effect against cardiac ischemia-reperfusion (I/R) injury and aimed to investigate the potential molecular mechanisms involved. Pretreatment of ex vivo and in vivo I/R-induced rat models by astragaloside IV significantly prevented the ratio of myocardium infarct size, systolic and diastolic dysfunction, and the production of creatine kinase and lactate dehydrogenase. Metabolic analyses showed that I/R injury caused a notable reduction of succinate and elevation of lysophospholipids, indicating excessive reactive oxygen species (ROS) generation driven by succinate's rapid reoxidization and glycerophospholipid degradation. Molecular validation mechanistically revealed that astragaloside IV stimulated nuclear factor (erythroid-derived 2)-like 2 (Nrf2) released from Kelch-like ECH-associated protein 1 (Keap1) and translocated to the nucleus to combine with musculoaponeurotic fibrosarcoma (Maf) to initiate the transcription of antioxidative gene heme oxygenase-1 (HO-1), which performed a wide range of ROS scavenging processes against pathological oxidative stress in the hearts. As expected, increasing succinate and decreasing lysophospholipid levels were observed in the astragaloside IV-pretreated group compared with the I/R model group. These results suggested that astragaloside IV ameliorated myocardial I/R injury by modulating succinate and lysophospholipid metabolism and scavenging ROS via the Nrf2 signal pathway.


Subject(s)
Lysophospholipids/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Saponins/therapeutic use , Triterpenes/therapeutic use , Animals , Lysophospholipids/pharmacology , Male , Oxidative Stress , Rats , Reactive Oxygen Species , Saponins/pharmacology , Succinic Acid , Triterpenes/pharmacology
11.
Am J Physiol Cell Physiol ; 317(3): C502-C512, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31241988

ABSTRACT

Sarcopenia, the age-associated loss of skeletal muscle mass and function, is coupled with declines in physical functioning leading to subsequent higher rates of disability, frailty, morbidity, and mortality. Aging and obesity independently contribute to muscle atrophy that is assumed to be a result of the activation of mutual physiological pathways. Understanding mechanisms contributing to the induction of skeletal muscle atrophy with aging and obesity is important for determining targets that may have pivotal roles in muscle loss in these conditions. We find that aging and obesity equally induce an anabolic resistance to acute skeletal muscle contraction as observed with decreases in anabolic signaling activation after contraction. Furthermore, treatment with the sphingosine-1-phosphate analog FTY720 for 4 wk increased lean mass and strength, and the anabolic signaling response to contraction was improved in obese but not older animals. To determine the role of chronic inflammation and different fatty acids on anabolic resistance in skeletal muscle cells, we overexpressed IKKß with and without exposure to saturated fatty acid (SFA; palmitic acid), polyunsaturated fatty acid (eicosapentaenoic acid), and monounsaturated fatty acid (oleic acid). We found that IKKß overexpression increased inflammation markers in muscle cells, and this chronic inflammation exacerbated anabolic resistance in response to SFA. Pretreatment with FTY720 reversed the inflammatory effects of palmitic acid in the muscle cells. Taken together, these data demonstrate chronic inflammation can induce anabolic resistance, SFA aggravates these effects, and FTY720 can reverse this by decreasing ceramide accumulation in skeletal muscle.


Subject(s)
Aging/drug effects , Fingolimod Hydrochloride/therapeutic use , Muscle Contraction/drug effects , Obesity/drug therapy , Sphingosine 1 Phosphate Receptor Modulators/therapeutic use , Aging/metabolism , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Fingolimod Hydrochloride/pharmacology , Lysophospholipids/pharmacology , Lysophospholipids/therapeutic use , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Obesity/metabolism , Random Allocation , Sarcopenia/drug therapy , Sarcopenia/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine/therapeutic use , Sphingosine 1 Phosphate Receptor Modulators/pharmacology
12.
J Neuroinflammation ; 16(1): 82, 2019 Apr 11.
Article in English | MEDLINE | ID: mdl-30975169

