Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Front Neurol ; 11: 611816, 2020.
Article in English | MEDLINE | ID: mdl-33384658

ABSTRACT

Exposure to blast overpressure waves is implicated as the major cause of ocular injuries and resultant visual dysfunction in veterans involved in recent combat operations. No effective therapeutic strategies have been developed so far for blast-induced ocular dysfunction. Lysophosphatidic acid (LPA) is a bioactive phospholipid generated by activated platelets, astrocytes, choroidal plexus cells, and microglia and is reported to play major roles in stimulating inflammatory processes. The levels of LPA in the cerebrospinal fluid have been reported to increase acutely in patients with traumatic brain injury (TBI) as well as in a controlled cortical impact (CCI) TBI model in mice. In the present study, we have evaluated the efficacy of a single intravenous administration of a monoclonal LPA antibody (25 mg/kg) given at 1 h post-blast for protection against injuries to the retina and associated ocular dysfunctions. Our results show that a single 19 psi blast exposure significantly increased the levels of several species of LPA in blood plasma at 1 and 4 h post-blast. The anti-LPA antibody treatment significantly decreased glial cell activation and preserved neuronal cell morphology in the retina on day 8 after blast exposure. Optokinetic measurements indicated that anti-LPA antibody treatment significantly improved visual acuity in both eyes on days 2 and 6 post-blast exposure. Anti-LPA antibody treatment significantly increased rod photoreceptor and bipolar neuronal cell signaling in both eyes on day 7 post-blast exposure. These results suggest that blast exposure triggers release of LPAs, which play a major role promoting blast-induced ocular injuries, and that a single early administration of anti-LPA antibodies provides significant protection.

2.
Am J Pathol ; 188(8): 1779-1793, 2018 08.
Article in English | MEDLINE | ID: mdl-30037420

ABSTRACT

Lysophosphatidic acid (LPA) levels increase in the cerebrospinal fluid and blood within 24 hours after traumatic brain injury (TBI), indicating it may be a biomarker for subsequent cellular pathology. However, no data exist that document this association after TBI. We, therefore, acquired matrix-assisted laser desorption ionization imaging mass spectrometry data of LPA, major LPA metabolites, and hemoglobin from adult rat brains at 1 and 3 hours after controlled cortical impact injury. Data were semiquantitatively assessed by signal intensity analysis normalized to naïve rat brains acquired concurrently. Gray and white matter pathology was assessed on adjacent sections using immunohistochemistry for cell death, axonal injury, and intracellular LPA, to determine the spatiotemporal patterning of LPA corresponding to pathology. The results revealed significant increases in LPA and LPA precursors at 1 hour after injury and robust enhancement in LPA diffusively throughout the brain at 3 hours after injury. Voxel-wise analysis of LPA by matrix-assisted laser desorption ionization and ß-amyloid precursor protein by immunohistochemistry in adjacent sections showed significant association, raising the possibility that LPA is linked to secondary axonal injury. Total LPA and metabolites were also present in remotely injured areas, including cerebellum and brain stem, and in particular thalamus, where intracellular LPA is associated with cell death. LPA may be a useful biomarker of cellular pathology after TBI.


Subject(s)
Biomarkers/metabolism , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Lysophospholipids/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Male , Rats , Rats, Sprague-Dawley
3.
Pain ; 158(11): 2181-2188, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29028747

ABSTRACT

Lysophosphatidic acid (LPA) is a bioactive lipid that impacts neurological outcomes after neurotrauma by inhibiting neuroregeneration, promoting inflammation, and contributing to behavioral deficits. Blocking LPA signaling with a novel anti-LPA monoclonal antibody (mAb) is neuroprotective after traumatic brain injury (TBI) if given to injured animals whose blood-brain barrier (BBB) has been compromised. It is hypothesized that the anti-LPA mAb could improve chronic pain initiated by TBI. However, poor brain penetration after systemic application of the antibody makes access to the central nervous system (CNS) problematic in situations where the BBB is intact. Our experiments investigated whether intranasal delivery of the anti-LPA mAb could bypass the BBB, allowing for direct entry of the antibody to certain areas of the CNS. When the humanized anti-LPA mAb, LT3114, was intranasally applied to injured rats within 30 minutes after mild TBI using the central lateral percussion model, enzyme-linked immunospecific assay and immunohistochemistry demonstrated antibody uptake to several areas in the CNS, including the area of cortical injury, the corpus callosum, cerebellum, and the subventricular region. Compared with control rats that received LT3114 but no TBI, TBI rats demonstrated significantly higher concentrations of intranasally administered LT3114 antibody in some tissues. In behavioral studies, a significant attenuation of mechanical allodynia after TBI was observed in the anti-LPA treatment group (P = 0.0079), when compared with vehicle controls within 14 days after TBI. These results suggest that intranasal application of the anti-LPA antibody directly accesses CNS sites involved in TBI-related pain and that this access attenuates pain sequelae to the neurotrauma.


Subject(s)
Antibodies/administration & dosage , Brain Injuries, Traumatic/complications , Hyperalgesia/etiology , Hyperalgesia/therapy , Lysophospholipids/immunology , Administration, Intranasal , Animals , Antibodies/blood , Antibodies/cerebrospinal fluid , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/cerebrospinal fluid , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Male , Pain Measurement , Rats , Rats, Sprague-Dawley
4.
Oncol Rep ; 38(4): 1932-1940, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28765947

ABSTRACT

Glioblastoma multiforme (GBM) is the most common primary, intracranial malignancy of the central nervous system. The standard treatment protocol, which involves surgical resection, and concurrent radiation with adjuvant temozolomide (TMZ), still imparts a grim prognosis. Ultimately, all GBMs exhibit recurrence or progression, developing resistance to standard treatment. This study demonstrates that GBMs acquire resistance to radiation via upregulation of acid ceramidase (ASAH1) and sphingosine­1-phosphate (Sph-1P). Moreover, inhibition of ASAH1 and Sph-1P, either with humanized monoclonal antibodies, small molecule drugs (i.e. carmofur), or a combination of both, led to suppression of GBM cell growth. These results suggest that ASAH1 and Sph-1P may be excellent targets for the treatment of new GBMs and recurrent GBMs, especially since the latter overexpresses ASAH1.


Subject(s)
Acid Ceramidase/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/radiotherapy , Glioblastoma/enzymology , Glioblastoma/radiotherapy , Acid Ceramidase/biosynthesis , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunohistochemistry , Lysophospholipids/metabolism , Neoplasm Recurrence, Local/enzymology , Neoplasm Recurrence, Local/pathology , Radiation Tolerance , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Up-Regulation
5.
Sci Signal ; 10(472)2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28351953

ABSTRACT

Sphingosine kinase 1 (SphK1) promotes cell proliferation and survival, and its abundance is often increased in tumors. SphK1 produces the signaling lipid sphingosine 1-phosphate (S1P), which activates signaling cascades downstream five G protein-coupled receptors (S1P1-5) to modulate vascular and immune system function and promote proliferation. We identified a new function of the SphK1-S1P pathway specifically in the control of mitosis. SphK1 depletion in HeLa cells caused prometaphase arrest, whereas its overexpression or activation accelerated mitosis. Increasing the abundance of S1P promoted mitotic progression, overrode the spindle assembly checkpoint (SAC), and led to chromosome segregation defects. S1P was secreted through the transporter SPNS2 and stimulated mitosis by binding to and activating S1P5 on the extracellular side, which then activated the intracellular phosphatidylinositol 3-kinase (PI3K)-AKT pathway. Knockdown of S1P5 prevented the S1P-induced spindle defect phenotype. RNA interference assays revealed that the mitotic kinase Polo-like kinase 1 (PLK1) was an important effector of S1P-S1P5 signaling-induced mitosis in HeLa cells. Our findings identify an extracellular signal and the downstream pathway that promotes mitotic progression and may indicate potential therapeutic targets to inhibit the proliferation of cancer cells.


Subject(s)
Chromosome Segregation/drug effects , Lysophospholipids/pharmacology , Mitosis/drug effects , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Animals , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , HeLa Cells , Humans , Mice, Knockout , Microscopy, Confocal , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Receptors, Lysosphingolipid/genetics , Sphingosine/pharmacology , Time-Lapse Imaging/methods , Polo-Like Kinase 1
6.
Oncotarget ; 6(15): 13803-21, 2015 May 30.
Article in English | MEDLINE | ID: mdl-25915662

ABSTRACT

Hypoxia promotes neovascularization, increased tumor growth, and therapeutic resistance. The transcription factor, hypoxia-inducible factor 1α (HIF-1α), has been reported as the master driver of adaptation to hypoxia. We previously identified the sphingosine kinase 1/sphingosine 1-phosphate (SphK1/S1P) pathway as a new modulator of HIF-1α under hypoxia. Taking advantage of a monoclonal antibody neutralizing extracellular S1P (sphingomab), we report that inhibition of S1P extracellular signaling blocks HIF-1α accumulation and activity in several cancer cell models exposed to hypoxia. In an orthotopic xenograft model of prostate cancer, we show that sphingomab reduces hypoxia and modifies vessel architecture within 5 days of treatment, leading to increased intratumoral blood perfusion. Supporting the notion that a transient vascular normalization of tumor vessels is the mechanism by which sphingomab exerts its effects, we demonstrate that administration of the antibody for 5 days before chemotherapy is more effective at local tumor control and metastatic dissemination than any other treatment scheduling. These findings validate sphingomab as a potential new normalization agent that could contribute to successful sensitization of hypoxic tumors to chemotherapy.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/immunology , Prostatic Neoplasms/drug therapy , Sphingosine/analogs & derivatives , Animals , Cell Hypoxia/drug effects , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Random Allocation , Signal Transduction , Sphingosine/antagonists & inhibitors , Sphingosine/immunology , Vascular Remodeling/drug effects
7.
Clin Cancer Res ; 21(8): 1925-1934, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25589614

ABSTRACT

PURPOSE: VEGFR2 tyrosine kinase inhibition (TKI) is a valuable treatment approach for patients with metastatic renal cell carcinoma (RCC). However, resistance to treatment is inevitable. Identification of novel targets could lead to better treatment for patients with TKI-naïve or -resistant RCC. EXPERIMENTAL DESIGN: In this study, we performed transcriptome analysis of VEGFR TKI-resistant tumors in a murine model and discovered that the SPHK-S1P pathway is upregulated at the time of resistance. We tested sphingosine-1-phosphate (S1P) pathway inhibition using an anti-S1P mAb (sphingomab), in two mouse xenograft models of RCC, and assessed tumor SPHK expression and S1P plasma levels in patients with metastatic RCC. RESULTS: Resistant tumors expressed several hypoxia-regulated genes. The SPHK1 pathway was among the most highly upregulated pathways that accompanied resistance to VEGFR TKI therapy. SPHK1 was expressed in human RCC, and the product of SPHK1 activity, S1P, was elevated in patients with metastatic RCC, suggesting that human RCC behavior could, in part, be due to overproduction of S1P. Sphingomab neutralization of extracellular S1P slowed tumor growth in both mouse models. Mice bearing tumors that had developed resistance to sunitinib treatment also exhibited tumor growth suppression with sphingomab. Sphingomab treatment led to a reduction in tumor blood flow as measured by MRI. CONCLUSIONS: Our findings suggest that S1P inhibition may be a novel therapeutic strategy in patients with treatment-naïve RCC and also in the setting of resistance to VEGFR TKI therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lysophospholipids/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, Vascular Endothelial Growth Factor/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Line, Tumor , Cluster Analysis , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Lysophospholipids/antagonists & inhibitors , Mice , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Sphingosine/antagonists & inhibitors , Sphingosine/metabolism , Transcriptome , Tumor Burden/drug effects , Up-Regulation , Xenograft Model Antitumor Assays
8.
Mol Oncol ; 8(7): 1181-95, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24768038

ABSTRACT

Sphingosine 1-phosphate (S1P) plays important roles in cell proliferation, differentiation or survival mainly through its surface G-protein-coupled receptors S1P1-5. Bone represents the major site of metastasis for prostate cancer (CaP) cells, which rely on bone-derived factors to support their proliferation and resistance to therapeutics. In the present work we have found that conditioned medium (CM) from the MC3T3 osteoblastic cell line or primary murine and human osteoblast-like cells, as well as co-culture with MC3T3 stimulate proliferation of CaP lines in S1P-dependent manner. In addition, osteoblastic-derived S1P induces resistance of CaP cells to therapeutics including chemotherapy and radiotherapy. When S1P release from osteoblastic cells is decreased (inhibition of SphK1, knock-down of SphK1 or the S1P transporter, Spns2 by siRNA) or secreted S1P neutralized with anti-S1P antibody, the proliferative and survival effects of osteoblasts on CaP cells are abolished. Because of the paracrine nature of the signaling, we studied the role of the S1P receptors expressed on CaP cells in the communication with S1P secreted by osteoblasts. Strategies aimed at down-regulating S1P1, S1P2 or S1P3 (siRNA, antagonists), established the exclusive role of the S1P/S1P1 signaling between osteoblasts and CaP cells. Bone metastases from CaP are associated with osteoblastic differentiation resulting in abnormal bone formation. We show that the autocrine S1P/S1P3 signaling is central during differentiation to mature osteoblasts by regulating Runx2 level, a key transcription factor involved in osteoblastic maturation. Importantly, differentiated osteoblasts exhibited enhanced secretion of S1P and further stimulated CaP cell proliferation in a S1P-dependent manner. By establishing the dual role of osteoblast-borne S1P on both osteoblastic differentiation and CaP cell proliferation and survival, we uncover the importance of S1P in the bone metastatic microenvironment, which may open a novel area of study for the treatment of CaP bone metastasis by targeting S1P.


Subject(s)
Bone Neoplasms/secondary , Lysophospholipids/metabolism , Osteoblasts/pathology , Prostate/pathology , Prostatic Neoplasms/pathology , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Cell Differentiation/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Drug Resistance, Neoplasm , Humans , Male , Mice , Osteoblasts/cytology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Signal Transduction , Sphingosine/metabolism
9.
J Neuroinflammation ; 11: 37, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24576351

ABSTRACT

BACKGROUND: Lysophosphatidic acid (LPA) is a bioactive phospholipid with a potentially causative role in neurotrauma. Blocking LPA signaling with the LPA-directed monoclonal antibody B3/Lpathomab is neuroprotective in the mouse spinal cord following injury. FINDINGS: Here we investigated the use of this agent in treatment of secondary brain damage consequent to traumatic brain injury (TBI). LPA was elevated in cerebrospinal fluid (CSF) of patients with TBI compared to controls. LPA levels were also elevated in a mouse controlled cortical impact (CCI) model of TBI and B3 significantly reduced lesion volume by both histological and MRI assessments. Diminished tissue damage coincided with lower brain IL-6 levels and improvement in functional outcomes. CONCLUSIONS: This study presents a novel therapeutic approach for the treatment of TBI by blocking extracellular LPA signaling to minimize secondary brain damage and neurological dysfunction.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/immunology , Immunoglobulin G/therapeutic use , Immunologic Factors/therapeutic use , Lysophospholipids/immunology , Adult , Aged, 80 and over , Animals , Brain Injuries/cerebrospinal fluid , Cytokines/metabolism , Disease Models, Animal , Female , Glasgow Coma Scale , Humans , Lysophospholipids/cerebrospinal fluid , Male , Mice , Mice, Inbred C57BL , Middle Aged , Single-Blind Method , Young Adult
10.
Am J Pathol ; 181(3): 978-92, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22819724

ABSTRACT

Evidence suggests a proinflammatory role of lysophosphatidic acid (LPA) in various pathologic abnormalities, including in the central nervous system. Herein, we describe LPA as an important mediator of inflammation after spinal cord injury (SCI) in zebrafish and mice. Furthermore, we describe a novel monoclonal blocking antibody raised against LPA that potently inhibits LPA's effect in vitro and in vivo. This antibody, B3, specifically binds LPA, prevents it from interacting with its complement of receptors, and blocks LPA's effects on the neuronal differentiation of human neural stem/progenitor cells, demonstrating its specificity toward LPA signaling. When administered systemically to mice subjected to SCI, B3 substantially reduced glial inflammation and neuronal death. B3-treated animals demonstrated significantly more neuronal survival upstream of the lesion site, with some functional improvement. This study describes the use of anti-LPA monoclonal antibody as a novel therapeutic approach for the treatment of SCI.


Subject(s)
Lysophospholipids/antagonists & inhibitors , Recovery of Function , Signal Transduction , Spinal Cord Injuries/pathology , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , CHO Cells , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cricetinae , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Inflammation/complications , Inflammation/pathology , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Mice , Microglia/drug effects , Microglia/pathology , Motor Activity/drug effects , Neurites/drug effects , Neurites/metabolism , Neuroprotective Agents/pharmacology , Receptors, Lysophosphatidic Acid/metabolism , Recovery of Function/drug effects , Signal Transduction/drug effects , Spinal Cord Injuries/complications , Spinal Cord Injuries/physiopathology , Zebrafish
11.
Int Rev Cell Mol Biol ; 296: 273-322, 2012.
Article in English | MEDLINE | ID: mdl-22559941

ABSTRACT

Lysophosphatidic acid (LPA) is a bioactive lipid that regulates a broad range of cellular effects in various cell types, leading to a variety of responses in tissues, including in the nervous system. LPA and its receptors are found in the nervous system, with different cellular and temporal profiles. Through its ability to target most cells of the nervous system and its induction of pleiotropic effects, LPA mediates events during neural development and adulthood. In this review, we summarize the current knowledge on the effects of LPA in the nervous system, during development and adulthood, and in various pathologies of the nervous system. We also explore potential LPA intervention strategies for anti-LPA therapeutics.


Subject(s)
Lysophospholipids/metabolism , Nervous System/embryology , Nervous System/metabolism , Animals , Humans , Nervous System/cytology , Nervous System/growth & development , Receptors, Lysophosphatidic Acid/biosynthesis
12.
PLoS One ; 6(1): e16571, 2011 Jan 31.
Article in English | MEDLINE | ID: mdl-21304987

ABSTRACT

BACKGROUND: Earlier we have shown that extracellular sphingosine-1-phosphate (S1P) induces migration of human pulmonary artery endothelial cells (HPAECs) through the activation of S1P(1) receptor, PKCε, and PLD2-PKCζ-Rac1 signaling cascade. As endothelial cells generate intracellular S1P, here we have investigated the role of sphingosine kinases (SphKs) and S1P lyase (S1PL), that regulate intracellular S1P accumulation, in HPAEC motility. METHODOLOGY/PRINCIPAL FINDINGS: Inhibition of SphK activity with a SphK inhibitor 2-(p-Hydroxyanilino)-4-(p-Chlorophenyl) Thiazole or down-regulation of Sphk1, but not SphK2, with siRNA decreased S1P(int), and attenuated S1P(ext) or serum-induced motility of HPAECs. On the contrary, inhibition of S1PL with 4-deoxypyridoxine or knockdown of S1PL with siRNA increased S1P(int) and potentiated motility of HPAECs to S1P(ext) or serum. S1P(ext) mediates cell motility through activation of Rac1 and IQGAP1 signal transduction in HPAECs. Silencing of SphK1 by siRNA attenuated Rac1 and IQGAP1 translocation to the cell periphery; however, knockdown of S1PL with siRNA or 4-deoxypyridoxine augmented activated Rac1 and stimulated Rac1 and IQGAP1 translocation to cell periphery. The increased cell motility mediated by down-regulation was S1PL was pertussis toxin sensitive suggesting "inside-out" signaling of intracellularly generated S1P. Although S1P did not accumulate significantly in media under basal or S1PL knockdown conditions, addition of sodium vanadate increased S1P levels in the medium and inside the cells most likely by blocking phosphatases including lipid phosphate phosphatases (LPPs). Furthermore, addition of anti-S1P mAb to the incubation medium blocked S1P(ext) or 4-deoxypyridoxine-dependent endothelial cell motility. CONCLUSIONS/SIGNIFICANCE: These results suggest S1P(ext) mediated endothelial cell motility is dependent on intracellular S1P production, which is regulated, in part, by SphK1 and S1PL.


Subject(s)
Aldehyde-Lyases/physiology , Cell Movement , Endothelial Cells/cytology , Lung/cytology , Lysophospholipids/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/physiology , Sphingosine/analogs & derivatives , Humans , Lysophospholipids/physiology , RNA, Small Interfering/pharmacology , Sphingosine/biosynthesis , Sphingosine/physiology
13.
Br J Pharmacol ; 162(6): 1225-38, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21091645

ABSTRACT

Sphingosine-1-phosphate (S1P) is a pleiotropic bioactive lipid thought to be dysregulated in a variety of disease conditions. In this review, we discuss the roles of S1P in cancer and in wet age-related macular degeneration. We also explore potential treatment strategies for these disorders, including the utility of anti-S1P antibodies acting as molecular sponges to neutralize dysregulated S1P in relevant tissues.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lysophospholipids/immunology , Lysophospholipids/metabolism , Neoplasms/therapy , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Wet Macular Degeneration/therapy , Drug Discovery , Humans , Molecular Targeted Therapy , Neoplasms/metabolism , Signal Transduction/drug effects , Sphingosine/immunology , Sphingosine/metabolism
14.
Cardiovasc Res ; 82(2): 303-12, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19228708

ABSTRACT

AIMS: Following injury, fibroblasts transform into myofibroblasts and produce extracellular matrix (ECM). Excess production of ECM associated with cardiac fibrosis severely inhibits cardiac function. Sphingosine-1-phosphate (S1P), a bioactive lysophospholipid, regulates the function of numerous cell types. In this study, we determined the role of S1P in promoting pro-fibrotic actions of cardiac fibroblasts (CFs). METHODS AND RESULTS: S1P-mediated effects on myofibroblast transformation, collagen production, and cross-talk with transforming growth factor-beta (TGF-beta) using mouse CF were examined. S1P increased alpha-smooth muscle actin (a myofibroblast marker) and collagen expression in a S1P2 receptor- and Rho kinase-dependent manner. TGF-beta increased sphingosine kinase 1 (SphK1; the enzyme responsible for S1P production) expression and activity. TGF-beta-stimulated collagen production was inhibited by SphK1 or S1P2 siRNA, a SphK inhibitor, and an anti-S1P monoclonal antibody. CONCLUSION: These findings suggest that TGF-beta-stimulated collagen production in CF involves 'inside-out' S1P signalling whereby S1P produced intracellularly by SphK1 can be released and act in an autocrine/paracrine fashion to activate S1P2 and increase collagen production.


Subject(s)
Collagen/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Lymphotoxin-alpha/pharmacology , Lysophospholipids/metabolism , Myocardium/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/analogs & derivatives , Animals , Antibodies, Monoclonal/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Lysophospholipids/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Myocardium/cytology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Signal Transduction/physiology , Sphingosine/antagonists & inhibitors , Sphingosine/metabolism , rho-Associated Kinases/metabolism
15.
Exp Eye Res ; 87(4): 367-75, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18687328

ABSTRACT

Sphingosine-1-phosphate (S1P) is a pleiotropic lysolipid that has recently been implicated in the regulation of tissue fibrosis. However, the fibrogenic potential of S1P in the eye has not previously been investigated. In the current study, we evaluated cells from the anterior and posterior segments of the eye for the presence of S1P and their potential ability to produce and respond to S1P. In addition, we investigated the regulatory role of S1P as a mediator of proliferation, cellular transformation and pro-fibrotic protein expression in human retinal pigmented epithelial cells. Expression of S1P receptors and sphingosine kinases (the enzymes that produce S1P) was examined using RT-PCR, and intracellular localization of S1P was examined using immunoblotting, immunohistochemistry and ELISA in primary human retinal pigmented epithelial (RPE) cells, primary human conjunctival fibroblasts (ConF), and primary human corneal fibroblasts (CF). RPE cell proliferation was determined using an MTT-based cell proliferation assay, and RPE myofibroblast transformation, collagen type I production and profibrotic protein expression were assessed using immunofluorescence, ELISA and immunoblot. S1P(1-3, 5) receptors and sphingosine kinases 1 and 2 were expressed and intracellular pools of S1P were detected in RPE cells, ConF and CF. S1P stimulated RPE cell proliferation in a dose- and time-dependent manner. S1P induced myofibroblast transformation of RPE cells, as indicated by increased alpha-smooth muscle actin (alpha-SMA) expression and its incorporation into prominent stress fibers, and promoted collagen type I production. S1P stimulated the expression of plasminogen activator inhibitor-1 (PAI-1) and heat shock protein 47 (HSP47), two proteins that are linked to increased tissue fibrosis. Combined, these data demonstrate that RPE cells, ConF and CF from the human eye not only have the molecular ability to produce and respond to S1P, but also contain S1P. Furthermore, S1P promotes proliferation, myofibroblast transformation, collagen production and pro-fibrotic protein expression by human RPE cells. These data suggest that S1P is a previously unrecognized mediator of profibrotic cellular function and signaling in the eye.


Subject(s)
Eye/metabolism , Lysophospholipids/physiology , Sphingosine/analogs & derivatives , Anterior Eye Segment/metabolism , Cell Proliferation/drug effects , Cell Transdifferentiation/drug effects , Cells, Cultured , Collagen Type I/biosynthesis , Dose-Response Relationship, Drug , Fibroblasts/cytology , Fibroblasts/drug effects , HSP47 Heat-Shock Proteins/metabolism , Humans , Lysophospholipids/analysis , Lysophospholipids/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Lysosphingolipid/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Sphingosine/analysis , Sphingosine/pharmacology , Sphingosine/physiology
16.
Am J Physiol Cell Physiol ; 294(1): C36-46, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17942639

ABSTRACT

Sphingosine 1-phosphate (S1P) mediates a number of cellular responses, including growth and proliferation. Skeletal muscle possesses the full enzymatic machinery to generate S1P and expresses the transcripts of S1P receptors. The aim of this work was to localize S1P receptors in rat skeletal muscle and to investigate whether S1P exerts a trophic action on muscle fibers. RT-PCR and Western blot analyses demonstrated the expression of S1P(1) and S1P(3) receptors by soleus muscle. Immunofluorescence revealed that S1P(1) and S1P(3) receptors are localized at the cell membrane of muscle fibers and in the T-tubule membranes. The receptors also decorate the nuclear membrane. S1P(1) receptors were also present at the neuromuscular junction. The possible trophic action of S1P was investigated by utilizing the denervation atrophy model. Rat soleus muscle was analyzed 7 and 14 days after motor nerve cut. During denervation, S1P was continuously delivered to the muscle through a mini osmotic pump. S1P and its precursor, sphingosine (Sph), significantly attenuated the progress of denervation-induced muscle atrophy. The trophic effect of Sph was prevented by N,N-dimethylsphingosine, an inhibitor of Sph kinase, the enzyme that converts Sph into S1P. Neutralization of circulating S1P by a specific antibody further demonstrated that S1P was responsible for the trophic effects of S1P during denervation atrophy. Denervation produced the down regulation of S1P(1) and S1P(3) receptors, regardless of the presence of the receptor agonist. In conclusion, the results suggest that S1P acts as a trophic factor of skeletal muscle.


Subject(s)
Lysophospholipids/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Antibodies , Cell Enlargement , Cell Membrane/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hypertrophy , Infusion Pumps, Implantable , Lysophospholipids/administration & dosage , Male , Muscle Denervation , Muscle, Skeletal/enzymology , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Muscular Atrophy/prevention & control , MyoD Protein/metabolism , Myogenin/metabolism , Myosin Heavy Chains/metabolism , Neuromuscular Junction/metabolism , Nuclear Envelope/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/immunology , Sciatic Nerve/surgery , Sphingosine/administration & dosage , Sphingosine/metabolism , Sphingosine/pharmacology , Time Factors
17.
Vaccine ; 25(12): 2279-87, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-17258845

ABSTRACT

In the midst of new investigations into the mechanisms of both delivery and protection of new vaccines and vaccine carriers, it has become clear that immunization with delivery mechanisms that do not involve living, replicating organisms are vastly preferred. In this report, non-replicating bacterial minicells simultaneously co-delivering the nucleoprotein (NP) of lymphocytic choriomeningitis virus (LCMV) and the corresponding DNA vaccine were tested for the ability to generate protective cellular immune responses in mice. It was found that good protection (89%) was achieved after intramuscular administration, moderate protection (31%) was achieved after intranasal administration, and less protection (7%) was achieved following gastric immunization. These results provide a solid foundation on which to pursue the use of bacterial minicells as a non-replicating vaccine delivery platform.


Subject(s)
Immunization/methods , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Nucleoproteins/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Administration, Intranasal , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , COS Cells , Chlorocebus aethiops , Cytokines/metabolism , Cytotoxicity, Immunologic/immunology , Drug Delivery Systems/methods , Escherichia coli/virology , Injections, Intramuscular , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/growth & development , Mice , Mice, Inbred C57BL , Nucleoproteins/genetics , Vero Cells , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
18.
Cell Microbiol ; 8(10): 1624-33, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16984417

ABSTRACT

The delivery of DNA to mammalian cells is of critical importance to the development of genetic vaccines, gene replacement therapies and gene silencing. For these applications, targeting, effective DNA transfer and vector safety are the major roadblocks in furthering development. In this report, we present a novel DNA delivery vehicle that makes use of protoplasted, achromosomal bacterial minicells. Transfer of plasmid DNA as measured by green fluorescent protein expression was found to occur in as high as 25% of cultured Cos-7 cells when a novel chimeric protein containing the D2-D5 region of invasin was expressed and displayed on the surface of protoplasted minicells. Based on endoplasmic reticulum stress and other responses, protoplasted minicells were non-toxic to recipient eukaryotic cells as a consequence of the transfection process. Taken together, these results suggest that bacterial minicells may represent a novel and promising gene delivery vehicle.


Subject(s)
Adhesins, Bacterial/genetics , Gene Transfer Techniques , Plasmids , Yersinia pseudotuberculosis/genetics , Animals , COS Cells , Chlorocebus aethiops , Electroporation , Green Fluorescent Proteins/genetics , Protoplasts , Recombinant Fusion Proteins/genetics , Transfection
19.
Vaccine ; 24(33-34): 6009-17, 2006 Aug 14.
Article in English | MEDLINE | ID: mdl-16806602

ABSTRACT

Recent events surrounding emerging infectious diseases, bioterrorism and increasing multidrug antibiotic resistance in bacteria have drastically increased current needs for effective vaccines. Many years of study have shown that live, attenuated pathogens are often more effective at delivering heterologous protein or DNA to induce protective immune responses. However, these vaccine carriers have inherent safety concerns that have limited their development and their use in many patient populations. Studies using nonliving delivery mechanisms have shown that providing both protein antigen and DNA encoding the antigen to an individual induces an improved, more protective immune response but rarely, if ever, are both delivered simultaneously. Here, non-replicating bacterial minicells derived from a commensal E. coli strain are shown to effectively induce antigen-specific immune responses after simultaneous protein and DNA delivery. These data demonstrate the potential use of achromosomal bacterial minicells as a vaccine carrier.


Subject(s)
Antibody Formation , Escherichia coli/genetics , Green Fluorescent Proteins/immunology , Vaccines, DNA/immunology , Vaccines, Synthetic/immunology , Administration, Intranasal , Animals , Antigen-Presenting Cells/immunology , Cell Line , Enzyme-Linked Immunosorbent Assay , Escherichia coli/immunology , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunity, Mucosal , Immunoglobulin A/blood , Immunoglobulin G/blood , Injections, Intramuscular , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Vaccines, DNA/administration & dosage , Vaccines, Synthetic/administration & dosage
20.
Cancer Cell ; 9(3): 225-38, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16530706

ABSTRACT

S1P has been proposed to contribute to cancer progression by regulating tumor proliferation, invasion, and angiogenesis. We developed a biospecific monoclonal antibody to S1P to investigate its role in tumorigenesis. The anti-S1P mAb substantially reduced tumor progression and in some cases eliminated measurable tumors in murine xenograft and allograft models. Tumor growth inhibition was attributed to antiangiogenic and antitumorigenic effects of the antibody. The anti-S1P mAb blocked EC migration and resulting capillary formation, inhibited blood vessel formation induced by VEGF and bFGF, and arrested tumor-associated angiogenesis. The anti-S1P mAb also neutralized S1P-induced proliferation, release of proangiogenic cytokines, and the ability of S1P to protect tumor cells from apoptosis in several tumor cell lines, validating S1P as a target for therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lysophospholipids/immunology , Neoplasm Invasiveness/prevention & control , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Sphingosine/analogs & derivatives , Animals , Antibody Specificity , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Sphingosine/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...