Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
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
2.
Pain ; 154(11): 2432-2440, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23891899

ABSTRACT

Many of the widely used anticancer drugs induce dose-limiting peripheral neuropathies that undermine their therapeutic efficacy. Animal models of chemotherapy-induced painful peripheral neuropathy (CIPN) evoked by a variety of drug classes, including taxanes, vinca alkaloids, platinum-complexes, and proteasome-inhibitors, suggest that the common underlying mechanism in the development of these neuropathies is mitotoxicity in primary nerve sensory axons (PNSAs) arising from reduced mitochondrial bioenergetics [eg adenosine triphosphate (ATP) production deficits due to compromised respiratory complex I and II activity]. The causative mechanisms of this mitotoxicity remain poorly defined. However, peroxynitrite, an important pro-nociceptive agent, has been linked to mitotoxicity in several disease states and may also drive the mitotoxicity associated with CIPN. Our findings reveal that the development of mechano-hypersensitivity induced by paclitaxel, oxaliplatin, and bortezomib was prevented by administration of the peroxynitrite decomposition catalyst Mn(III) 5,10,15,20-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin (MnTE-2-PyP(5+)) without interfering with their anti-tumor effects. Peak CIPN was associated with the nitration and inactivation of superoxide dismutase in the mitochondria, but not in the cytosol, as well as a significant decrease in ATP production within the PNSAs; all of these events were attenuated by MnTE-2-PyP(5+). Our results provide continued support for the role of mitotoxicity in the development of CIPN across chemotherapeutic drug classes, and identify peroxynitrite as a key mediator in these processes, thereby providing the rationale towards development of "peroxynitrite-targeted" therapeutics for CIPN.


Subject(s)
Antineoplastic Agents/adverse effects , Axons/physiology , Energy Metabolism/physiology , Neuralgia/chemically induced , Neuralgia/metabolism , Peripheral Nerves/physiology , Peroxynitrous Acid/physiology , Sensory Receptor Cells/physiology , Superoxide Dismutase/metabolism , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Boronic Acids/pharmacology , Bortezomib , Hyperalgesia/drug therapy , Hyperalgesia/psychology , Male , Mitochondria/drug effects , Mitochondria/ultrastructure , Neoplasm Transplantation , Organoplatinum Compounds/pharmacology , Oxaliplatin , Paclitaxel/pharmacology , Physical Stimulation , Protein Processing, Post-Translational/physiology , Pyrazines/pharmacology , Rats , Rats, Sprague-Dawley
3.
Pain ; 154(7): 978-86, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23590939

ABSTRACT

Treatment of severe pain by morphine, the gold-standard opioid and a potent drug in our arsenal of analgesic medications, is limited by the eventual development of hyperalgesia and analgesic tolerance. We recently reported that systemic administration of a peroxynitrite (PN) decomposition catalyst (PNDC) or superoxide dismutase mimetic attenuates morphine hyperalgesia and antinociceptive tolerance and reduces PN-mediated mitochondrial nitroxidative stress in the spinal cord. These results suggest the potential involvement of spinal PN signaling in this setting; which was examined in the present study. PN removal with intrathecal delivery of manganese porphyrin-based dual-activity superoxide/PNDCs, MnTE-2-PyP(5+) and the more lipophilic MnTnHex-2-PyP(5+), blocked hyperalgesia and antinociceptive tolerance in rats. Noteworthy is that intrathecal MnTnHex-2-PyP(5+) prevented nitration and inactivation of mitochondrial manganese superoxide dismutase. Mitochondrial manganese superoxide dismutase inactivation enhances the superoxide-to-PN pathway by preventing the dismutation of superoxide to hydrogen peroxide, thus providing an important enzymatic source for PN formation. Additionally, intrathecal MnTnHex-2-PyP(5+) attenuated neuroimmune activation by preventing the activation of nuclear factor kappa B, extracellular-signal-regulated kinase and p38 mitogen activated protein kinases, and the enhanced levels of proinflammatory cytokines, interleukin (IL)-1ß and IL-6, while increasing anti-inflammatory cytokines, IL-4 and IL-10. The role of PN was further confirmed using intrathecal or oral delivery of the superoxide-sparing PNDC, SRI-110. These results suggest that mitochondrial-derived PN triggers the activation of several biochemical pathways engaged in the development of neuroinflammation in the spinal cord that are critical to morphine hyperalgesia and tolerance, further supporting the potential of targeting PN as an adjunct to opiates to maintain pain relief.


Subject(s)
Hyperalgesia/chemically induced , Hyperalgesia/immunology , Mitochondria/immunology , Morphine/adverse effects , Neuroimmunomodulation/immunology , Peroxynitrous Acid/immunology , Spinal Cord/immunology , Analgesics/adverse effects , Analgesics, Opioid/adverse effects , Animals , Drug Interactions/immunology , Drug Tolerance/immunology , Hyperalgesia/prevention & control , Male , Mitochondria/drug effects , Neuroimmunomodulation/drug effects , Peroxynitrous Acid/administration & dosage , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Treatment Outcome
4.
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
5.
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
6.
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
7.
J Med Chem ; 54(24): 8658-69, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22082008

ABSTRACT

Redox-active metalloporphyrins represent the most well-characterized class of catalysts capable of attenuating oxidative stress in vivo through the direct interception and decomposition of superoxide and peroxynitrite. While many interesting pharmacological probes have emerged from these studies, few catalysts have been developed with pharmaceutical properties in mind. Herein, we describe our efforts to identify new Mn(III)-porphyrin systems with enhanced membrane solubilizing properties. To this end, seven new Mn(III)-tetracyclohexenylporphyin (TCHP) analogues, 7, 10, 12, 15, and 16a-c, have been prepared in which the beta-fused cyclohexenyl rings provide a means to shield the charged metal center from the membrane during passive transport. Compounds 7, 15, and 16a-c have been shown to be orally active and potent analgesics in a model of carrageenan-induced thermal hyperalgesia. In addition, oral administration of compound 7 (10-100 mg/kg, n=5) has been shown to dose dependently reverse mechano-allodynia in the CCI model of chronic neuropathic pain.


Subject(s)
Analgesics/chemical synthesis , Manganese , Metalloporphyrins/chemical synthesis , Peroxynitrous Acid/metabolism , Administration, Oral , Analgesics/chemistry , Analgesics/pharmacology , Animals , Boronic Acids/chemistry , Carrageenan , Catalysis , Chronic Pain/drug therapy , Electrochemistry , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Metalloporphyrins/chemistry , Metalloporphyrins/pharmacology , Mice , Neuralgia/drug therapy , Peroxynitrous Acid/chemistry , Rats , Sciatic Nerve/injuries , Structure-Activity Relationship , Superoxide Dismutase/chemistry
8.
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
9.
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
10.
J Parasitol ; 96(4): 758-64, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20738200

ABSTRACT

Chagas' disease, induced by Trypanosoma cruzi , is a common cause of infectious myocarditis. Recent clinical treatment trials and vaccine studies indicate that chagasic immunopathology is directed against the parasite and not self-antigens. Therefore, vaccines may have the potential to protect against disease progression. Certain combinations of mouse and parasite strains produce significant histopathology and can be used for safety analyses of new vaccination strategies. The goals of this study were to determine (1) whether the development of chagasic cardiomyopathy in the murine model could be identified by electrocardiogram (ECG); and (2) whether these potential chagasic ECG changes would correlate with histopathologic findings. Groups of BALB/c, C57BL/6, and C3H mice were infected with different parasite strains (Tulahuén, Brazil, or Sylvio-X10/4) and evaluated weekly by ECG. Selected tissues from subsets of mice were harvested periodically for blinded histologic evaluation. Significantly increased proportions of BALB/c mice infected with Brazil and Tulahuén strain parasites displayed prolonged QT intervals. Prolonged mean QT intervals detected in infected BALB/c mice significantly correlated with chagasic histopathologic changes. These results indicate that ECG can be used as a non-invasive method to screen for immune-mediated damage resulting in chagasic cardiomyopathy in the murine model.


Subject(s)
Chagas Cardiomyopathy/diagnosis , Electrocardiography , Animals , Chagas Cardiomyopathy/pathology , Disease Models, Animal , Female , Inflammation/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, SCID , Muscle, Skeletal/pathology , Myocardium/pathology , Statistics, Nonparametric
11.
Neurosci Lett ; 483(2): 85-9, 2010 Oct 11.
Article in English | MEDLINE | ID: mdl-20637262

ABSTRACT

The role of superoxide and its active byproduct peroxynitrite as mediators of nociceptive signaling is emerging. We have recently reported that nitration and inactivation of spinal mitochondrial superoxide dismutase (MnSOD) provides a critical source of these reactive oxygen and nitrogen species during central sensitization associated with the development of morphine-induced hyperalgesia and antinociceptive tolerance. In this study, we demonstrate that activation of spinal NADPH oxidase is another critical source for superoxide generation. Indeed, the development of morphine-induced hyperalgesia and antinociceptive tolerance was associated with increased activation of NADPH oxidase and superoxide release. Co-administration of morphine with systemic delivery of two structurally unrelated NADPH oxidase inhibitors namely apocynin or diphenyleneiodonium (DPI), blocked NADPH oxidase activation and the development of hyperalgesia and antinociceptive tolerance at doses devoid of behavioral side effects. These results suggest that activation of spinal NADPH oxidase contributes to the development of morphine-induced hyperalgesia and antinociceptive tolerance. The role of spinal NADPH oxidase was confirmed by showing that intrathecal delivery of apocynin blocked these events. Our results are the first to implicate the contribution of NADPH oxidase as an enzymatic source of superoxide and thus peroxynitrite in the development of central sensitization associated with morphine-induced hyperalgesia and antinociceptive tolerance. These results continue to support the critical role of these reactive oxygen and nitrogen species in pain while advancing our knowledge of their biomolecular sources.


Subject(s)
Drug Tolerance/physiology , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Morphine/toxicity , NADPH Oxidases/chemistry , NADPH Oxidases/physiology , Spinal Cord/metabolism , Superoxides/chemistry , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacology , Animals , Hyperalgesia/enzymology , Male , Mice , Morphine/metabolism , NADPH Oxidases/antagonists & inhibitors , Spinal Cord/drug effects , Spinal Cord/enzymology , Superoxides/metabolism
12.
Neurosci Lett ; 463(1): 49-53, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19631718

ABSTRACT

Opiates, like morphine, are the most effective analgesics for treating acute and chronic severe pain, but their use is limited by the development of analgesic tolerance and hypersensitivity to innocuous and noxious stimuli. Because opioids are a mainstay of pain management, restoring their efficacy has great clinical importance. We have recently demonstrated that spinal ceramide, a sphingolipid signaling molecule plays a central role in the development of morphine antinociceptive tolerance. We now report that ceramide upregulation in dorsal horn tissues in response to chronic morphine administration is associated with significant neuronal apoptosis. Inhibition of ceramide biosynthesis attenuated both the increase in neuronal apoptosis and the development of antinociceptive tolerance. These findings indicate that spinal ceramide upregulation is a key pro-apoptotic event that occurs upstream of the development of morphine antinociceptive tolerance and support the rationale for development of inhibitors of ceramide biosynthesis as adjuncts to opiates for the management of chronic pain.


Subject(s)
Analgesics, Opioid/adverse effects , Apoptosis , Ceramides/biosynthesis , Drug Tolerance , Morphine/adverse effects , Posterior Horn Cells/drug effects , Spinal Cord/metabolism , Analgesics, Opioid/administration & dosage , Animals , Ceramides/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Activation , Male , Mice , Morphine/administration & dosage , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism , Posterior Horn Cells/metabolism , Posterior Horn Cells/pathology , Spinal Cord/pathology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...