ABSTRACT

BACKGROUND: Neuropathic pain is caused by sensory nerve injury, but effective treatments are currently lacking. Microglia are activated in the spinal dorsal horn after sensory nerve injury and contribute to neuropathic pain. Accordingly, molecules expressed by these cells are considered potential targets for therapeutic strategies. Our previous gene screening study using a mouse model of motor nerve injury showed that the G-protein-coupled receptor 34 gene (GPR34) is induced by nerve injury. Because GPR34 is now considered a microglia-enriched gene, we explored the possibility that it might be involved in microglial activation in the dorsal horn in a mouse model of neuropathic pain. METHODS: mRNA expression of GPR34 and pro-inflammatory molecules was determined by quantitative real-time PCR in wild-type and GPR34-deficient mice with L4 spinal nerve injury. In situ hybridization was used to identify GPR34 expression in microglia, and immunohistochemistry with the microglial marker Iba1 was performed to examine microglial numbers and morphology. Mechanical sensitivity was evaluated by the von Frey hair test. Liquid chromatography-tandem mass spectrometry quantified expression of the ligand for GPR34, lysophosphatidylserine (LysoPS), in the dorsal horn, and a GPR34 antagonist was intrathecally administrated to examine the effect of inhibiting LysoPS-GPR34 signaling on mechanical sensitivity. RESULTS: GPR34 was predominantly expressed by microglia in the dorsal horn after L4 nerve injury. There were no histological differences in microglial numbers or morphology between WT and GPR34-deficient mice. However, nerve injury-induced pro-inflammatory cytokine expression levels in microglia and pain behaviors were significantly attenuated in GPR34-deficient mice. Furthermore, the intrathecal administration of the GPR34 antagonist reduced neuropathic pain. CONCLUSIONS: Inhibition of GPR34-mediated signal by GPR34 gene deletion reduced nerve injury-induced neuropathic pain by suppressing pro-inflammatory responses of microglia without affecting their morphology. Therefore, the suppression of GPR34 activity may have therapeutic potential for alleviating neuropathic pain.


Subject(s)
Microglia/metabolism , Neuralgia/metabolism , Neuralgia/pathology , Receptors, Lysophospholipid/metabolism , Spinal Cord/pathology , Analysis of Variance , Animals , Calcium-Binding Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Interferon Regulatory Factors/metabolism , Lysophospholipids/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Neuralgia/drug therapy , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Pain Measurement , Pain Threshold/physiology , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Receptors, Lysophospholipid/antagonists & inhibitors , Receptors, Lysophospholipid/genetics , Time Factors
13.
FASEB J ; 33(4): 5181-5195, 2019 04.
Article in English | MEDLINE | ID: mdl-30629456

ABSTRACT

Because the association between sphingosine 1-phosphate (S1P)/apolipoprotein M (ApoM) and chronic kidney diseases has not been established, we investigated the involvement of S1P/ApoM in the phenotypes of IgA nephropathy in hyper-IgA (HIGA) mice. The overexpression of ApoM in adenoviral gene transfer ameliorated the phenotypes of IgA nephropathy in HIGA mice, whereas the knockdown of ApoM with siRNA caused deterioration. When ApoM-overexpressing HIGA mice were treated with VPC23019, an antagonist against S1P receptor 1 (S1P1) and 3 (S1P3), we observed that the protective effects of ApoM were reversed, whereas JTE013, an antagonist against S1P2, did not inhibit the effects. We also found that S1P bound to albumin accelerated the proliferation of MES13 cells and the fibrotic changes of HK2 cells, which were inhibited by JTE013, whereas S1P bound to ApoM suppressed these changes, which were inhibited by VPC23019. These results suggest that S1P bound to ApoM possesses properties protective against the phenotypes of IgA nephropathy through S1P1 and S1P3, whereas S1P bound to albumin exerts deteriorating effects through S1P2. ApoM may be useful as a therapeutic target to treat or retard the progression of IgA nephropathy.-Kurano, M., Tsuneyama, K., Morimoto, Y., Nishikawa, M., Yatomi, Y. Apolipoprotein M suppresses the phenotypes of IgA nephropathy in hyper-IgA mice.


Subject(s)
Apolipoproteins M/therapeutic use , Glomerulonephritis, IGA/blood , Glomerulonephritis, IGA/drug therapy , Immunoglobulin A/blood , Lysophospholipids/therapeutic use , Sphingosine/analogs & derivatives , Animals , Blotting, Western , Cell Line , Creatinine/blood , Creatinine/urine , Female , Glomerulonephritis, IGA/metabolism , Glomerulonephritis, IGA/urine , Immunoglobulin A/metabolism , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/therapeutic use , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Sphingosine-1-Phosphate Receptors/metabolism
14.
Nat Commun ; 9(1): 4030, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279412

ABSTRACT

Asthma accounts for 380,000 deaths a year. Carotid body denervation has been shown to have a profound effect on airway hyper-responsiveness in animal models but a mechanistic explanation is lacking. Here we demonstrate, using a rat model of asthma (OVA-sensitized), that carotid body activation during airborne allergic provocation is caused by systemic release of lysophosphatidic acid (LPA). Carotid body activation by LPA involves TRPV1 and LPA-specific receptors, and induces parasympathetic (vagal) activity. We demonstrate that this activation is sufficient to cause acute bronchoconstriction. Moreover, we show that prophylactic administration of TRPV1 (AMG9810) and LPA (BrP-LPA) receptor antagonists prevents bradykinin-induced asthmatic bronchoconstriction and, if administered following allergen exposure, reduces the associated respiratory distress. Our discovery provides mechanistic insight into the critical roles of carotid body LPA receptors in allergen-induced respiratory distress and suggests alternate treatment options for asthma.


Subject(s)
Acrylamides/therapeutic use , Asthma/prevention & control , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Carotid Body/metabolism , Lysophospholipids/therapeutic use , Receptors, Lysophosphatidic Acid/metabolism , TRPV Cation Channels/metabolism , Acrylamides/pharmacology , Animals , Asthma/etiology , Asthma/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Lysophospholipids/pharmacology , Male , Rats, Inbred BN , Rats, Sprague-Dawley , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , TRPV Cation Channels/antagonists & inhibitors
15.
Article in English | MEDLINE | ID: mdl-29462674

ABSTRACT

Lysophosphatidic acid (LPA) is a bioactive phospholipid that induces diverse biological responses. Recently, we found that LPA ameliorates NSAIDs-induced gastric ulcer in mice. Here, we quantified LPA in 21 medicinal herbs used for treatment of gastrointestinal (GI) disorders. We found that half of them contained LPA at relatively high levels (40-240 µg/g) compared to soybean seed powder (4.6 µg/g), which we previously identified as an LPA-rich food. The LPA in peony (Paeonia lactiflora) root powder is highly concentrated in the lipid fraction that ameliorates indomethacin-induced gastric ulcer in mice. Synthetic 18:1 LPA, peony root LPA and peony root lipid enhanced prostaglandin E2 production in a gastric cancer cell line, MKN74 cells that express LPA2 abundantly. These materials also prevented indomethacin-induced cell death and stimulated the proliferation of MKN74 cells. We found that LPA was present in stomach fluids at 2.4 µM, which is an effective LPA concentration for inducing a cellular response in vitro. These results indicated that LPA is one of the active components of medicinal herbs for the treatment of GI disorder and that orally administered LPA-rich herbs may augment the protective actions of endogenous LPA on gastric mucosa.


Subject(s)
Dinoprostone/metabolism , Indomethacin/adverse effects , Lysophospholipids/therapeutic use , Plants, Medicinal/chemistry , Animals , Cell Death/drug effects , Cell Proliferation/drug effects , Mice , Signal Transduction/drug effects , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Stomach Ulcer/metabolism
16.
Int J Mol Med ; 41(3): 1704-1714, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29286094

ABSTRACT

Inhibition of histone deacetylase-2 (HDAC2), which is a prohypertrophic factor in the heart, can functionally attenuate cardiac hypertrophy. The present study aimed to investigate whether sphingosine­1­phosphate (S1P), which has recently been reported to suppress HDAC2 activity, could ameliorate the cardiac hypertrophic response and improve cardiac function in mice with transverse aortic constriction (TAC), as well as to determine the underlying mechanisms. Briefly, 8­week­old male C57BL/6 mice were randomly divided into sham, TAC and TAC + S1P groups; the results indicated that S1P treatment attenuated TAC­induced cardiac dysfunction. In addition, heart size and the expression levels of fetal cardiac genes were reduced in the TAC + S1P group compared with in the TAC group. Furthermore, in cultured H9c2 cells exposed to phenylephrine, S1P was revealed to decrease cardiomyocyte size and the exaggerated expression of fetal cardiac genes. The present study also demonstrated that S1P had no effect on HDAC2 expression, but it did suppress its activity and increase acetylation of histone H3 in vivo and in vitro. Krüppel­like factor 4 (KLF4) is an antihypertrophic transcriptional regulator, which mediates HDAC inhibitor­induced prevention of cardiac hypertrophy; in the present study, KLF4 was upregulated by S1P. Finally, the results indicated that S1P receptor 2 (S1PR2) may be involved in the antihypertrophic effects, whereas the suppressive effects of S1P on HDAC2 activity were independent of S1PR2. In conclusion, the present study demonstrated that S1P treatment may ameliorate the cardiac hypertrophic response, which may be partly mediated by the suppression of HDAC2 activity and the upregulation of KLF4; it was suggested that S1PR2 may also be involved. Therefore, S1P may be considered a potential therapy for the treatment of heart diseases caused by cardiac hypertrophy.


Subject(s)
Cardiomegaly/drug therapy , Cardiomegaly/enzymology , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Lysophospholipids/therapeutic use , Sphingosine/analogs & derivatives , Animals , Aorta/pathology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cells, Cultured , Constriction, Pathologic , Electrocardiography , Hemodynamics/drug effects , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Lysophospholipids/administration & dosage , Lysophospholipids/pharmacology , Male , Mice, Inbred C57BL , Models, Biological , Phenylephrine , RNA, Small Interfering/metabolism , Rats , Receptors, Lysosphingolipid/metabolism , Sphingosine/administration & dosage , Sphingosine/pharmacology , Sphingosine/therapeutic use , Up-Regulation/drug effects
17.
Arch Med Res ; 48(2): 147-155, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28625317

ABSTRACT

BACKGROUND AND AIMS: Although preconditioning of sphingosine 1-phosphate (S1P) has been shown to protect myocytes from hypoxia reoxgenation injury in vitro, the role of S1P postconditioning on myocardial ischemia reperfusion injury (MIRI) in vivo and its related mechanism are unknown. The aim of this study was to investigate the protective role of sphingosine 1-phosphate (S1P) postconditioning in MIRI via its effects on mitochondrial signaling and Akt/Gsk3ß phosphorylation. METHODS: Rats were subjected to MIRI, consisting of 30 min of ischemia followed by 120 min of reperfusion, with S1P administered at the beginning of the reperfusion. Myocardial infarct size and apoptotic index were measured by triphenyltetrazolium (TTC) and terminal deoxynucleotide transferase dUTP nick-end labeling (TUNEL) assays, respectively. Akt and Gsk3ß phosphorylation, caspase-3 cleavage, and cytochrome c translocation were assessed by western blot. Mitochondrial permeability transition pore (MPTP) opening and mitochondrial membrane potential (MMP, ΔΨ) were also examined to determine overall mitochondrial function. RESULTS: S1P postconditioning significantly decreased myocardial infarct size and apoptosis, as well as enhanced Akt and Gsk3ß phosphorylation, attenuated caspase-3 cleavage and cytosolic cytochrome c translocation, and inhibited MPTP opening, which subsequently preserved Δψ. Electron microscopy also confirmed that S1P helped maintain myocardial mitochondria integrity. Moreover, the protective effects of S1P treatment were blocked by cotreatment with a PI3K inhibitor, LY294002. CONCLUSIONS: These results suggest that S1P postconditioning protects against MIRI by regulating mitochondrial signaling and Akt/Gsk3ß phosphorylation.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Ischemic Postconditioning , Lysophospholipids/pharmacology , Mitochondria, Heart/drug effects , Myocardial Reperfusion Injury/prevention & control , Proto-Oncogene Proteins c-akt/metabolism , Sphingosine/analogs & derivatives , Animals , Chromones/pharmacology , Cytochromes c/metabolism , Lysophospholipids/therapeutic use , Male , Membrane Potential, Mitochondrial , Mitochondria, Heart/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Morpholines/pharmacology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Transport , Rats, Wistar , Signal Transduction , Sphingosine/pharmacology , Sphingosine/therapeutic use
18.
Autoimmun Rev ; 16(5): 495-503, 2017 May.
Article in English | MEDLINE | ID: mdl-28279838

ABSTRACT

Inflammatory bowel diseases (IBD), including ulcerative colitis and Crohn's disease, involve an inappropriate immune reaction in the digestive tract, causing a variety of disabling symptoms. The advent of monoclonal antibodies (anti-tumor necrosis factor, anti-integrin, anti-interleukin -23) has revolutionized IBD management. Nevertheless, these agents, with potential for immunogenicity, are associated with high rates of response loss and disease relapse over time. They are also associated with high production costs. Sphingosine-1-phosphate (S1P), a membrane-derived lysophospholipid signaling molecule, is implicated in a vast array of physiological and pathophysiological processes, primarily via extracellular activation of S1P1-S1P5 receptors. S1P1, S1P4 and S1P5 are involved in regulation of the immune system, while S1P2 and S1P3 may be associated with cardiovascular, pulmonary, and theoretical cancer-related risks. Targeting S1P receptors for inflammatory conditions has been successful in clinical trials leading to approval of the non-selective S1P modulator, fingolimod, for relapsing forms of multiple sclerosis. However, the association of this non-selective S1P modulator with serious adverse events provides the rationale for developing more selective S1P receptor modulators. Until recently, three S1P modulators with differing selectivity for S1P receptors were in clinical development for IBD: ozanimod (RPC1063), etrasimod (APD334) and amiselimod (MT-1303). The development of amiselimod has been stopped as Biogen are currently focusing on other drugs in its portfolio. Following encouraging results from the Phase 2 TOUCHSTONE trial, a Phase 3 trial of the S1P modulator ozanimod in patients with moderate-to-severe ulcerative colitis is ongoing. Etrasimod is also being tested in a phase 2 trial in ulcerative colitis. These pipeline medications can be administered orally and may avoid the formation of anti-drug antibodies that can lead to treatment failure with injectable biologic therapies for IBD. Data from ongoing clinical trials will establish the relationship between the selectivity of S1P modulators and their safety and efficacy in IBD, as well as their potential place in the clinical armamentarium for IBD.


Subject(s)
Inflammatory Bowel Diseases/drug therapy , Lysophospholipids/therapeutic use , Sphingosine/analogs & derivatives , Humans , Inflammatory Bowel Diseases/pathology , Lysophospholipids/metabolism , Sphingosine/metabolism , Sphingosine/therapeutic use
19.
Gynecol Endocrinol ; 33(6): 476-479, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28277139

ABSTRACT

To investigate whether sphingosine-1-phosphate (S1P), an apoptosis-inhibitor would be able to inhibit chemotherapy induced human granulosa cell apoptosis. Cultures of primary granulosa cells were isolated from women undergoing in vitro fertilization (IVF). MTT assay was used to measure the optimum concentration of CTX and S1P acts on human granulosa cells. Granulosa cells were added with pertussis toxin (PTX), the PI3K inhibitor LY294002. Western blot analysis was used to analyze the signaling pathway of proteins and cell apoptosis. We found that S1P (10 mm) statistically significantly decreased granulosa cell apoptosis after cyclophosphamide (CTX) treatment. The decreased cell apoptosis induced by S1P was abolished after treatment with LY294002, PI3K inhibitor. CONCLUSIONS: Treatment with S1P can inhibit the CTX-induced granulosa cell apoptosis. The S1P protective effect is mediated by activating the PI3K/Akt pathway.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Cyclophosphamide/adverse effects , Granulosa Cells/drug effects , Lysophospholipids/therapeutic use , Primary Ovarian Insufficiency/prevention & control , Sphingosine/analogs & derivatives , Apoptosis/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Female , Humans , Lysophospholipids/pharmacology , Phosphorylation/drug effects , Primary Ovarian Insufficiency/chemically induced , Proto-Oncogene Proteins c-akt/metabolism , Sphingosine/pharmacology , Sphingosine/therapeutic use
20.
Mol Hum Reprod ; 22(12): 852-866, 2016 12.
Article in English | MEDLINE | ID: mdl-27645281

ABSTRACT

STUDY QUESTION: Are follicular fluid (FF) sphingosine-1-phosphate (S1P) levels in patients at risk of developing ovarian hyperstimulation syndrome (OHSS) altered and in part responsible for the high vascular permeability observed in these patients. STUDY ANSWER: FF S1P levels are lower in FF from patients at risk of OHSS and treatment with S1P may reduce vascular permeability in these patients. WHAT IS KNOWN ALREADY: Although advances have been made in the diagnosis, and management of OHSS and in basic knowledge of its development, complete prevention has proven difficult. STUDY DESIGN, SIZE, DURATION: A total of 40 FF aspirates were collected from patients undergoing ART. The women (aged 25-39 years old) were classified into a control group (n = 20) or a group at risk of OHSS (n = 20). The EA.hy926 endothelial cell line was used to assess the efffects of FF from patients at risk of OHSS with or without the addition of S1P. An animal model that develops OHSS in immature Sprague-Dawley rats were also used. PARTICIPANTS/MATERIALS, SETTING, METHODS: Migration assays, confocal microscopy analysis of actin filaments, immunoblotting and quail chorioallantoic membrane (CAM) assays of in-vivo angiogenesis were performed and statistical comparisons between groups were made. MAIN RESULTS AND THE ROLE OF CHANCE: The S1P concentration was significantly lower in FF from patients at risk of OHSS (P = 0.03). The addition of S1P to this FF decreased cell migration (P < 0.05) and prevented VE-cadherin phosphorylation in endothelial cells (P < 0.05). S1P in the FF from patients at risk of OHSS increased the levels of VE-cadherin (P < 0.05), N-cadherin (P < 0.05) and ß-catenin (P < 0.05), and partially reversed actin redistribution in endothelial cells. The addition of S1P in FF from patients at risk of OHSS also decreased the levels of vascular endothelial growth factor (VEGF121; P < 0.01) and S1P lyase (SPL; P < 0.05) and increased the levels of S1PR1 (P < 0.05) in endothelial cells. In CAMs incubated with FF from patients at risk of OHSS with S1P, the number of vessel branch points decreased while the periendothelial cell coverage increased. Additionally, in a rat OHSS model, we demonstrated that vascular permeability and VEGF121 and its receptor KDR expression were increased in the OHSS group compared to the control group and that S1P administration decreased these parameters. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: The results of this study were generated from an in-vitro system. This model reflects the microvasculature in vivo. Even though the ideal model would be the use of human endothelial cells from the ovary, it is obviously not possible to carry out this kind of approach in ovaries of patients from ART. More studies will be necessary to delineate the effects of S1P in the pathogenesis of OHSS. Hence, clinical studies are needed in order to choose the most appropriate method of prevention and management. WIDER IMPLICATIONS OF THE FINDINGS: The use of bioactive sphingolipid metabolites may contribute to finding better and safer therapeutic strategies for the treatment of OHSS and other human diseases that display aberrant vascular leakage. STUDY FUNDING/COMPETING INTERESTS: This work was supported by grants ANPCyT (PICT 2012-897), CONICET (PIP 5471), Roemmers and Baron Foundation, Argentina. The authors declare no conflict of interest.


Subject(s)
Lysophospholipids/pharmacology , Ovarian Hyperstimulation Syndrome/metabolism , Ovary/metabolism , Sphingosine/analogs & derivatives , Adult , Capillary Permeability/drug effects , Cell Line , Endothelial Cells/drug effects , Female , Follicular Fluid/metabolism , Humans , Immunoblotting , Lysophospholipids/therapeutic use , Microscopy, Confocal , Ovarian Hyperstimulation Syndrome/drug therapy , Ovary/drug effects , Sphingosine/pharmacology , Sphingosine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL