Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Pharmacol Exp Ther ; 383(2): 172-181, 2022 11.
Article in English | MEDLINE | ID: mdl-36116795

ABSTRACT

Emerging evidence implicates the G-protein coupled receptor (GPCR) GPR183 in the development of neuropathic pain. Further investigation of the signaling pathways downstream of GPR183 is needed to support the development of GPR183 antagonists as analgesics. In rodents, intrathecal injection of its ligand, 7α,25-dihydroxycholesterol (7α,25-OHC), causes time-dependent development of mechano-and cold- allodynia (behavioral hypersensitivity). These effects are blocked by the selective small molecule GPR183 antagonist, SAE-14. However, the molecular mechanisms engaged downstream of GPR183 in the spinal cord are not known. Here, we show that 7α,25-OHC-induced behavioral hypersensitivity is Gα i dependent, but not ß-arrestin 2-dependent. Non-biased transcriptomic analyses of dorsal-horn spinal cord (DH-SC) tissues harvested at the time of peak hypersensitivity implicate potential contributions of mitogen-activated protein kinase (MAPK) and nuclear factor κB (NF-κB). In support, we found that the development of 7α,25-OHC/GPR183-induced mechano-allodynia was associated with significant activation of MAPKs (extracellular signal-regulated kinase [ERK], p38) and redox-sensitive transcription factors (NF-κB) and increased formation of inflammatory and neuroexcitatory cytokines. SAE-14 blocked these effects and behavioral hypersensitivity. Our findings provide novel mechanistic insight into how GPR183 signaling in the spinal cord produces hypersensitivity through MAPK and NF-κB activation. SIGNIFICANCE STATEMENT: Using a multi-disciplinary approach, we have characterized the molecular mechanisms underpinning 7α,25-OHC/GPR183-induced hypersensitivity in mice. Intrathecal injections of the GPR183 agonist 7α,25-OHC induce behavioral hypersensitivity, and these effects are blocked by the selective GPR183 antagonist SAE-14. We found that 7α,25-OHC-induced allodynia is dependent on MAPK and NF-κB signaling pathways and results in an increase in pro-inflammatory cytokine expression. This study provides a first insight into how GPR183 signaling in the spinal cord is pronociceptive.


Subject(s)
Hyperalgesia , NF-kappa B , Animals , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hyperalgesia/chemically induced , Ligands , Mice , NF-kappa B/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Proc Natl Acad Sci U S A ; 116(21): 10557-10562, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31068460

ABSTRACT

Neuropathic pain afflicts millions of individuals and represents a major health problem for which there is limited effective and safe therapy. Emerging literature links altered sphingolipid metabolism to nociceptive processing. However, the neuropharmacology of sphingolipid signaling in the central nervous system in the context of chronic pain remains largely unexplored and controversial. We now provide evidence that sphingosine-1-phosphate (S1P) generated in the dorsal horn of the spinal cord in response to nerve injury drives neuropathic pain by selectively activating the S1P receptor subtype 1 (S1PR1) in astrocytes. Accordingly, genetic and pharmacological inhibition of S1PR1 with multiple antagonists in distinct chemical classes, but not agonists, attenuated and even reversed neuropathic pain in rodents of both sexes and in two models of traumatic nerve injury. These S1PR1 antagonists retained their ability to inhibit neuropathic pain during sustained drug administration, and their effects were independent of endogenous opioid circuits. Moreover, mice with astrocyte-specific knockout of S1pr1 did not develop neuropathic pain following nerve injury, thereby identifying astrocytes as the primary cellular substrate of S1PR1 activity. On a molecular level, the beneficial reductions in neuropathic pain resulting from S1PR1 inhibition were driven by interleukin 10 (IL-10), a potent neuroprotective and anti-inflammatory cytokine. Collectively, our results provide fundamental neurobiological insights that identify the cellular and molecular mechanisms engaged by the S1PR1 axis in neuropathic pain and establish S1PR1 as a target for therapeutic intervention with S1PR1 antagonists as a class of nonnarcotic analgesics.


Subject(s)
Astrocytes/metabolism , Neuralgia/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sulfones/therapeutic use , Triazoles/therapeutic use , Animals , Drug Evaluation, Preclinical , Female , Interleukin-10/metabolism , Male , Mice , Neuralgia/drug therapy , Neuralgia/etiology , Rats, Sprague-Dawley , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Sulfones/pharmacology , Triazoles/pharmacology
3.
J Neuroinflammation ; 17(1): 314, 2020 Oct 22.
Article in English | MEDLINE | ID: mdl-33092620

ABSTRACT

Opioid therapies for chronic pain are undermined by many adverse side effects that reduce their efficacy and lead to dependence, abuse, reduced quality of life, and even death. We have recently reported that sphingosine-1-phosphate (S1P) 1 receptor (S1PR1) antagonists block the development of morphine-induced hyperalgesia and analgesic tolerance. However, the impact of S1PR1 antagonists on other undesirable side effects of opioids, such as opioid-induced dependence, remains unknown. Here, we demonstrate that naloxone-precipitated morphine withdrawal in mice altered de novo sphingolipid metabolism in the dorsal horn of the spinal cord and increased S1P that accompanied the manifestation of several withdrawal behaviors. Blocking de novo sphingolipid metabolism with intrathecal administration of myriocin, an inhibitor of serine palmitoyltransferase, blocked naloxone-precipitated withdrawal. Noteworthy, we found that competitive (NIBR-15) and functional (FTY720) S1PR1 antagonists attenuated withdrawal behaviors in mice. Mechanistically, at the level of the spinal cord, naloxone-precipitated withdrawal was associated with increased glial activity and formation of the potent inflammatory/neuroexcitatory cytokine interleukin-1ß (IL-1ß); these events were attenuated by S1PR1 antagonists. These results provide the first molecular insight for the role of the S1P/S1PR1 axis during opioid withdrawal. Our data identify S1PR1 antagonists as potential therapeutics to mitigate opioid-induced dependence and support repurposing the S1PR1 functional antagonist FTY720, which is FDA-approved for multiple sclerosis, as an opioid adjunct.


Subject(s)
Analgesics, Opioid/adverse effects , Central Nervous System/metabolism , Morphine/adverse effects , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Sphingosine-1-Phosphate Receptors/metabolism , Substance Withdrawal Syndrome/metabolism , Animals , Central Nervous System/drug effects , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Male , Mice , Mice, Inbred BALB C , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Rodentia , Substance Withdrawal Syndrome/drug therapy
4.
J Pharmacol Exp Ther ; 375(2): 367-375, 2020 11.
Article in English | MEDLINE | ID: mdl-32913007

ABSTRACT

Neuropathic pain is a debilitating public health concern for which novel non-narcotic therapeutic targets are desperately needed. Using unbiased transcriptomic screening of the dorsal horn spinal cord after nerve injury we have identified that Gpr183 (Epstein-Barr virus-induced gene 2) is upregulated after chronic constriction injury (CCI) in rats. GPR183 is a chemotactic receptor known for its role in the maturation of B cells, and the endogenous ligand is the oxysterol 7α,25-dihydroxycholesterol (7α,25-OHC). The role of GPR183 in the central nervous system is not well characterized, and its role in pain is unknown. The profile of commercially available probes for GPR183 limits their use as pharmacological tools to dissect the roles of this receptor in pathophysiological settings. Using in silico modeling, we have screened a library of 5 million compounds to identify several novel small-molecule antagonists of GPR183 with nanomolar potency. These compounds are able to antagonize 7α,25-OHC-induced calcium mobilization in vitro with IC50 values below 50 nM. In vivo intrathecal injections of these antagonists during peak pain after CCI surgery reversed allodynia in male and female mice. Acute intrathecal injection of the GPR183 ligand 7α,25-OHC in naïve mice induced dose-dependent allodynia. Importantly, this effect was blocked using our novel GPR183 antagonists, suggesting spinal GPR183 activation as pronociceptive. These studies are the first to reveal a role for GPR183 in neuropathic pain and identify this receptor as a potential target for therapeutic intervention. SIGNIFICANCE STATEMENT: We have identified several novel GPR183 antagonists with nanomolar potency. Using these antagonists, we have demonstrated that GPR183 signaling in the spinal cord is pronociceptive. These studies are the first to reveal a role for GPR183 in neuropathic pain and identify it as a potential target for therapeutic intervention.


Subject(s)
Neuralgia/metabolism , Oxysterols/metabolism , Receptors, G-Protein-Coupled/metabolism , Spinal Cord/metabolism , Animals , Female , HL-60 Cells , Humans , Male , Mice , Neuralgia/drug therapy , Neuralgia/pathology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction , Spinal Cord/pathology
5.
J Pharmacol Exp Ther ; 374(2): 331-341, 2020 08.
Article in English | MEDLINE | ID: mdl-32434943

ABSTRACT

Treating chronic pain by using opioids, such as morphine, is hampered by the development of opioid-induced hyperalgesia (OIH; increased pain sensitivity), antinociceptive tolerance, and withdrawal, which can contribute to dependence and abuse. In the central nervous system, the purine nucleoside adenosine has been implicated in beneficial and detrimental actions of morphine, but the extent of their interaction remains poorly understood. Here, we demonstrate that morphine-induced OIH and antinociceptive tolerance in rats is associated with a twofold increase in adenosine kinase (ADK) expression in the dorsal horn of the spinal cord. Blocking ADK activity in the spinal cord provided greater than 90% attenuation of OIH and antinociceptive tolerance through A3 adenosine receptor (A3AR) signaling. Supplementing adenosine signaling with selective A3AR agonists blocked OIH and antinociceptive tolerance in rodents of both sexes. Engagement of A3AR in the spinal cord with an ADK inhibitor or A3AR agonist was associated with reduced dorsal horn of the spinal cord expression of the NOD-like receptor pyrin domain-containing 3 (60%-75%), cleaved caspase 1 (40%-60%), interleukin (IL)-1ß (76%-80%), and tumor necrosis factor (50%-60%). In contrast, the neuroinhibitory and anti-inflammatory cytokine IL-10 increased twofold. In mice, A3AR agonists prevented the development of tolerance in a model of neuropathic pain and reduced naloxone-dependent withdrawal behaviors by greater than 50%. These findings suggest A3AR-dependent adenosine signaling is compromised during sustained morphine to allow the development of morphine-induced adverse effects. These findings raise the intriguing possibility that A3AR agonists may be useful adjunct to opioids to manage their unwanted effects. SIGNIFICANCE STATEMENT: The development of hyperalgesia and antinociceptive tolerance during prolonged opioid use are noteworthy opioid-induced adverse effects that reduce opioid efficacy for treating chronic pain and increase the risk of dependence and abuse. We report that in rodents, these adverse effects are due to reduced adenosine signaling at the A3AR, resulting in NOD-like receptor pyrin domain-containing 3-interleukin-1ß neuroinflammation in spinal cord. These effects are attenuated by A3AR agonists, suggesting that A3AR may be a target for therapeutic intervention with selective A3AR agonist as opioid adjuncts.


Subject(s)
Analgesics/adverse effects , Drug Tolerance , Hyperalgesia/chemically induced , Morphine/adverse effects , Receptor, Adenosine A3/metabolism , Signal Transduction/drug effects , Substance Withdrawal Syndrome/etiology , Adenosine/metabolism , Animals , Female , Hyperalgesia/metabolism , Interleukin-10/metabolism , Interleukin-1beta/biosynthesis , Male , Rats , Rats, Sprague-Dawley , Time Factors
6.
Purinergic Signal ; 16(3): 367-377, 2020 09.
Article in English | MEDLINE | ID: mdl-32720036

ABSTRACT

Prodrugs (MRS7422, MRS7476) of highly selective A3 adenosine receptor (AR) agonists Cl-IB-MECA and MRS5698, respectively, were synthesized by succinylation of the 2' and 3' hydroxyl groups, and the parent, active drug was shown to be readily liberated upon incubation with liver esterases. The prodrug MRS7476 had greatly increased aqueous solubility compared with parent MRS5698 and was fully efficacious and with a longer duration than MRS7422 in reversing mouse neuropathic pain (chronic constriction injury model, 3 µmol/kg, p.o.), a known A3AR effect. MRS7476 (5 mg/kg, p.o., twice daily) was found to protect against non-alcoholic steatohepatitis (NASH) in the STAM mouse model, indicated by the NAFLD activity score. Hepatocyte ballooning, IL-10 production, and liver histology were significantly normalized in the MRS7476-treated mice, but not liver fibrosis (no change in ACTA2 levels) or inflammation. Hepatic expression of ADORA3 in human NAFLD patients was 1.9-fold lower compared to normal controls. Adora3 expression determined by qPCR in primary mouse liver was associated with the stellate cells, and its mouse full body A3AR knockout worsened liver markers of inflammation and steatosis. Thus, we have introduced a reversible prodrug strategy that enables water solubility and in vivo activity of masked A3AR agonists in models of two disease conditions.


Subject(s)
Adenosine A3 Receptor Agonists/chemistry , Drug Design , Neuralgia/drug therapy , Prodrugs/chemistry , Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine/therapeutic use , Adenosine A3 Receptor Agonists/therapeutic use , Animals , Disease Models, Animal , Inflammation/prevention & control , Mice , Non-alcoholic Fatty Liver Disease/prevention & control , Prodrugs/therapeutic use
7.
J Neurosci ; 35(15): 6057-67, 2015 04 15.
Article in English | MEDLINE | ID: mdl-25878279

ABSTRACT

More than 1.5 billion people worldwide suffer from chronic pain, yet current treatment strategies often lack efficacy or have deleterious side effects in patients. Adenosine is an inhibitory neuromodulator that was previously thought to mediate antinociception through the A1 and A2A receptor subtypes. We have since demonstrated that A3AR agonists have potent analgesic actions in preclinical rodent models of neuropathic pain and that A3AR analgesia is independent of adenosine A1 or A2A unwanted effects. Herein, we explored the contribution of the GABA inhibitory system to A3AR-mediated analgesia using well-characterized mouse and rat models of chronic constriction injury (CCI)-induced neuropathic pain. The deregulation of GABA signaling in pathophysiological pain states is well established: GABA signaling can be hampered by a reduction in extracellular GABA synthesis by GAD65 and enhanced extracellular GABA reuptake via the GABA transporter, GAT-1. In neuropathic pain, GABAAR-mediated signaling can be further disrupted by the loss of the KCC2 chloride anion gradient. Here, we demonstrate that A3AR agonists (IB-MECA and MRS5698) reverse neuropathic pain via a spinal mechanism of action that modulates GABA activity. Spinal administration of the GABAA antagonist, bicuculline, disrupted A3AR-mediated analgesia. Furthermore, A3AR-mediated analgesia was associated with reductions in CCI-related GAD65 and GAT-1 serine dephosphorylation as well as an enhancement of KCC2 serine phosphorylation and activity. Our results suggest that A3AR-mediated reversal of neuropathic pain increases modulation of GABA inhibitory neurotransmission both directly and indirectly through protection of KCC2 function, underscoring the unique utility of A3AR agonists in chronic pain.


Subject(s)
Adenosine A3 Receptor Agonists/therapeutic use , Analgesics/therapeutic use , Sciatica/drug therapy , Signal Transduction/drug effects , Symporters/metabolism , gamma-Aminobutyric Acid/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/therapeutic use , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Male , Mice , Pain Threshold/drug effects , Pyridines/pharmacology , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Sciatica/complications , Signal Transduction/physiology , Spinal Nerve Roots/metabolism , Spinal Nerve Roots/pathology , Thiazoles/pharmacology , Thiazoles/therapeutic use , Thioglycolates/pharmacology , Thioglycolates/therapeutic use , K Cl- Cotransporters
8.
J Pharmacol Exp Ther ; 356(2): 474-82, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26606937

ABSTRACT

Adenosine can induce hypothermia, as previously demonstrated for adenosine A1 receptor (A1AR) agonists. Here we use the potent, specific A3AR agonists MRS5698, MRS5841, and MRS5980 to show that adenosine also induces hypothermia via the A3AR. The hypothermic effect of A3AR agonists is independent of A1AR activation, as the effect was fully intact in mice lacking A1AR but abolished in mice lacking A3AR. A3AR agonist-induced hypothermia was attenuated by mast cell granule depletion, demonstrating that the A3AR hypothermia is mediated, at least in part, via mast cells. Central agonist dosing had no clear hypothermic effect, whereas peripheral dosing of a non-brain-penetrant agonist caused hypothermia, suggesting that peripheral A3AR-expressing cells drive the hypothermia. Mast cells release histamine, and blocking central histamine H1 (but not H2 or H4) receptors prevented the hypothermia. The hypothermia was preceded by hypometabolism and mice with hypothermia preferred a cooler environmental temperature, demonstrating that the hypothermic state is a coordinated physiologic response with a reduced body temperature set point. Importantly, hypothermia is not required for the analgesic effects of A3AR agonists, which occur with lower agonist doses. These results support a mechanistic model for hypothermia in which A3AR agonists act on peripheral mast cells, causing histamine release, which stimulates central histamine H1 receptors to induce hypothermia. This mechanism suggests that A3AR agonists will probably not be useful for clinical induction of hypothermia.


Subject(s)
Hypothermia/metabolism , Receptor, Adenosine A3/metabolism , Receptors, Histamine H1/metabolism , Adenosine A3 Receptor Agonists/pharmacology , Animals , Dose-Response Relationship, Drug , Hypothermia/chemically induced , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred C57BL
9.
Brain ; 138(Pt 1): 28-35, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25414036

ABSTRACT

Chronic pain is a global burden that promotes disability and unnecessary suffering. To date, efficacious treatment of chronic pain has not been achieved. Thus, new therapeutic targets are needed. Here, we demonstrate that increasing endogenous adenosine levels through selective adenosine kinase inhibition produces powerful analgesic effects in rodent models of experimental neuropathic pain through the A3 adenosine receptor (A3AR, now known as ADORA3) signalling pathway. Similar results were obtained by the administration of a novel and highly selective A3AR agonist. These effects were prevented by blockade of spinal and supraspinal A3AR, lost in A3AR knock-out mice, and independent of opioid and endocannabinoid mechanisms. A3AR activation also relieved non-evoked spontaneous pain behaviours without promoting analgesic tolerance or inherent reward. Further examination revealed that A3AR activation reduced spinal cord pain processing by decreasing the excitability of spinal wide dynamic range neurons and producing supraspinal inhibition of spinal nociception through activation of serotonergic and noradrenergic bulbospinal circuits. Critically, engaging the A3AR mechanism did not alter nociceptive thresholds in non-neuropathy animals and therefore produced selective alleviation of persistent neuropathic pain states. These studies reveal A3AR activation by adenosine as an endogenous anti-nociceptive pathway and support the development of A3AR agonists as novel therapeutics to treat chronic pain.


Subject(s)
Neuralgia/metabolism , Neurons/metabolism , Receptor, Adenosine A3/metabolism , Spinal Cord/metabolism , Adenosine/pharmacology , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hyperalgesia/diagnosis , Hyperalgesia/physiopathology , Male , Medulla Oblongata/drug effects , Medulla Oblongata/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morpholines/pharmacology , Morpholines/therapeutic use , Naloxone/administration & dosage , Neuralgia/drug therapy , Neuralgia/genetics , Neuralgia/pathology , Neurons/drug effects , Pain Measurement , Pain Threshold/drug effects , Purinergic P1 Receptor Antagonists/pharmacology , Pyridines/pharmacology , Pyridines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A3/genetics , Spinal Cord/drug effects , Spinal Cord/pathology , Time Factors
10.
J Biol Chem ; 289(30): 21082-97, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24876379

ABSTRACT

The ceramide-sphingosine 1-phosphate (S1P) rheostat is important in regulating cell fate. Several chemotherapeutic agents, including paclitaxel (Taxol), involve pro-apoptotic ceramide in their anticancer effects. The ceramide-to-S1P pathway is also implicated in the development of pain, raising the intriguing possibility that these sphingolipids may contribute to chemotherapy- induced painful peripheral neuropathy, which can be a critical dose-limiting side effect of many widely used chemotherapeutic agents.We demonstrate that the development of paclitaxel-induced neuropathic pain was associated with ceramide and S1P formation in the spinal dorsal horn that corresponded with the engagement of S1P receptor subtype 1 (S1PR(1))- dependent neuroinflammatory processes as follows: activation of redox-sensitive transcription factors (NFκB) and MAPKs (ERK and p38) as well as enhanced formation of pro-inflammatory and neuroexcitatory cytokines (TNF-α and IL-1ß). Intrathecal delivery of the S1PR1 antagonist W146 reduced these neuroinflammatory processes but increased IL-10 and IL-4, potent anti-inflammatory/ neuroprotective cytokines. Additionally, spinal W146 reversed established neuropathic pain. Noteworthy, systemic administration of the S1PR1 modulator FTY720 (Food and Drug Administration- approved for multiple sclerosis) attenuated the activation of these neuroinflammatory processes and abrogated neuropathic pain without altering anticancer properties of paclitaxel and with beneficial effects extended to oxaliplatin. Similar effects were observed with other structurally and chemically unrelated S1PR1 modulators (ponesimod and CYM-5442) and S1PR1 antagonists (NIBR-14/15) but not S1PR1 agonists (SEW2871). Our findings identify for the first time the S1P/S1PR1 axis as a promising molecular and therapeutic target in chemotherapy-induced painful peripheral neuropathy, establish a mechanistic insight into the biomolecular signaling pathways, and provide the rationale for the clinical evaluation of FTY720 in chronic pain patients.


Subject(s)
Antineoplastic Agents, Phytogenic/adverse effects , Neuralgia/chemically induced , Neuralgia/enzymology , Paclitaxel/adverse effects , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , Anilides/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cytokines/metabolism , Enzyme Activation/drug effects , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/pharmacology , Indans/pharmacology , Lysophospholipids/metabolism , Male , Neuralgia/drug therapy , Organophosphonates/pharmacology , Oxadiazoles/pharmacology , Paclitaxel/pharmacology , Propylene Glycols/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Lysosphingolipid/antagonists & inhibitors , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , Thiazoles/pharmacology , Thiophenes/pharmacology
11.
Bioorg Med Chem Lett ; 24(15): 3302-6, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24969016

ABSTRACT

2-Arylethynyl-(N)-methanocarba adenosine 5'-methyluronamides containing rigid N(6)-(trans-2-phenylcyclopropyl) and 2-phenylethynyl groups were synthesized as agonists for probing structural features of the A3 adenosine receptor (AR). Radioligand binding confirmed A3AR selectivity and N(6)-1S,2R stereoselectivity for one diastereomeric pair. The environment of receptor-bound, conformationally constrained N(6) groups was explored by docking to an A3AR homology model, indicating specific hydrophobic interactions with the second extracellular loop able to modulate the affinity profile. 2-Pyridylethynyl derivative 18 was administered orally in mice to reduce chronic neuropathic pain in the chronic constriction injury model.


Subject(s)
Adenosine A3 Receptor Antagonists/pharmacology , Nucleosides/pharmacology , Receptor, Adenosine A3/metabolism , Adenosine A3 Receptor Antagonists/administration & dosage , Adenosine A3 Receptor Antagonists/chemistry , Animals , CHO Cells , Chronic Pain/drug therapy , Cricetulus , Crystallography, X-Ray , Disease Models, Animal , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Ligands , Mice , Models, Molecular , Molecular Structure , Nucleosides/administration & dosage , Nucleosides/chemistry , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 24(14): 3088-91, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24863744

ABSTRACT

DDD-028 (4), a novel pentacyclic pyridoindolobenzazepine derivative was evaluated in vitro for receptor binding affinity and in vivo for analgesic activity using rodent models of neuropathic and inflammatory pain. DDD-028 does not bind to opioid, cannabinoid, dopamine, or histamine receptors. DDD-028 is very active even at the low oral dose of 1-5 mg/kg in both neuropathic, (spinal nerve ligation and chronic constriction injury) and inflammatory (Complete Freund's Adjuvant Induced) models of pain. DDD-028 appears to be about 6-fold more potent than pregabalin and indomethacin. Visual observation of all the animals used in these studies indicated that DDD-028 is well tolerated without any sedation. Thus, DDD-028 seems to be a promising candidate for the treatment of neuropathic and inflammatory pain without the possible side effects or abuse potential associated with opioid or cannabinoid activities.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Azepines/pharmacology , Carbolines/pharmacology , Constriction, Pathologic/drug therapy , Inflammation/drug therapy , Neuralgia/drug therapy , Spinal Nerves/drug effects , Analgesics/administration & dosage , Analgesics/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Azepines/administration & dosage , Azepines/chemistry , Carbolines/administration & dosage , Carbolines/chemistry , Chronic Disease , Mice , Molecular Structure , Pain Measurement , Rats , Spinal Nerves/pathology
13.
J Neurosci ; 32(32): 10797-808, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22875915

ABSTRACT

Peroxynitrite (PN, ONOO(-)) is a potent oxidant and nitrating agent that contributes to pain through peripheral and spinal mechanisms, but its supraspinal role is unknown. We present evidence here that PN in the rostral ventromedial medulla (RVM) is essential for descending nociceptive modulation in rats during inflammatory and neuropathic pain through PN-mediated suppression of opioid signaling. Carrageenan-induced thermal hyperalgesia was associated with increased 3-nitrotyrosine (NT), a PN biomarker, in the RVM. Furthermore, intra-RVM microinjections of the PN decomposition catalyst Fe(III)-5,10,15,20-tetrakis(N-methyl-pyridinium-4-yl)porphyrin (FeTMPyP(5+)) dose-dependently reversed this thermal hyperalgesia. These effects of FeTMPyP(5+) were abrogated by intra-RVM naloxone, implicating potential interplay between PN and opioids. In support, we identified NT colocalization with the endogenous opioid enkephalin (ENK) in the RVM during thermal hyperalgesia, suggesting potential in situ interactions. To address the functional significance of such interactions, we exposed methionine-enkephalin (MENK) to PN and identified the major metabolite, 3-nitrotyrosine-methionine-sulfoxide (NSO)-MENK, using liquid chromatography-mass spectrometry. Next, we isolated, purified, and tested NSO-MENK for opioid receptor binding affinity and analgesic effects. Compared to MENK, this NSO-MENK metabolite lacked appreciable binding affinity for δ, µ, and κ opioid receptors. Intrathecal injection of NSO-MENK in rats did not evoke antinociception, suggesting that PN-mediated chemical modifications of ENK suppress opioid signaling. When extended to chronic pain, intra-RVM FeTMPyP(5+) produced naloxone-sensitive reversal of mechanical allodynia in rats following chronic constriction injury of the sciatic nerve. Collectively, our data reveal the central role of PN in RVM descending facilitation during inflammatory and neuropathic pain potentially through anti-opioid activity.


Subject(s)
Hyperalgesia/drug therapy , Medulla Oblongata/metabolism , Opioid Peptides/metabolism , Pain/drug therapy , Peroxynitrous Acid/administration & dosage , Signal Transduction/drug effects , Analysis of Variance , Animals , CD11b Antigen/metabolism , Carrageenan/adverse effects , Cell Line, Transformed , Chromatography, Liquid , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Electrochemical Techniques , Enkephalin, Methionine/therapeutic use , Glial Fibrillary Acidic Protein/metabolism , Humans , Hyperalgesia/chemically induced , Hyperalgesia/pathology , Injections, Spinal , Male , Medulla Oblongata/drug effects , Metalloporphyrins/therapeutic use , Microinjections , Neuroglia/metabolism , Neurons/metabolism , Pain Measurement , Phosphopyruvate Hydratase/metabolism , Protein Binding/drug effects , Radioligand Assay , Rats , Rats, Sprague-Dawley , Tyrosine/analogs & derivatives , Tyrosine/metabolism
14.
J Neurosci ; 32(18): 6149-60, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22553021

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) accompanied by chronic neuropathic pain is a major dose-limiting side effect of a large number of antitumoral agents including paclitaxel (Taxol). Thus, CIPN is one of most common causes of dose reduction and discontinuation of what is otherwise a life-saving therapy. Neuropathological changes in spinal cord are linked to CIPN, but the causative mediators and mechanisms remain poorly understood. We report that formation of peroxynitrite (PN) in response to activation of nitric oxide synthases and NADPH oxidase in spinal cord contributes to neuropathological changes through two mechanisms. The first involves modulation of neuroexcitatory and proinflammatory (TNF-α and IL-1ß) and anti-inflammatory (IL-10 and IL-4) cytokines in favor of the former. The second involves post-translational nitration and modification of glia-derived proteins known to be involved in glutamatergic neurotransmission (astrocyte-restricted glutamate transporters and glutamine synthetase). Targeting PN with PN decomposition catalysts (PNDCs) not only blocked the development of paclitaxel-induced neuropathic pain without interfering with antitumor effects, but also reversed it once established. Herein, we describe our mechanistic study on the role(s) of PN and the prevention of neuropathic pain in rats using known PNDCs (FeTMPyP(5+) and MnTE-2-PyP(5+)). We also demonstrate the prevention of CIPN with our two new orally active PNDCs, SRI6 and SRI110. The improved chemical design of SRI6 and SRI110 also affords selectivity for PN over other reactive oxygen species (such as superoxide). Our findings identify PN as a critical determinant of CIPN, while providing the rationale toward development of superoxide-sparing and "PN-targeted" therapeutics.


Subject(s)
Cytokines/metabolism , Drug Delivery Systems/methods , Neuralgia/metabolism , Paclitaxel/adverse effects , Peroxynitrous Acid/metabolism , Spinal Cord/metabolism , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Male , Neuralgia/chemically induced , Neuralgia/prevention & control , Paclitaxel/therapeutic use , Peroxynitrous Acid/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects
15.
FASEB J ; 26(5): 1855-65, 2012 May.
Article in English | MEDLINE | ID: mdl-22345405

ABSTRACT

Clinical management of chronic neuropathic pain is limited by marginal effectiveness and unacceptable side effects of current drugs. We demonstrate A(3) adenosine receptor (A(3)AR) agonism as a new target-based therapeutic strategy. The development of mechanoallodynia in a well-characterized mouse model of neuropathic pain following chronic constriction injury of the sciatic nerve was rapidly and dose-dependently reversed by the A(3)AR agonists: IB-MECA, its 2-chlorinated analog (Cl-IB-MECA), and the structurally distinct MRS1898. These effects were naloxone insensitive and thus are not opioid receptor mediated. IB-MECA was ≥1.6-fold more efficacious than morphine and >5-fold more potent. In addition, IB-MECA was equally efficacious as gabapentin (Neurontin) or amitriptyline, but respectively >350- and >75-fold more potent. Besides its potent standalone ability to reverse established mechanoallodynia, IB-MECA significantly increased the antiallodynic effects of all 3 analgesics. Moreover, neuropathic pain development in rats caused by widely used chemotherapeutics in the taxane (paclitaxel), platinum-complex (oxaliplatin), and proteasome-inhibitor (bortezomib) classes was blocked by IB-MECA without antagonizing their antitumor effect. A(3)AR agonist effects were blocked with A(3)AR antagonist MRS1523, but not with A(1)AR (DPCPX) or A(2A)AR (SCH-442416) antagonists. Our findings provide the scientific rationale and pharmacological basis for therapeutic development of A(3)AR agonists for chronic pain.


Subject(s)
Adenosine A3 Receptor Agonists/therapeutic use , Neuralgia/drug therapy , Pain Management/methods , Animals , Chronic Disease , Male , Mice , Rats , Rats, Sprague-Dawley , Rotarod Performance Test
16.
J Med Chem ; 66(13): 9076-9094, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37382926

ABSTRACT

P2Y14 receptor (P2Y14R) is activated by extracellular UDP-glucose, a damage-associated molecular pattern that promotes inflammation in the kidney, lung, fat tissue, and elsewhere. Thus, selective P2Y14R antagonists are potentially useful for inflammatory and metabolic diseases. The piperidine ring size of potent, competitive P2Y14R antagonist (4-phenyl-2-naphthoic acid derivative) PPTN 1 was varied from 4- to 8-membered rings, with bridging/functional substitution. Conformationally and sterically modified isosteres included N-containing spirocyclic (6-9), fused (11-13), and bridged (14, 15) or large (16-20) ring systems, either saturated or containing alkene or hydroxy/methoxy groups. The alicyclic amines displayed structural preference. An α-hydroxyl group increased the affinity of 4-(4-((1R,5S,6r)-6-hydroxy-3-azabicyclo[3.1.1]heptan-6-yl)phenyl)-7-(4-(trifluoromethyl)phenyl)-2-naphthoic acid 15 (MRS4833) compared to 14 by 89-fold. 15 but not its double prodrug 50 reduced airway eosinophilia in a protease-mediated asthma model, and orally administered 15 and prodrugs reversed chronic neuropathic pain (mouse CCI model). Thus, we identified novel drug leads having in vivo efficacy.


Subject(s)
Receptors, Purinergic P2 , Mice , Animals , Receptors, Purinergic P2/metabolism , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Uridine Diphosphate Glucose/metabolism
17.
FASEB J ; 25(8): 2782-91, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21551240

ABSTRACT

Inflammatory pain represents an important unmet clinical need with important socioeconomic implications. Ceramide, a potent proinflammatory sphingolipid, has been shown to elicit mechanical hyperalgesia, but the mechanisms remain largely unknown. We now demonstrate that, in addition to mechanical hyperalgesia, intraplantar injection of ceramide (10 µg) led to the development of thermal hyperalgesia that was dependent on induction of the inducible cyclooxygenase (COX-2) and subsequent increase of prostaglandin E(2) (PGE(2)). The development of mechanical and thermal hyperalgesia and increased production of PGE(2) was blocked by NS-398 (15-150 ng), a selective COX-2 inhibitor. The importance of the COX-2 to PGE(2) pathway in ceramide signaling was underscored by the findings that intraplantar injection of a monoclonal PGE(2) antibody (4 µg) blocked the development of hyperalgesia. Our results further revealed that COX-2 induction is regulated by NF-κB and p38 kinase activation, since intraplantar injection of SC-514 (0.1-1 µg) or SB 203580 (1-10 µg), well-characterized inhibitors of NF-κB and p38 kinase activation, respectively, blocked COX-2 induction and increased formation of PGE(2) and thermal hyperalgesia in a dose-dependent manner. Moreover, activation of NF-κB was dependent on upstream activation of p38 MAPK, since SB 203580 (10 µg) blocked p65 phosphorylation, whereas p38 kinase phosphorylation was unaffected by NF-κB inhibition by SC-514 (1 µg). Our findings not only provide mechanistic insight into the signaling pathways engaged by ceramide in the development of hyperalgesia, but also provide a potential pharmacological basis for developing inhibitors targeting the ceramide metabolic-to-COX-2 pathway as novel analgesics.


Subject(s)
Ceramides/toxicity , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , NF-kappa B/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Ceramides/administration & dosage , Ceramides/metabolism , Cyclooxygenase 2 Inhibitors/administration & dosage , Dinoprostone/antagonists & inhibitors , Extremities , Hot Temperature , Hyperalgesia/prevention & control , Imidazoles/administration & dosage , Injections , Male , Models, Biological , NF-kappa B/antagonists & inhibitors , Pain Threshold/drug effects , Pain Threshold/physiology , Pyridines/administration & dosage , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Thiophenes/administration & dosage , Touch , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
18.
J Med Chem ; 65(4): 3434-3459, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35113556

ABSTRACT

High affinity phenyl-piperidine P2Y14R antagonist 1 (PPTN) was modified with piperidine bridging moieties to probe receptor affinity and hydrophobicity. Various 2-azanorbornane, nortropane, isonortropane, isoquinuclidine, and ring-opened cyclopentylamino derivatives preserved human P2Y14R affinity (fluorescence binding assay), and their pharmacophoric overlay was compared. Enantiomeric 2-azabicyclo[2.2.1]hept-5-en-3-one precursors assured stereochemically unambiguous, diverse products. Pure (S,S,S) 2-azanorbornane enantiomer 15 (MRS4738) displayed higher affinity than 1 (3-fold higher affinity than enantiomer 16) and in vivo antihyperallodynic and antiasthmatic activity. Its double prodrug 143 (MRS4815) dramatically reduced lung inflammation in a mouse asthma model. Related lactams 21-24 and dicarboxylate 42 displayed intermediate affinity and enhanced aqueous solubility. Isoquinuclidine 34 (IC50 15.6 nM) and isonortropanol 30 (IC50 21.3 nM) had lower lipophilicity than 1. In general, rigidified piperidine derivatives did not lower lipophilicity dramatically, except those rings with multiple polar groups. P2Y14R molecular modeling based on a P2Y12R structure showed stable and persistent key interactions for compound 15.


Subject(s)
Piperidines/chemistry , Purinergic P2 Receptor Antagonists/pharmacology , Animals , Mice , Purinergic P2 Receptor Antagonists/chemistry , Structure-Activity Relationship
19.
J Neurosci ; 30(46): 15400-8, 2010 Nov 17.
Article in English | MEDLINE | ID: mdl-21084596

ABSTRACT

The clinical efficacy of opiates for pain control is severely limited by analgesic tolerance and hyperalgesia. Herein we show that chronic morphine upregulates both the sphingolipid ceramide in spinal astrocytes and microglia, but not neurons, and spinal sphingosine-1-phosphate (S1P), the end-product of ceramide metabolism. Coadministering morphine with intrathecal administration of pharmacological inhibitors of ceramide and S1P blocked formation of spinal S1P and development of hyperalgesia and tolerance in rats. Our results show that spinally formed S1P signals at least in part by (1) modulating glial function because inhibiting S1P formation blocked increased formation of glial-related proinflammatory cytokines, in particular tumor necrosis factor-α, interleukin-1ßα, and interleukin-6, which are known modulators of neuronal excitability, and (2) peroxynitrite-mediated posttranslational nitration and inactivation of glial-related enzymes (glutamine synthetase and the glutamate transporter) known to play critical roles in glutamate neurotransmission. Inhibitors of the ceramide metabolic pathway may have therapeutic potential as adjuncts to opiates in relieving suffering from chronic pain.


Subject(s)
Analgesics, Opioid/pharmacology , Neuroglia/drug effects , Neuroglia/physiology , Sphingolipids/physiology , Analgesics, Opioid/therapeutic use , Animals , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Lysophospholipids/physiology , Male , Pain Measurement/drug effects , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Sphingosine/analogs & derivatives , Sphingosine/physiology
20.
J Am Chem Soc ; 133(12): 4200-3, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21370860

ABSTRACT

We report a new series of biscyclohexano-fused Mn(III) complexes of bis(hydroxyphenyl)dipyrromethenes, 4a-c, as potent and orally active peroxynitrite scavengers. Complexes 4a-c are shown to reduce peroxynitrite through a two-electron mechanism, thereby forming the corresponding Mn(V)O species, which were characterized by UV, NMR, and LC-MS methods. Mn(III) complex 4b and its strained BODIPY analogue 9b were analyzed by X-ray crystallography. Finally, complex 4a is shown to be an orally active and potent analgesic in a model carrageenan-induced hyperalgesia known to be driven by the overproduction of peroxynitrite.


Subject(s)
Free Radical Scavengers/chemistry , Manganese/chemistry , Organometallic Compounds/chemistry , Peroxynitrous Acid/chemistry , Porphobilinogen/analogs & derivatives , Crystallography, X-Ray , Free Radical Scavengers/chemical synthesis , Models, Molecular , Molecular Structure , Organometallic Compounds/chemical synthesis , Porphobilinogen/chemistry , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL