Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Vasc Res ; 57(4): 178-184, 2020.
Article in English | MEDLINE | ID: mdl-32434183

ABSTRACT

BACKGROUND: Lysophosphatidic acid (LPA) is a small phospholipid-signaling molecule, which can alter responses to stress in the central nervous system. OBJECTIVE: We hypothesized that exogenous LPA would increase the size of infarct and reduce microregional O2 supply/consumption balance after cerebral ischemia-reperfusion. METHODS: This was tested in isoflurane-anesthetized rats with middle cerebral artery blockade for 1 h and reperfusion for 2 h with or without LPA (1 mg/kg, at 30, 60, and 90 min after reperfusion). Regional cerebral blood flow was determined using a C14-iodoantipyrine autoradiographic technique. Regional small-vessel (20-60 µm in diameter) arterial and venous oxygen saturations were determined microspectrophotometrically. RESULTS: There were no significant hemodynamic or arterial blood gas differences between groups. The control ischemic-reperfused cortex had a similar O2 consumption to the contralateral cortex. However, microregional O2 supply/consumption balance was significantly reduced in the ischemic-reperfused cortex with many areas of low O2 saturation (43 of 80 veins with O2 saturation below 50%). LPA did not significantly alter cerebral blood flow, but it did significantly increase O2 extraction and consumption of the ischemic-reperfused region. It also significantly increased the number of small veins with low O2 saturations in the reperfused region (76 of 80 veins with O2 saturation below 50%). This was associated with a significantly increased cortical infarct size after LPA administration (11.4 ± 0.5% control vs. 16.4 ± 0.6% LPA). CONCLUSION: This suggests that LPA reduces cell survival and that it is associated with an increase in the number of small microregions with reduced local oxygen balance after cerebral ischemia-reperfusion.


Subject(s)
Cerebral Cortex/blood supply , Cerebral Cortex/drug effects , Cerebrovascular Circulation/drug effects , Infarction, Middle Cerebral Artery/pathology , Lysophospholipids/toxicity , Microcirculation/drug effects , Oxygen Consumption/drug effects , Oxygen/blood , Reperfusion Injury/pathology , Animals , Cell Death/drug effects , Cerebral Cortex/pathology , Cerebral Veins/drug effects , Cerebral Veins/pathology , Cerebral Veins/physiopathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/physiopathology , Male , Rats, Inbred F344 , Reperfusion Injury/blood , Reperfusion Injury/physiopathology
2.
J Stroke Cerebrovasc Dis ; 29(10): 105029, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32912542

ABSTRACT

BACKGROUND: We investigated whether exogenous lysophosphatidic acid (LPA), a phospholipid extracellular signaling molecule, would increase infarct size and blood-brain barrier (BBB) disruption during the early stage of cerebral ischemia-reperfusion, and whether it works through Akt-mTOR-S6K1 intracellular signaling. MATERIAL AND METHODS: Rats were given either vehicle or LPA 1 mg/kg iv three times during reperfusion after one hour of middle cerebral artery (MCA) occlusion. In another group, prior to administration of LPA, 30 mg/kg of PF-4708671, an S6K1 inhibitor, was injected. After one hour of MCA occlusion and two hours of reperfusion the transfer coefficient (Ki) of 14C-α-aminoisobutyric acid and the volume of 3H-dextran distribution were determined to measure the degree of BBB disruption. At the same time, the size of infarct was determined and western blot analysis was performed to determine the levels of phosphorylated Akt (p-Akt) and phosphorylated S6 (pS6). RESULTS: LPA increased the Ki in the ischemic-reperfused cortex (+43%) when compared with Control rats and PF-4708671 pretreatment prevented the increase of Ki by LPA. LPA increased the percentage of cortical infarct out of total cortical area (+36%) and PF-4708671 pretreatment prevented the increase of the infarct size. Exogenous LPA did not significantly change the levels of p-Akt as well as pS6 in the ischemic-reperfused cortex. CONCLUSION: Our data demonstrate that the increase in BBB disruption could be one of the reasons of the increased infarct size by LPA. S6K1 may not be the major target of LPA. A decrease of LPA during early cerebral ischemia-reperfusion might be beneficial for neuronal survival.


Subject(s)
Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Cerebral Cortex/drug effects , Infarction, Middle Cerebral Artery/therapy , Lysophospholipids/toxicity , Reperfusion Injury/chemically induced , Reperfusion , Animals , Blood-Brain Barrier/physiopathology , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats, Inbred F344 , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Ribosomal Protein S6 Kinases/metabolism
3.
Cell Physiol Biochem ; 53(6): 921-932, 2019.
Article in English | MEDLINE | ID: mdl-31778305

ABSTRACT

BACKGROUND/AIMS: Lysophosphatidic acid (LPA) is a phospholipid signal molecule that regulates many cellular processes both physiological and pathological. Moreover, its high plasma concentrations are toxic for several cellular types, including erythrocytes (RBC), as it acts as a pro-thrombotic and pro-atherogenic agent. It is therefore essential to explore the potential protective role of nutrition in protecting cells from the possible toxic effects of high plasma concentrations of LPA by testing bioactive nutrients. In particular, our focus was on hydroxytyrosol (HT), a phenolic antioxidant occurring naturally in virgin olive oil, investigating its possible protective effect in preventing LPA-induced programmed cell death (eryptosis) in human RBC. METHODS: Intact RBC were incubated in the presence of 2.5 µM LPA and increasing concentrations of HT. Phosphatidylserine (PS) exposure with cell shrinkage, influx of extracellular calcium (Ca2+), adenosine triphosphate (ATP) and glutathione levels were measured by FACS analysis. In addition, confocal laser scanning microscopy was used to determine RBC morphological alterations, as well as microvesicle formation. RESULTS: Our study confirms that LPA-induced eryptosis is characterized by PS exposure at the cell surface, with cell shrinkage and ATP and glutathione depletion; (Ca2+) influx is also a key event that triggers eryptosis. Here we report for the first time that cell co-incubation with LPA and in quantities as low as 0.1 µM HT causes a significant decrease in PS-exposing RBC, in addition to providing significant protection from the decrease in cell volume. Moreover, treatment of RBC with HT counters the influx of extracellular Ca2+ and completely restores ATP and glutathione content at 1 µM. Finally, under the same experimental conditions, HT exerts a protective effect on RBC morphological changes and microvescicle release, completely restoring the typical biconcave shape at 1 µM. CONCLUSION: Taken together, the findings reported in this paper point to a novel biological effect for HT in preventing programmed suicidal death in anucleated cells and indicate that prevention from LPA toxic effects may represent an additional mechanism responsible for the health-promoting effect of this dietary phenol which has been claimed, particularly related to cardiovascular diseases.


Subject(s)
Eryptosis/drug effects , Lysophospholipids/toxicity , Phenylethyl Alcohol/analogs & derivatives , Phosphatidylserines/pharmacology , Adenosine Triphosphate/metabolism , Calcium/metabolism , Cell Size/drug effects , Erythrocytes/cytology , Erythrocytes/drug effects , Erythrocytes/metabolism , Glutathione/metabolism , Humans , Phenylethyl Alcohol/pharmacology , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism
4.
Cell Physiol Biochem ; 45(4): 1472-1486, 2018.
Article in English | MEDLINE | ID: mdl-29466791

ABSTRACT

BACKGROUND/AIMS: Hypertrophic ligamentum flavum (LF) is a major cause of lumbar spinal stenosis. Our previous work showed that high levels of lysophosphatidic acid (LPA) expression are positively correlated with LF hypertrophy. This study aimed to further unveil how LPA regulates LF hypertrophy Methods: We studied LPAR1 expression in human LF cells using PCR and western blotting. Cell viability cell cycle, apoptosis rate and molecular mechanisms were assayed in LPAR1 knockdown or overexpression LF cells. LF hypertrophy and the molecular mechanism was confirmed in human samples and in in vivo studies. RESULTS: The expression of LPA and its receptor LPAR1 is significantly higher in tissues or cells harvested from hypertrophic LF compared to healthy controls. Moreover, LPA promoted LF cell proliferation by interacting with LPAR1. This conclusion is supported by the fact that depletion or overexpression of LPAR1 changed the effect of LPA on LF cell proliferation. LPA also inhibits apoptosis in LF cells through the receptor LPAR1. Importantly, we demonstrated that the LPA-LPAR1 interaction initiated Akt phosphorylation and determined cell proliferation and apoptosis. Our in vitro findings were supported by our in vivo evidence that lyophilized LPA significantly induced LF hypertrophy via the LPAR1-Akt signaling pathway. More importantly, targeted inhibition of LPAR1 by Ki16425 with a gel sponge implant effectively reduced LPA-associated LF hypertrophy. Taken together, these data indicate that LPA binds to the receptor LPAR1 to induce LF cell proliferation and inhibit apoptosis by activating AKT signaling cascades. Targeting this signaling cascade with Ki16425 is a potential therapeutic strategy for preventing LF hypertrophy. CONCLUSION: LPA-LPAR1-Akt activation is positively correlated with the proliferation and survival of LF cells. LPAR1 could be a target for new drugs and the development of new therapeutic methods for treating LF hypertrophy.


Subject(s)
Ligamentum Flavum/drug effects , Lysophospholipids/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Hypertrophy/chemically induced , Hypertrophy/prevention & control , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Ligamentum Flavum/cytology , Ligamentum Flavum/metabolism , Lumbar Vertebrae/abnormalities , Lumbar Vertebrae/diagnostic imaging , Lysophospholipids/analysis , Male , Phosphorylation/drug effects , Propionates/pharmacology , Propionates/therapeutic use , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics
5.
J Physiol ; 595(8): 2681-2698, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28176353

ABSTRACT

KEY POINTS: Lysophosphatidic acid (LPA) is an itch mediator, but not a pain mediator by a cheek injection model. Dorsal root ganglion neurons directly respond to LPA depending on transient receptor potential ankyrin 1 (TRPA1) and vanilloid 1 (TRPV1). LPA-induced itch-related behaviours are decreased in TRPA1-knockout (KO), TRPV1KO or TRPA1TRPV1 double KO mice. TRPA1 and TRPV1 channels are activated by intracellular LPA, but not by extracellular LPA following LPA5 receptor activation with an activity of Ca2+ -independent phospholipase A2 and phospholipase D. Intracellular LPA interaction sites of TRPA1 are KK672-673 and KR977-978 (K: lysine, R: arginine). ABSTRACT: Intractable and continuous itch sensations often accompany diseases such as atopic dermatitis, neurogenic lesions, uremia and cholestasis. Lysophosphatidic acid (LPA) is an itch mediator found in cholestatic itch patients and it induces acute itch and pain in experimental rodent models. However, the molecular mechanism by which LPA activates peripheral sensory neurons remains unknown. In this study, we used a cheek injection method in mice to reveal that LPA induced itch-related behaviours but not pain-related behaviours. The LPA-induced itch behaviour and cellular effects were dependent on transient receptor potential ankyrin 1 (TRPA1) and vanilloid 1 (TRPV1), which are important for itch signal transduction. We also found that, among the six LPA receptors, the LPA5 receptor had the greatest involvement in itching. Furthermore, we demonstrated that phospholipase D (PLD) plays a critical role downstream of LPA5 and that LPA directly and intracellularly activates TRPA1 and TRPV1. These results suggest a unique mechanism by which cytoplasmic LPA produced de novo could activate TRPA1 and TRPV1. We conclude that LPA-induced itch is mediated by LPA5 , PLD, TRPA1 and TRPV1 signalling, and thus targeting TRPA1, TRPV1 or PLD could be effective for cholestatic itch interventions.


Subject(s)
Lysophospholipids/toxicity , Phospholipase D/physiology , Pruritus/metabolism , Receptors, Lysophosphatidic Acid/physiology , TRPV Cation Channels/physiology , Transient Receptor Potential Channels/physiology , Animals , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pruritus/chemically induced , Signal Transduction/drug effects , Signal Transduction/physiology , TRPA1 Cation Channel
6.
J Neurosci ; 35(28): 10224-35, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26180199

ABSTRACT

Lysophosphatidic acid (LPA) is an extracellular lipid mediator involved in many physiological functions that signals through six known G-protein-coupled receptors (LPA1-LPA6). A wide range of LPA effects have been identified in the CNS, including neural progenitor cell physiology, astrocyte and microglia activation, neuronal cell death, axonal retraction, and development of neuropathic pain. However, little is known about the involvement of LPA in CNS pathologies. Herein, we demonstrate for the first time that LPA signaling via LPA1 contributes to secondary damage after spinal cord injury. LPA levels increase in the contused spinal cord parenchyma during the first 14 d. To model this potential contribution of LPA in the spinal cord, we injected LPA into the normal spinal cord, revealing that LPA induces microglia/macrophage activation and demyelination. Use of a selective LPA1 antagonist or mice lacking LPA1 linked receptor-mediated signaling to demyelination, which was in part mediated by microglia. Finally, we demonstrate that selective blockade of LPA1 after spinal cord injury results in reduced demyelination and improvement in locomotor recovery. Overall, these results support LPA-LPA1 signaling as a novel pathway that contributes to secondary damage after spinal cord contusion in mice and suggest that LPA1 antagonism might be useful for the treatment of acute spinal cord injury. SIGNIFICANCE STATEMENT: This study reveals that LPA signaling via LPA receptor type 1 activation causes demyelination and functional deficits after spinal cord injury.


Subject(s)
Demyelinating Diseases/etiology , Receptors, Lysophosphatidic Acid/metabolism , Spinal Cord Injuries/complications , Spinal Cord Injuries/pathology , Spinal Cord/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Demyelinating Diseases/drug therapy , Demyelinating Diseases/pathology , Disease Models, Animal , Evoked Potentials, Motor/drug effects , Evoked Potentials, Motor/genetics , Female , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/metabolism , Lysophospholipids/toxicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Microglia/ultrastructure , Motor Activity/drug effects , Motor Activity/genetics , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/ultrastructure , Receptors, Lysophosphatidic Acid/deficiency , Spinal Cord/drug effects , Spinal Cord Injuries/etiology , Time Factors
7.
J Neural Transm (Vienna) ; 122(3): 487-94, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25119538

ABSTRACT

Lysophosphatidic acid (LPA) is a potent bioactive lipid mediator with diverse biological properties. We previously found altered expression of the LPA-related genes in rodents after treatment with sertraline, which is widely used to treat anxiety disorders and depression. However, little is known about the behavioral effects of LPA. In the present study, we investigated the behavioral effects of intracerebroventricular injection of LPA in adult mice. LPA did not significantly affect spontaneous locomotor activity, suggesting that LPA does not induce hyperactivity, ataxia, or sedation. We next investigated the emotional effects of LPA via the hole-board test. LPA significantly increased the number of head-dips in a dose- and time-related manner. A significant induction of head-dip counts occurred 15 and 30 min after LPA administration. To clarify the involvement of LPA receptors, we examined the effect of the non-selective LPA1-4 receptor antagonist, 1-bromo-3(S)-hydroxy-4-(palmitoyloxy)butyl-phosphonate (BrP-LPA) co-administered with LPA. BrP-LPA dose-dependently inhibited LPA-induced head-dip counts. We next investigated anxiety-like behavior via the elevated plus-maze test. LPA significantly reduced the percentage of time spent in the open arms and BrP-LPA dose-dependently inhibited this anxiety-like behavior. In conclusion, LPA induced anxiety-like behavior in mice via LPA receptors. Our results suggest that LPA signaling plays an important role in regulating anxiety in mice.


Subject(s)
Anxiety/chemically induced , Anxiety/metabolism , Lysophospholipids/toxicity , Receptors, Lysophosphatidic Acid/metabolism , Analysis of Variance , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Lysophospholipids/pharmacology , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Reaction Time/drug effects , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Time Factors
8.
Mediators Inflamm ; 2015: 492659, 2015.
Article in English | MEDLINE | ID: mdl-26576074

ABSTRACT

Initial and recurrent stroke produces central nervous system (CNS) damage, involving neuroinflammation. Receptor-mediated S1P signaling can influence neuroinflammation and has been implicated in cerebral ischemia through effects on the immune system. However, S1P-mediated events also occur within the brain itself where its roles during stroke have been less well studied. Here we investigated the involvement of S1P signaling in initial and recurrent stroke by using a transient middle cerebral artery occlusion/reperfusion (M/R) model combined with analyses of S1P signaling. Gene expression for S1P receptors and involved enzymes was altered during M/R, supporting changes in S1P signaling. Direct S1P microinjection into the normal CNS induced neuroglial activation, implicating S1P-initiated neuroinflammatory responses that resembled CNS changes seen during initial M/R challenge. Moreover, S1P microinjection combined with M/R potentiated brain damage, approximating a model for recurrent stroke dependent on S1P and suggesting that reduction in S1P signaling could ameliorate stroke damage. Delivery of FTY720 that removes S1P signaling with chronic exposure reduced damage in both initial and S1P-potentiated M/R-challenged brain, while reducing stroke markers like TNF-α. These results implicate direct S1P CNS signaling in the etiology of initial and recurrent stroke that can be therapeutically accessed by S1P modulators acting within the brain.


Subject(s)
Brain Ischemia/prevention & control , Lysophospholipids/physiology , Receptors, Lysosphingolipid/antagonists & inhibitors , Sphingosine/analogs & derivatives , Stroke/prevention & control , Animals , Blood-Brain Barrier , Brain Ischemia/etiology , Fingolimod Hydrochloride/pharmacology , Lysophospholipids/toxicity , Male , Mice , Mice, Inbred ICR , Microinjections , Neuroglia/drug effects , Neuroglia/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Signal Transduction , Sphingosine/physiology , Sphingosine/toxicity , Stroke/etiology , Tumor Necrosis Factor-alpha/analysis
9.
J Neurosci ; 33(6): 2582-92, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23392686

ABSTRACT

The biolipid sphingosine-1-phosphate (S1P) is an essential modulator of innate immunity, cell migration, and wound healing. It is released locally upon acute tissue injury from endothelial cells and activated thrombocytes and, therefore, may give rise to acute post-traumatic pain sensation via a yet elusive molecular mechanism. We have used an interdisciplinary approach to address this question, and we find that intradermal injection of S1P induced significant licking and flinching behavior in wild-type mice and a dose-dependent flare reaction in human skin as a sign of acute activation of nociceptive nerve terminals. Notably, S1P evoked a small excitatory ionic current that resulted in nociceptor depolarization and action potential firing. This ionic current was preserved in "cation-free" solution and blocked by the nonspecific Cl(-) channel inhibitor niflumic acid and by preincubation with the G-protein inhibitor GDP-ß-S. Notably, S1P(3) receptor was detected in virtually all neurons in human and mouse DRG. In line with this finding, S1P-induced neuronal responses and spontaneous pain behavior in vivo were substantially reduced in S1P(3)(-/-) mice, whereas in control S1P(1) floxed (S1P(1)(fl/fl)) mice and mice with a nociceptor-specific deletion of S1P(1)(-/-) receptor (SNS-S1P(1)(-/-)), neither the S1P-induced responses in vitro nor the S1P-evoked pain-like behavior was altered. Therefore, these findings indicate that S1P evokes significant nociception via G-protein-dependent activation of an excitatory Cl(-) conductance that is largely mediated by S1P(3) receptors present in nociceptors, and point to these receptors as valuable therapeutic targets for post-traumatic pain.


Subject(s)
Lysophospholipids/toxicity , Pain Measurement/methods , Pain/metabolism , Receptors, Lysosphingolipid/physiology , Sphingosine/analogs & derivatives , Adult , Animals , Cells, Cultured , Double-Blind Method , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pain/chemically induced , Pain Measurement/drug effects , Sphingosine/toxicity
10.
Biochim Biophys Acta ; 1831(8): 1386-94, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23711961

ABSTRACT

Earlier, our study demonstrated that lysophosphatidic acid (LPA) receptor mediated cardiomyocyte hypertrophy. However, the subtype-specific functions for LPA1 and LPA3 receptors in LPA-induced hypertrophy have not been distinguished. Growing evidence indicates that microRNAs (miRNAs) are involved in the pathogenesis of cardiac hypertrophy by down-regulating target molecules. The present work therefore aimed at elucidating the functions mediated by different subtypes of LPA receptors and investigating the modulatory role of miRNAs during LPA induced hypertrophy. Experiments were done with cultured neonatal rat cardiomyocytes (NRCMs) exposed to LPA and we showed that knockdown of LPA1 by small interfering RNA (siRNA) enhanced LPA-induced cardiomyocyte hypertrophy, whereas LPA3 silencing repressed hypertrophy. miR-23a, a pro-hypertrophic miRNA, was up-regulated by LPA in cardiomyocytes and its down-regulation reduced LPA-induced cardiomyocyte hypertrophy. Importantly, luciferase reporter assay confirmed LPA1 to be a target of miR-23a, indicating that miR-23a is involved in mediating the LPA-induced cardiomyocyte hypertrophy by targeting LPA1. In addition, knockdown of LPA3, but not LPA1, eliminated miR-23a elevation induced by LPA. And PI3K inhibitor, LY294002, effectively prevented LPA-induced miR-23a expression in cardiomyocytes, suggesting that LPA might induce miR-23a elevation by activating LPA3 and PI3K/AKT pathway. These findings identified opposite subtype-specific functions for LPA1 and LPA3 in mediating cardiomyocyte hypertrophy and indicated LPA1 to be a target of miR-23a, which discloses a link between miR-23a and the LPA receptor signaling in cardiomyocyte hypertrophy.


Subject(s)
Cardiomegaly/metabolism , MicroRNAs/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction , Animals , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/pathology , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Lysophospholipids/toxicity , MicroRNAs/genetics , Morpholines/pharmacology , Muscle Proteins/genetics , Myocytes, Cardiac/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Lysophosphatidic Acid/genetics
11.
J Neurosci ; 32(33): 11330-42, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22895716

ABSTRACT

Interleukin (IL)-1ß and IL-18 play critical roles in the induction of chronic pain hypersensitivity. Their inactive forms are activated by caspase-1. However, little is known about the mechanism underlying the activation of pro-caspase-1. There is increasing evidence that cathepsin B (CatB), a typical lysosomal cysteine protease, is involved in the pro-caspase-1 activation and the subsequent maturation of IL-1ß and IL-18. In this context, CatB is considered to be an important molecular target to control chronic pain. However, no information is currently available about the role of CatB in chronic pain hypersensitivity. We herein show that CatB deficiency or the intrathecal administration of CA-074Me, a specific CatB inhibitor, significantly inhibited the induction of complete Freund's adjuvant-induced tactile allodynia in mice without affecting peripheral inflammation. In contrast, CatB deficiency did not affect the nerve injury-induced tactile allodynia. Furthermore, CatB deficiency or CA-074Me treatment significantly inhibited the maturation and secretion of IL-1ß and IL-18 by cultured microglia following treatment with the neuroactive glycoprotein chromogranin A (CGA), but not with ATP. Moreover, the IL-1ß expression in spinal microglia and the induction of tactile allodynia following the intrathecal administration of CGA depended on CatB, whereas those induced by the intrathecal administration of ATP or lysophosphatidic acid were CatB independent. These results strongly suggest that CatB is an essential enzyme for the induction of chronic inflammatory pain through its activation of pro-caspase-1, which subsequently induces the maturation and secretion of IL-1ß and IL-18 by spinal microglia. Therefore, CatB-specific inhibitors may represent a useful new strategy for treating inflammation-associated pain.


Subject(s)
Cathepsin B/metabolism , Chronic Pain/etiology , Chronic Pain/pathology , Inflammation/complications , Microglia/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Analysis of Variance , Animals , CD11b Antigen/metabolism , CD4 Antigens/metabolism , Calcium-Binding Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cathepsin B/deficiency , Cells, Cultured , Chromogranin A/administration & dosage , Chronic Pain/drug therapy , Chronic Pain/genetics , Cyclooxygenase 2/metabolism , Dipeptides/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Freund's Adjuvant/toxicity , Functional Laterality , Ganglia, Spinal/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Inflammation/chemically induced , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lysophospholipids/toxicity , Lysosomes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Microglia/pathology , Motor Activity/drug effects , Motor Activity/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Nerve Tissue Proteins/metabolism , Pain Threshold/drug effects , RNA, Small Interfering/metabolism , Spinal Cord/pathology , Transfection
12.
Cell Physiol Biochem ; 32(2): 253-63, 2013.
Article in English | MEDLINE | ID: mdl-23942151

ABSTRACT

BACKGROUND: Transcriptional co-activator with PDZ-binding motif (TAZ), a downstream effector of the Hippo pathway, has been reported to regulate organ size, tissue homeostasis, and tumorigenesis by acting as a transcriptional co-activator. Lysophosphatidic acid (LPA) is a bioactive lipid implicated in tumorigenesis and metastasis of ovarian cancer through activation of G protein-coupled receptors. However, the involvement of TAZ in LPA-induced tumorigenesis of ovarian cancer has not been elucidated. METHODS: In order to demonstrate the role of TAZ in LPA-stimulated tumorigenesis, the effects of LPA on TAZ expression and cell migration were determined by Western blotting and chemotaxis analyses in R182 human epithelial ovarian cancer cells. RESULTS AND CONCLUSION: Treatment of R182 cells with the LPA receptor inhibitor Ki16425 blocked LPA-induced cell migration. In addition, transfection of R182 cells with small interfering RNA specific for LPA receptor 1 resulted in abrogation of LPA-stimulated cell migration. LPA induced phosphorylation of ERK and p38 MAP kinase in R182 cells and pretreatment of cells with the MEK-ERK pathway inhibitor U0126, but not the p38 MAPK inhibitor SB202190, resulted in abrogation of LPA-induced cell migration. Pretreatment of R182 cells with U0126 attenuated LPA-induced mRNA levels of TAZ and its transcriptional target genes, such as CTGF and CYR61, without affecting phosphorylation level of YAP. These results suggest that MEK-ERK pathway plays a key role in LPA-induced cell migration and mRNA expression of TAZ in R182 cells, without affecting stability of TAZ protein. In addition, small interfering RNA-mediated silencing of TAZ expression attenuated LPA-stimulated migration of R182 cells. These results suggest that TAZ plays a key role in LPA-stimulated migration of epithelial ovarian cancer cells.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Lysophospholipids/toxicity , Butadienes/pharmacology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Isoxazoles/pharmacology , Neoplasms, Glandular and Epithelial/drug therapy , Nitriles/pharmacology , Ovarian Neoplasms/drug therapy , Propionates/pharmacology , Protein Stability/drug effects , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins
13.
Sci Rep ; 11(1): 658, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436724

ABSTRACT

Scratching is one of the most important behaviours in experimental animals because it can reflect itching and/or psychological stress. Here, we aimed to establish a novel method to detect scratching using deep neural network. Scratching was elicited by injecting a chemical pruritogen lysophosphatidic acid to the back of a mouse, and behaviour was recorded using a standard handy camera. Images showing differences between two consecutive frames in each video were generated, and each frame was manually labelled as showing scratching behaviour or not. Next, a convolutional recurrent neural network (CRNN), composed of sequential convolution, recurrent, and fully connected blocks, was constructed. The CRNN was trained using the manually labelled images and then evaluated for accuracy using a first-look dataset. Sensitivity and positive predictive rates reached 81.6% and 87.9%, respectively. The predicted number and durations of scratching events correlated with those of the human observation. The trained CRNN could also successfully detect scratching in the hapten-induced atopic dermatitis mouse model (sensitivity, 94.8%; positive predictive rate, 82.1%). In conclusion, we established a novel scratching detection method using CRNN and showed that it can be used to study disease models.


Subject(s)
Behavior, Animal/drug effects , Dermatitis, Atopic/diagnosis , Disease Models, Animal , Lysophospholipids/toxicity , Neural Networks, Computer , Pruritus/diagnosis , Animals , Dermatitis, Atopic/chemically induced , Female , Male , Mice , Mice, Inbred BALB C , Pruritus/chemically induced
14.
J Neurochem ; 113(4): 1002-11, 2010 May.
Article in English | MEDLINE | ID: mdl-20420580

ABSTRACT

Lysophosphatidic acid receptor (LPA(1)) signaling initiates neuropathic pain through demyelination of the dorsal root (DR). Although LPA is found to cause down-regulation of myelin proteins underlying demyelination, the detailed mechanism remains to be determined. In the present study, we found that a single intrathecal injection of LPA evoked a dose- and time-dependent down-regulation of myelin-associated glycoprotein (MAG) in the DR through LPA(1) receptor. A similar event was also observed in ex vivo DR cultures. Interestingly, LPA-induced down-regulation of MAG was significantly inhibited by calpain inhibitors (calpain inhibitor X, E-64 and E-64d) and LPA markedly induced calpain activation in the DR. The pre-treatment with calpain inhibitors attenuated LPA-induced neuropathic pain behaviors such as hyperalgesia and allodynia. Moreover, we found that sciatic nerve injury activates calpain activity in the DR in a LPA(1) receptor-dependent manner. The E-64d treatments significantly blocked nerve injury-induced MAG down-regulation and neuropathic pain. However, there was no significant calpain activation in the DR by complete Freund's adjuvant treatment, and E-64d failed to show anti-hyperalgesic effects in this inflammation model. The present study provides strong evidence that LPA-induced calpain activation plays a crucial role in the manifestation of neuropathic pain through MAG down-regulation in the DR.


Subject(s)
Calpain/metabolism , Demyelinating Diseases/metabolism , Myelin-Associated Glycoprotein/metabolism , Peripheral Nervous System Diseases/metabolism , Sensory Receptor Cells/metabolism , Spinal Nerve Roots/metabolism , Animals , Cysteine Proteinase Inhibitors/pharmacology , Demyelinating Diseases/etiology , Demyelinating Diseases/physiopathology , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Activation/physiology , Leucine/analogs & derivatives , Leucine/pharmacology , Lysophospholipids/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurotoxins/toxicity , Peripheral Nervous System Diseases/physiopathology , Receptors, Lysophosphatidic Acid/drug effects , Receptors, Lysophosphatidic Acid/metabolism , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/physiopathology , Sensory Receptor Cells/pathology , Spinal Nerve Roots/pathology
15.
J Exp Med ; 199(6): 763-74, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15007093

ABSTRACT

Neointimal lesions are characterized by accumulation of cells within the arterial wall and are a prelude to atherosclerotic disease. Here we report that a brief exposure to either alkyl ether analogs of the growth factor-like phospholipid lysophosphatidic acid (LPA), products generated during the oxidative modification of low density lipoprotein, or to unsaturated acyl forms of LPA induce progressive formation of neointima in vivo in a rat carotid artery model. This effect is completely inhibited by the peroxisome proliferator-activated receptor (PPAR)gamma antagonist GW9662 and mimicked by PPARgamma agonists Rosiglitazone and 1-O-hexadecyl-2-azeleoyl-phosphatidylcholine. In contrast, stearoyl-oxovaleryl phosphatidylcholine, a PPARalpha agonist and polypeptide epidermal growth factor, platelet-derived growth factor, and vascular endothelial growth factor failed to elicit neointima. The structure-activity relationship for neointima induction by LPA analogs in vivo is identical to that of PPARgamma activation in vitro and disparate from that of LPA G protein-coupled receptor activation. Neointima-inducing LPA analogs up-regulated the CD36 scavenger receptor in vitro and in vivo and elicited dedifferentiation of cultured vascular smooth muscle cells that was prevented by GW9662. These results suggest that selected LPA analogs are important novel endogenous PPARgamma ligands capable of mediating vascular remodeling and that activation of the nuclear transcription factor PPARgamma is both necessary and sufficient for neointima formation by components of oxidized low density lipoprotein.


Subject(s)
Anilides/pharmacology , Arteriosclerosis/chemically induced , Carotid Artery Diseases/chemically induced , Lysophospholipids/toxicity , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Analysis of Variance , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Calmodulin-Binding Proteins/metabolism , Cells, Cultured , DNA Primers , Disease Models, Animal , Growth Substances/metabolism , Ligands , Lipoproteins, LDL/metabolism , Male , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Structure-Activity Relationship , Thiazolidinediones/toxicity , Time Factors , Transcription Factors/agonists
16.
Pain ; 160(1): 269-278, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30211781

ABSTRACT

Joint neuropathic pain occurs in a subset of arthritis patients, and lysophosphatidic acid (LPA) has been implicated as a mediator of joint neuropathy. The mechanism by which LPA promotes neuropathic pain is unknown but may be related to altered signalling of the voltage-gated sodium channel Nav1.8 located on nociceptors. Because arthritis and neuropathic pain are more prevalent in females, this study aimed to explore potential sex differences in the development of LPA-induced joint neuropathy and whether Nav1.8 played a role in the associated neuropathic pain. Joint neuropathy was induced in male and female Wistar rats (179-284 g) by intra-articular injection of 50-µg LPA. Pain behaviour was assessed over 21 days using von Frey hair algesiometry. On day 21, electrophysiological recordings of joint primary afferents were conducted to measure peripheral sensitisation. Saphenous nerve morphology and expression of the nerve-damage marker ATF3 and Nav1.8 in ipsilateral dorsal root ganglions were compared on the basis of sex. The analgesic properties of the selective Nav1.8 antagonist A-803467 was determined in pain behaviour and electrophysiology experiments. Females developed more severe mechanical allodynia than males after LPA treatment. Lysophosphatidic acid caused more pronounced demyelination of the saphenous nerve in females, but no sex differences were observed in the expression of ATF3 or Nav1.8 in dorsal root ganglion neurones. Blockade of Nav1.8 channels with A-803467 resulted in a decrease in joint mechanosensitivity and secondary allodynia with females exhibiting a greater response. These findings suggest that LPA has sex-specific effects on joint neuropathy and Nav1.8 gating, which should be considered when treating neuropathic arthritis patients.


Subject(s)
Arthralgia/chemically induced , Knee Joint/pathology , Lysophospholipids/toxicity , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Sex Characteristics , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Aniline Compounds/pharmacology , Animals , Disease Models, Animal , Exploratory Behavior , Female , Furans/pharmacology , Hyperalgesia/chemically induced , Male , NAV1.8 Voltage-Gated Sodium Channel/genetics , Pain Measurement , Rats , Rats, Wistar , Stilbamidines/metabolism
17.
J Vis Exp ; (151)2019 09 27.
Article in English | MEDLINE | ID: mdl-31609319

ABSTRACT

Itch was defined as "an unpleasant cutaneous sensation that provokes a desire to scratch" by Rothman in 1941. In mouse models, scratch bouts are typically counted to evaluate itch induced by pruritogens. However, previous reports have shown that algesic substances also induce scratching behaviors in a mouse neck injection model, which is the most common test used for scratching behaviors. This finding makes it difficult to study itch in mice.  In contrast, capsaicin, a common algogen, reduced scratching behaviors in some neck injection experiments. Therefore, the effect of pain on scratching behaviors remains unclear. It is thus necessary to develop a method to concurrently investigate itch and pain sensation using behavioral tests. Here, a cheek injection model is introduced which can be used to simultaneously measure pain- and itch-related behaviors. In this model, pruritogens induce scratching behaviors while algesic substances induce wiping behaviors. Using this model, lysophosphatidic acid (LPA), an itch mediator found in cholestatic patients with itch, is shown to exclusively induce itch but not pain. However, in mouse models, LPA has been reported to be both a pruritogen and an algogen. Investigation into the effects of LPA in a mouse cheek injection model showed that LPA only induced scratching, but not wiping behaviors. This indicates that LPA acts as a pruritogen similarly in mice and humans, and demonstrates the utility of a cheek injection model for itch research.


Subject(s)
Behavior, Animal/drug effects , Capsaicin/administration & dosage , Cheek , Lysophospholipids/toxicity , Pain/psychology , Pruritus/psychology , Animals , Antipruritics/administration & dosage , Disease Models, Animal , Injections , Male , Mice , Mice, Inbred C57BL , Pain/chemically induced , Pain/drug therapy , Pain/pathology , Pruritus/chemically induced , Pruritus/drug therapy , Pruritus/pathology
18.
Sci Adv ; 5(10): eaax2011, 2019 10.
Article in English | MEDLINE | ID: mdl-31633020

ABSTRACT

Posthemorrhagic hydrocephalus (PHH) in premature infants is a common neurological disorder treated with invasive neurosurgical interventions. Patients with PHH lack effective therapeutic interventions and suffer chronic comorbidities. Here, we report a murine lysophosphatidic acid (LPA)-induced postnatal PHH model that maps neurodevelopmentally to premature infants, a clinically accessible high-risk population, and demonstrates ventriculomegaly with increased intracranial pressure. Administration of LPA, a blood-borne signaling lipid, acutely disrupted the ependymal cells that generate CSF flow, which was followed by cell death, phagocytosis, and ventricular surface denudation. This mechanism is distinct from a previously reported fetal model that induces PHH through developmental alterations. Analyses of LPA receptor-null mice identified LPA1 and LPA3 as key mediators of PHH. Pharmacological blockade of LPA1 prevented PHH in LPA-injected animals, supporting the medical tractability of LPA receptor antagonists in preventing PHH and negative CNS sequelae in premature infants.


Subject(s)
Infant, Premature, Diseases/pathology , Receptors, Lysophosphatidic Acid/metabolism , Animals , Animals, Newborn , Apoptosis , Brain/metabolism , Brain/pathology , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Ependyma/cytology , Ependyma/metabolism , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Infant, Premature, Diseases/chemically induced , Infant, Premature, Diseases/prevention & control , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Lysophospholipids/toxicity , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Phagocytosis , Propionates/pharmacology , Propionates/therapeutic use , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics
19.
Neurochem Int ; 129: 104515, 2019 10.
Article in English | MEDLINE | ID: mdl-31369778

ABSTRACT

Lysophosphatidic acid (LPA) is a glycerophospholipid that can be detected in serum, saliva and cerebrospinal fluid. However, the effect of LPA on neuronal death and survival has not been fully determined. In the present study, we investigated the potential neurotoxic effect of LPA in primary cultured cortical neurons. Treatment with LPA (0.5, 1 and 5 µM) markedly decreased neuronal viability, increased lactate dehydrogenase (LDH) release and promoted apoptosis in cortical neurons. The results of western blot showed that LPA increased the expression of endoplasmic reticulum (ER) stress associated factors, and the protein misfolding inhibitor 4-phenylbutyric acid (4-PBA) attenuated LPA-induced toxicity. In addition, treatment with LPA did not alter the expression and distribution of Homer1 in cortical neurons. The protein levels of metabotropic glutamate receptor 1 (mGluR1), but not metabotropic glutamate receptor 5 (mGluR5), were significantly increased by LPA at 12 and 24 h after treatment. Knockdown of Homer1 using specific siRNA partially prevented the LPA-induced neurotoxicity and ER stress. Furthermore, the results of Ca2+ imaging showed that treatment with LPA induced intracellular Ca2+ release, which could be partially prevented by 4-PBA and downregulation of Homer1. The LPA-induced intracellular Ca2+ release was associated with ER Ca2+ release through the Homer1-mGluR1 pathway. In summary, our results showed that LPA treatment induced ER stress and apoptosis in cortical neurons, and its neurotoxicity was partially mediated by Ca2+ release from the ER via the Homer1/mGluR1 pathway.


Subject(s)
Calcium Signaling/drug effects , Endoplasmic Reticulum Stress/drug effects , Homer Scaffolding Proteins/physiology , Lysophospholipids/toxicity , Nerve Tissue Proteins/physiology , Neurons/drug effects , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Down-Regulation , Female , Homer Scaffolding Proteins/antagonists & inhibitors , Homer Scaffolding Proteins/biosynthesis , Homer Scaffolding Proteins/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/metabolism , Phenylbutyrates/pharmacology , Primary Cell Culture , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptor, Metabotropic Glutamate 5/genetics , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/genetics
20.
Neuropharmacology ; 155: 150-161, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31145906

ABSTRACT

Lysophosphatidic acid (LPA) is a bioactive lipid mediator of inflammation that binds to its specific cell surface G protein coupled receptors (LPA1-6). It is reported that LPA induced cell apoptosis by targeting LPA1, while LPA1 blockade eliminated LPS-induced production of peritoneal neutrophil chemokines and cytokines. Previous studies have shown that Saikosaponin-d (SSd) mitigated depressive-like behaviors in rats exposed to chronic unpredictable mild stress (CUMS), as well as corticosterone-induced apoptosis in PC12 cells. The present study explored the role of SSd during modulating LPA1 mediated neuronal apoptosis in LPS-stimulated mice. The phenomenon that SSd alleviated LPS-induced depressive-like behaviors were observed by open field test (OPT), forced swim test (FST) and tail suspension test (TST). SSd inhibited the protein expression of LPA1 both in the CA1 and CA3 region of the hippocampus. Moreover, SSd significantly decreased the levels of RhoA, ROCK2, p-p38, p-ERK, p-p65, p-IκBα in LPS-stimulated mice as well as in LPA-stimulated SH-SY5Y cells. Additionally, SSd significantly decreased the expression of LPA1 and the degree of neuronal apoptosis in SH-SY5Y cells which were co-cultured with LPS-stimulated BV2 microglia. These results suggested that SSd improved LPS-induced depressive-like behaviors in mice and suppressed neuronal apoptosis by regulating LPA1/RhoA/ROCK2 signaling pathway.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Apoptosis/physiology , Depression/metabolism , Neurons/metabolism , Oleanolic Acid/analogs & derivatives , Receptors, Lysophosphatidic Acid/metabolism , Saponins/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Coculture Techniques , Depression/chemically induced , Depression/drug therapy , Dose-Response Relationship, Drug , Humans , Lysophospholipids/metabolism , Lysophospholipids/toxicity , Mice , Mice, Inbred ICR , Neurons/drug effects , Oleanolic Acid/metabolism , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Random Allocation , Saponins/pharmacology , Saponins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL