Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Neurosci Methods ; 402: 110006, 2024 02.
Article in English | MEDLINE | ID: mdl-37967672

ABSTRACT

BACKGROUND: Delivery of therapeutics via indwelling intrathecal catheters is highly efficacious for targeting of pain, spasticity, neuraxial cancer and neurodegenerative disorders. However, current catheter designs have some major limitations. Given limited CSF flow, fixed intrathecal volume and the large distance of the rostro-caudal spinal axis, current intrathecal delivery routes fail to achieve adequate drug distribution. Additionally open catheter systems are plagued with cellular ingrowth and debris accumulation if used intermittently. NEW METHOD: RESULTS/COMPARISON WITH EXISTING METHOD(S): High speed imaging showed micro-valve catheters greatly increase fluid exit velocities compared to typical open-ended catheters, which prevents pooling of injectate proximal to the opening. When implanted intrathecally in rats, small injection volumes (7.5 µL) of dye or AAV9-RFP, resulted in an even rostro-caudal distribution along the spinal axis and robust transfection of neurons from cervical to lumbar dorsal root ganglia. In contrast, such injections with an open-ended catheter resulted in localized distribution and transfection proximal to the delivery site. Our poly micro-valve catheter design resulted in equivalent transfection rates of cervical DRG neurons using 100x lower titer of AAV9-RFP. Unlike open port catheters, no debris accumulation was observed in the lumen of implanted catheters, showing potential for long-term intermittent use. CONCLUSIONS: This catheter platform, suitable for small animal models is easily scalable for human use and addresses many of the problems observed with common catheter systems.


Subject(s)
Catheterization , Catheters, Indwelling , Humans , Rats , Animals , Catheterization/methods , Pain , Central Nervous System , Injections, Spinal/methods
2.
Neurotoxicology ; 88: 155-167, 2022 01.
Article in English | MEDLINE | ID: mdl-34801587

ABSTRACT

Spinally-administered local anesthetics provide effective perioperative anesthesia and/or analgesia for children of all ages. New preparations and drugs require preclinical safety testing in developmental models. We evaluated age-dependent efficacy and safety following 1 % preservative-free 2-chloroprocaine (2-CP) in juvenile Sprague-Dawley rats. Percutaneous lumbar intrathecal 2-CP was administered at postnatal day (P)7, 14 or 21. Mechanical withdrawal threshold pre- and post-injection evaluated the degree and duration of sensory block, compared to intrathecal saline and naive controls. Tissue analyses one- or seven-days following injection included histopathology of spinal cord, cauda equina and brain sections, and quantification of neuronal apoptosis and glial reactivity in lumbar spinal cord. Following intrathecal 2-CP or saline at P7, outcomes assessed between P30 and P72 included: spinal reflex sensitivity (hindlimb thermal latency, mechanical threshold); social approach (novel rat versus object); locomotor activity and anxiety (open field with brightly-lit center); exploratory behavior (rearings, holepoking); sensorimotor gating (acoustic startle, prepulse inhibition); and learning (Morris Water Maze). Maximum tolerated doses of intrathecal 2-CP varied with age (1.0 µL/g at P7, 0.75 µL/g at P14, 0.5 µL/g at P21) and produced motor and sensory block for 10-15 min. Tissue analyses found no significant differences across intrathecal 2-CP, saline or naïve groups. Adult behavioral measures showed expected sex-dependent differences, that did not differ between 2-CP and saline groups. Single maximum tolerated in vivo doses of intrathecal 2-CP produced reversible spinal anesthesia in juvenile rodents without detectable evidence of developmental neurotoxicity. Current results cannot be extrapolated to repeated dosing or prolonged infusion.


Subject(s)
Neurotoxicity Syndromes/etiology , Procaine/analogs & derivatives , Animals , Caspase 3/metabolism , Cauda Equina/anatomy & histology , Cauda Equina/drug effects , Female , Injections, Spinal , Male , Morris Water Maze Test/drug effects , Motor Activity/drug effects , Procaine/administration & dosage , Procaine/toxicity , Rats , Rats, Sprague-Dawley , Sensory Gating/drug effects
3.
Anesthesiology ; 131(1): 132-147, 2019 07.
Article in English | MEDLINE | ID: mdl-31225809

ABSTRACT

BACKGROUND: As the meningeally derived, fibroblast-rich, mass-produced by intrathecal morphine infusion is not produced by all opiates, but reduced by mast cell stabilizers, the authors hypothesized a role for meningeal mast cell/fibroblast activation. Using the guinea pig, the authors asked: (1) Are intrathecal morphine masses blocked by opiate antagonism?; (2) Do opioid agonists not producing mast cell degranulation or fibroblast activation produce masses?; and (3) Do masses covary with Mas-related G protein-coupled receptor signaling thought to mediate mast cell degranulation? METHODS: In adult male guinea pigs (N = 66), lumbar intrathecal catheters connected to osmotic minipumps (14 days; 0.5 µl/h) were placed to deliver saline or equianalgesic concentrations of morphine sulfate (33 nmol/h), 2',6'-dimethyl tyrosine-(Tyr-D-Arg-Phe-Lys-NH2) (abbreviated as DMT-DALDA; 10 pmol/h; µ agonist) or PZM21 (27 nmol/h; biased µ agonist). A second pump delivered subcutaneous naltrexone (25 µg/h) in some animals. After 14 to 16 days, animals were anesthetized and perfusion-fixed. Drug effects on degranulation of human cultured mast cells, mouse embryonic fibroblast activation/migration/collagen formation, and Mas-related G protein-coupled receptor activation (PRESTO-Tango assays) were determined. RESULTS: Intrathecal infusion of morphine, DMT-DALDA or PZM21, but not saline, comparably increased thermal thresholds for 7 days. Spinal masses proximal to catheter tip, composed of fibroblast/collagen type I (median: interquartile range, 0 to 4 scale), were produced by morphine (2.3: 2.0 to 3.5) and morphine plus naltrexone (2.5: 1.4 to 3.1), but not vehicle (1.2: 1.1 to 1.5), DMT-DALDA (1.0: 0.6 to 1.3), or PZM21 (0.5: 0.4 to 0.8). Morphine in a naloxone-insensitive fashion, but not PZM21 or DMT-DALDA, resulted in mast cell degranulation and fibroblast proliferation/collagen formation. Morphine-induced fibroblast proliferation, as mast cell degranulation, is blocked by cromolyn. Mas-related G protein-coupled receptor activation was produced by morphine and TAN67 (∂-opioid agonist), but not by PZM21, TRV130 (mu biased ligand), or DMT-DALDA. CONCLUSIONS: Opiates that activate Mas-related G protein-coupled receptor will degranulate mast cells, activate fibroblasts, and result in intrathecal mass formation. Results suggest a mechanistically rational path forward to safer intrathecal opioid therapeutics.


Subject(s)
Cell Degranulation/drug effects , Fibroblasts/drug effects , Mast Cells/drug effects , Morphine/pharmacology , Receptors, G-Protein-Coupled/physiology , Spine/drug effects , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Guinea Pigs , Humans , Infusions, Spinal , Male , Models, Animal , Morphine/administration & dosage , Signal Transduction/physiology
4.
Neuromodulation ; 22(7): 790-798, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31124198

ABSTRACT

BACKGROUND: We determined whether intrathecally delivering the same daily dose of morphine (MS) at a fixed concentration of 25 mg/mL by periodic boluses versus continuous infusion would reduce intrathecal mass (IMs) formation in dogs. METHODS: Adult dogs (hound cross, n = 32) were implanted with intrathecal catheters connected to SynchroMed II infusion pumps. Animals were randomly assigned to receive infusion of 0.48 mL/day of saline or MS dosing (12 mg/day at 25 mg/mL) as boluses: x1 (q24hour), x2 (q12hour), x4 (q6hour), or x8 (q3hour) given at the rate of 1000 µL/hour, or as a continuous infusion (25 mg/mL/20 µL/hour). RESULTS: With IT saline, minimal pathology was noted. In contrast, animals receiving morphine displayed spinally compressing durally derived masses with the maximal cross-sectional area being greatest near the catheter tip. Histopathology showed that IMs consisted of fibroblasts in a collagen (type 1) matrix comprised of newly formed collagen near the catheter and mature collagen on the periphery of the mass. The rank order of median cross-sectional mass area (mm2 ) was: Saline: 0.7 mm2 ; x2: 1.8 mm2 ; x4: 2.7 mm2 ; x1: 2.7 mm2 ; x8: 4.2 mm2 ; Continuous: 8.1 mm2 , with statistical difference from saline being seen with continuous (p < 0.0001) and x8 (p < 0.05). Bench studies with a 2D diffusion chamber confirmed an increase in dye distribution and lower peak concentrations after bolus delivery versus continuous infusion of dye. CONCLUSIONS: Using multiple bolus dosing, IMs were reduced as compared to continuous infusion, suggesting relevance of bolus delivery in yielding reduced intrathecal masses.


Subject(s)
Analgesics, Opioid/administration & dosage , Infusion Pumps, Implantable/trends , Morphine/administration & dosage , Spinal Cord/drug effects , Spinal Cord/pathology , Analgesics, Opioid/adverse effects , Animals , Dogs , Drug Administration Schedule , Female , Infusion Pumps, Implantable/adverse effects , Injections, Spinal/adverse effects , Injections, Spinal/instrumentation , Injections, Spinal/trends , Male , Morphine/adverse effects , Random Allocation
5.
Pain Med ; 20(5): 897-906, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30590777

ABSTRACT

OBJECTIVE: This study modeled image-guided epidural drug delivery to test whether intraprocedural distribution of pre-injected contrast reliably predicts the neuroanatomical reach of resiniferatoxin-mediated nociceptive neurolysis. METHODS: Swine (N = 12) received unilateral L4-S2 computed tomography fluoroscopy injections by a blinded neuroradiologist; 0.25 mL of contrast was pre-injected to confirm dorsal periganglionic targeting, followed by a 0.5-mL injection of 5 µg of resiniferatoxin/Tween80 or vehicle control. Epidural contrast distribution was graded according to maximum medial excursion. Spinal cord substance P immunostaining quantified the magnitude and anatomical range of resiniferatoxin activity. RESULTS: Periganglionic injection was well tolerated by all animals without development of neurological deficits or other complications. Swine were a suitable model of human clinical spinal intervention. The transforaminal approach was used at all L4 and 50% of L5 segments; the remaining segments were approached by the interlaminar route. All injections were successful with unilateral contrast distribution for all resiniferatoxin injections (N = 28). Immunohistochemistry showed bilateral ablation of substance P+ fibers entering the spinal cord of all resiniferatoxin-treated segments. The intensity of substance P immunostaining in treated segments fell below the lower 99% confidence interval of controls, defining the knockout phenotype. Substance P knockout occurred over a narrow range and was uncorrelated to the anatomical distribution of pre-injected contrast. CONCLUSIONS: Periganglionic resiniferatoxin/Tween80 induced bilateral ablation of spinal cord substance P despite exclusively unilateral targeting. These data suggest that the location of pre-injected contrast is an imperfect surrogate for the neuroanatomical range of drugs delivered to the dorsal epidural compartment that may fail to predict contralateral drug effects.


Subject(s)
Diterpenes/administration & dosage , Nerve Block/methods , Neurotoxins/administration & dosage , Animals , Female , Fluoroscopy/methods , Injections, Epidural , Spinal Nerve Roots/drug effects , Swine , Therapy, Computer-Assisted/methods
6.
Vet Anaesth Analg ; 45(2): 212-226, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29361418

ABSTRACT

OBJECTIVE: To evaluate target engagement of intracisternally (IC) delivered TRPV1 agonist, resiniferatoxin (RTX), as measured by primary afferent and dorsal horn substance P immunoreactivity (sP-IR), histopathology and thermal escape latencies in dogs. STUDY DESIGN: Prospective experimental trial. ANIMALS: Fourteen adult male Beagle dogs, weighing 10.3-13.2 kg; 11 dogs surviving to scheduled euthanasia. METHODS: Anesthetized dogs were randomly assigned to be administered IC RTX (3.6 µg, 0.1 mL kg-1) in a hyperbaric (hRTX, n = 6), normobaric (nRTX, n = 4) vehicle or a hyperbaric vehicle (hVehicle, n = 4). Over 16 days, animals were examined for thoracic and pelvic limb paw thermal withdrawal latencies and neurologic function. Spinal cords, trigeminal ganglia and dorsal root ganglia (DRGs) were assessed for morphologic changes and sP-IR. RESULTS: IC RTX in anesthetized dogs resulted in a < 1 hour increase in blood pressure. Acute reactions leading to euthanasia within 8 hours occurred in three dogs (two hRTX, one nRTX). All other animals recovered with normal neurologic, bowel and bladder function. Final groups were: vehicle n = 4, hRTX n = 4 and nRTX n = 3. Animals in nRTX and hRTX showed increases in escape latencies in thoracic paws and, to a lesser extent, in pelvic paws, correlating to a loss of sP-IR in cervical cord with smaller reductions in thoracic and lumbar cord. In animals surviving to euthanasia, thickening of the arachnoid membrane (predominantly in the cervical region) was the most consistent change. This change, present in controls, was interpreted to be vehicle related. There was no evidence of structural changes in brain and spinal cord. CONCLUSIONS AND CLINICAL RELEVANCE: IC RTX produced localized loss of spinal and DRG sP with a corresponding thermal analgesia, absent motor impairment or spinal pathology. Loss of three animals emphasizes the need to refine the use of this promising therapeutic modality in managing companion animal pain.


Subject(s)
Diterpenes/pharmacology , Dogs , Nervous System/drug effects , Neurotoxins/pharmacology , Anesthesia/veterinary , Animals , Blood Chemical Analysis/veterinary , Brain/drug effects , Cervical Cord/drug effects , Diterpenes/administration & dosage , Diterpenes/blood , Injections, Intraventricular , Male , Nervous System/pathology , Neurotoxins/administration & dosage , Neurotoxins/blood , Pain Threshold/drug effects , Substance P/metabolism , TRPV Cation Channels/drug effects
7.
Mater Today (Kidlington) ; 21(9): 951-959, 2018 Nov.
Article in English | MEDLINE | ID: mdl-31156331

ABSTRACT

Engineered nerve guidance conduits (NGCs) have been demonstrated for repairing peripheral nerve injuries. However, there remains a need for an advanced biofabrication system to build NGCs with complex architectures, tunable material properties, and customizable geometrical control. Here, a rapid continuous 3D-printing platform was developed to print customizable NGCs with unprecedented resolution, speed, flexibility, and scalability. A variety of NGC designs varying in complexity and size were created including a life-size biomimetic branched human facial NGC. In vivo implantation of NGCs with microchannels into complete sciatic nerve transections of mouse models demonstrated the effective directional guidance of regenerating sciatic nerves via branching into the microchannels and extending toward the distal end of the injury site. Histological staining and immunostaining further confirmed the progressive directional nerve regeneration and branching behavior across the entire NGC length. Observational and functional tests, including the von Frey threshold test and thermal test, showed promising recovery of motor function and sensation in the ipsilateral limbs grafted with the 3D-printed NGCs.

8.
Anesthesiology ; 125(2): 378-94, 2016 08.
Article in English | MEDLINE | ID: mdl-27272672

ABSTRACT

BACKGROUND: Intrathecal infusion of opioids in dogs, sheep, and humans produces local space-occupying masses. To develop a small-animal model, the authors examined effects of intrathecal catheterization and morphine infusion in guinea pigs. METHODS: Under isoflurane, polyethylene or polyurethane catheters were advanced from the cisterna magna to the lumbar enlargement. Drugs were delivered as a bolus through the externalized catheter or continuously by subcutaneous minipumps. Hind paw withdrawal to a thermal stimulus was assessed. Spinal histopathology was systematically assessed in a blinded fashion. To assist in determining catheter placement, ex vivo images were obtained using magnetic resonance imaging in several animals. Canine spinal tissue from previous intrathecal morphine studies was analyzed in parallel. RESULTS: (1) Polyethylene (n = 30) and polyurethane (n = 25) catheters were implanted in the lumbar intrathecal space. (2) Bolus intrathecal morphine produced a dose-dependent (20 to 40 µg/10 µl) increase in thermal escape latencies. (3) Absent infusion, a catheter-associated distortion of the spinal cord and a fibrotic investment were noted along the catheter tract (polyethylene > polyurethane). (4) Intrathecal morphine infusion (25 mg/ml/0.5 µl/h for 14 days) resulted in intrathecal masses (fibroblasts, interspersed collagen, lymphocytes, and macrophages) arising from meninges proximal to the catheter tip in both polyethylene- and polyurethane-catheterized animals. This closely resembles mass histopathology from intrathecal morphine canine studies. CONCLUSIONS: Continuous intrathecal infusion of morphine leads to pericatheter masses that morphologically resemble those observed in dogs and humans. This small-animal model may be useful for studying spinal drug toxicology in general and the biology of intrathecal granuloma formation in particular.


Subject(s)
Analgesics, Opioid/adverse effects , Catheterization/methods , Drug Delivery Systems/methods , Granuloma/chemically induced , Injections, Spinal/methods , Morphine/adverse effects , Spinal Cord Diseases/chemically induced , Animals , Catheters , Cisterna Magna , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Granuloma/pathology , Guinea Pigs , Magnetic Resonance Imaging , Male , Meninges/pathology , Polyethylene , Polyurethanes , Spinal Cord Diseases/pathology
9.
Pain ; 155(4): 674-684, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24333775

ABSTRACT

We addressed the hypothesis that intraplantar botulinum toxin B (rimabotulinumtoxin B: BoNT-B) has an early local effect upon peripheral afferent terminal releasing function and, over time, will be transported to the central terminals of the primary afferent. Once in the terminals it will cleave synaptic protein, block spinal afferent transmitter release, and thereby prevent spinal nociceptive excitation and behavior. In mice, C57Bl/6 males, intraplantar BoNT-B (1 U) given unilaterally into the hind paw had no effect upon survival or motor function, but ipsilaterally decreased: (1) intraplantar formalin-evoked flinching; (2) intraplantar capsaicin-evoked plasma extravasation in the hind paw measured by Evans blue in the paw; (3) intraplantar formalin-evoked dorsal horn substance P (SP) release (neurokinin 1 [NK1] receptor internalization); (4) intraplantar formalin-evoked dorsal horn neuronal activation (c-fos); (5) ipsilateral dorsal root ganglion (DRG) vesicle-associated membrane protein (VAMP); (6) ipsilateral SP release otherwise evoked bilaterally by intrathecal capsaicin; (7) ipsilateral activation of c-fos otherwise evoked bilaterally by intrathecal SP. These results indicate that BoNT-B, after unilateral intraplantar delivery, is taken up by the peripheral terminal, is locally active (blocking plasma extravasation), is transported to the ipsilateral DRG to cleave VAMP, and is acting presynaptically to block release from the spinal peptidergic terminal. The observations following intrathecal SP offer evidence for a possible transsynaptic effect of intraplantar BoNT. These results provide robust evidence that peripheral BoNT-B can alter peripheral and central terminal release from a nociceptor and attenuate downstream nociceptive processing via a presynaptic effect, with further evidence suggesting a possible postsynaptic effect.


Subject(s)
Afferent Pathways/physiology , Anti-Dyskinesia Agents/pharmacology , Botulinum Toxins/pharmacology , Nociceptors/drug effects , Pain Threshold/drug effects , Spinal Cord/metabolism , Afferent Pathways/drug effects , Animals , Botulinum Toxins, Type A , Capsaicin/adverse effects , Functional Laterality/drug effects , Gene Expression Regulation/drug effects , Hindlimb Suspension , Male , Mice , Mice, Inbred C57BL , Muscle Strength/drug effects , Pain/chemically induced , Pain/metabolism , Pain/pathology , Posterior Horn Cells/drug effects , Receptors, Neurokinin-1/metabolism , Spinal Cord/drug effects , Substance P/metabolism , Time Factors
10.
Anesthesiology ; 119(5): 1163-77, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24051388

ABSTRACT

BACKGROUND: Neurokinin-1 receptors (NK1-rs) located on superficial dorsal horn neurons are essential for integration of nociceptive input. Intrathecal injection of substance P-saporin (SP-SAP) leads to local loss of spinal NK1-r (+) neurons suggesting its potential as a therapeutic agent for chronic pain. The authors determined, in a canine model, effects of lumbar intrathecal SP-SAP. METHODS: Distribution of SP-SAP and Saporin was determined in plasma, lumbar cerebrospinal fluid, and tissue. Safety of intrathecal SP-SAP was determined in four groups (six dogs each) administered 0 (0.9% saline), 1.5, 15, or 150 µg SP-SAP through lumbar intrathecal catheters. Behavioral, physiologic, and biochemical variables were assessed. Spinal tissues were collected at 7 and approximately 90 days, or earlier if significant morbidity developed, and analyzed for NK1-r (+) neuron loss and histopathology. RESULTS: SP-SAP and Saporin were detectable in lumbar cerebrospinal fluid for up to 4 and 24 h, respectively. Animals receiving intrathecal saline, 1.5, or 15 µg of SP-SAP showed no persistent neurologic deficits. Three animals receiving 150 µg of SP-SAP developed pelvic limb paraparesis and were euthanized prematurely. Immunohistochemistry and in situ hybridization cell counts confirmed a significant reduction in NK1-r (+) in superficial dorsal horn neurons from lumbar spinal cord after intrathecal administration of 15 and 150 µg of SP-SAP. A significant loss of NK1-r neurons in the lumbar ventral horn occurred only with 150-µg SP-SAP. CONCLUSION: Intrathecal 15-µg SP-SAP reduced dorsal, but not ventral, NK1-r (+) neurons at the spinal level of delivery with minimal side effects, whereas 150-µg SP-SAP resulted in motor neuron toxicity.


Subject(s)
Neurokinin-1 Receptor Antagonists , Ribosome Inactivating Proteins, Type 1/pharmacology , Spinal Cord/metabolism , Substance P/analogs & derivatives , Animals , Behavior, Animal/drug effects , Blood Pressure/drug effects , Body Temperature/drug effects , Body Weight/drug effects , Dogs , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , In Situ Hybridization , Injections, Spinal , Neurologic Examination , Neurotoxicity Syndromes/pathology , Ophthalmoscopy , Phenotype , Receptors, Neurokinin-1/metabolism , Ribosome Inactivating Proteins, Type 1/pharmacokinetics , Ribosome Inactivating Proteins, Type 1/toxicity , Saporins , Spinal Cord/drug effects , Substance P/pharmacokinetics , Substance P/pharmacology , Substance P/toxicity , Tissue Distribution
11.
FASEB J ; 27(5): 1939-49, 2013 May.
Article in English | MEDLINE | ID: mdl-23382512

ABSTRACT

Previously, we observed significant increases in spinal 12-lipoxygenase (LOX) metabolites, in particular, hepoxilins, which contribute to peripheral inflammation-induced tactile allodynia. However, the enzymatic sources of hepoxilin synthase (HXS) activity in rats remain elusive. Therefore, we overexpressed each of the 6 rat 12/15-LOX enzymes in HEK-293T cells and measured by LC-MS/MS the formation of HXB3, 12-HETE, 8-HETE, and 15-HETE from arachidonic acid (AA) at baseline and in the presence of LOX inhibitors (NDGA, AA-861, CDC, baicalein, and PD146176) vs. vehicle-treated and mock-transfected controls. We detected the following primary intrinsic activities: 12-LOX (Alox12, Alox15), 15-LOX (Alox15b), and HXS (Alox12, Alox15). Similar to human and mouse orthologs, proteins encoded by rat Alox12b and Alox12e possessed minimal 12-LOX activity with AA as substrate, while eLOX3 (encoded by Aloxe3) exhibited HXS without 12-LOX activity when coexpressed with Alox12b or supplemented with 12-HpETE. CDC potently inhibited HXS and 12-LOX activity in vitro (relative IC50s: CDC, ~0.5 and 0.8 µM, respectively) and carrageenan-evoked tactile allodynia in vivo. Notably, peripheral inflammation significantly increased spinal eLOX3; intrathecal pretreatment with either siRNA targeting Aloxe3 or an eLOX3-selective antibody attenuated the associated allodynia. These findings implicate spinal eLOX3-mediated hepoxilin synthesis in inflammatory hyperesthesia and underscore the importance of developing more selective 12-LOX/HXS inhibitors.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Hyperalgesia/etiology , Intramolecular Oxidoreductases/metabolism , Animals , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/drug effects , HEK293 Cells , Humans , Lipoxygenase Inhibitors/pharmacology , Male , Rats
12.
Anesthesiology ; 118(3): 664-78, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23426209

ABSTRACT

BACKGROUND: Intrathecal morphine forms granulomas that arise from the adjacent arachnoid membrane. The authors propose that these inflammatory cells exit the meningeal vasculature secondary to meningeal mast cell degranulation. METHODS: Three sets of experiments were accomplished in dogs: (1) ex vivo meningeal mast cell degranulation (histamine release was measured ex vivo from canine dura incubated with opiates); (2) in vivo cutaneous mast cell degranulation (flare areas on the dog abdomen were measured after subcutaneous opiates); and (3) in vivo granuloma pharmacology. Dogs with lumbar intrathecal catheters received infusion of intrathecal saline or intrathecal morphine. Intrathecal morphine dogs received (1) no other treatment (control); (2) twice-daily subcutaneous naltrexone; (3) intrathecal co-infusion of cromolyn; or (4) twice-daily subcutaneous cromolyn for the 24- to 28-day study course. RESULTS: Morphine but not fentanyl evoked dural histamine release, which was blocked by cromolyn but not naloxone. Wheal/flare was produced by subcutaneous morphine, methadone, hydromorphone, but not fentanyl, and was unaffected by naltrexone but prevented by cromolyn. Granulomas occurred in all dogs receiving intrathecal morphine (15 of 15); subcutaneous naltrexone had no effect on granulomas (six of six) but was reduced by concurrent intrathecal cromolyn (zero of five) or twice-daily subcutaneous cromolyn (one of five). CONCLUSIONS: The pharmacology of cutaneous/dural mast cell degranulation and intrathecal granulomas are comparable, not mediated by opioid receptors, and reduced by agents preventing mast cell degranulation. If an agent produces cutaneous mast cell degranulation at concentrations produced by intrathecal delivery, the agent may initiate granulomas.


Subject(s)
Granuloma/chemically induced , Mast Cells/drug effects , Mast Cells/pathology , Meninges/drug effects , Morphine/administration & dosage , Morphine/adverse effects , Administration, Cutaneous , Animals , Dogs , Female , Granuloma/metabolism , Granuloma/pathology , Histamine Release/drug effects , Histamine Release/physiology , Injections, Spinal , Male , Meninges/pathology
13.
Neuromodulation ; 16(5): 459-66; discussion 466, 2013.
Article in English | MEDLINE | ID: mdl-23170763

ABSTRACT

BACKGROUND: We hypothesize that intrathecal (IT) granulomas arising from the IT infusion of several opiates may result from the degranulation of meningeal mast cells (MC). Given functional covariance between cutaneous and meningeal MC, we propose that opioids that do not degranulate cutaneous MC will not produce a granuloma. An opioid meeting this criteria is the phenylpiperadine alfentanil HCl. METHODS: Three experiments were accomplished in dogs. 1) Cutaneous MC degranulation. Flare areas on the dog abdomen were measured after intradermal alfentanil, morphine, or compound 48-80. 2) Dose ranging of analgesic effects of IT alfentanil infusion. Dogs with lumbar IT catheters received continuous infusion for 24 hours of different concentrations (1-20 mg/mL/d) of alfentanil and analgesic effects were assessed. 3) Granuloma inducing effects. Dogs received IT alfentanil (20 mg/mL/d; N = 5; 22-28 days) or morphine (12 mg/mL/d; N = 3; 22-30 days) and spinal cord harvested for histopathology after 22-30 days of infusion. RESULTS: 1) Intradermal morphine (10 mg/mL) and compound 48-80 (1 mg/mL) but not alfentanil at concentrations up to 20 mg/mL produced a cutaneous flare. IT alfentanil infusion produced increases in thermal escape latency at concentrations as low as 2 mg/mL/day. A significant depression of arousal was noted in the dogs receiving 20 mg/mL. Over the 22- to 30-day infusion period, morphine (12 mg/mL/day) resulted in granulomas in all three animals examined whereas IT alfentanil at 20 mg/mL/day failed to initiate a granuloma in any animal. CONCLUSIONS: These results support the hypothesis linking MC degranulation and IT granulomas.


Subject(s)
Alfentanil/administration & dosage , Analgesics, Opioid/administration & dosage , Granuloma/chemically induced , Mast Cells/physiology , Skin/pathology , Analysis of Variance , Animals , Dogs , Dose-Response Relationship, Drug , Injections, Spinal , Morphine/administration & dosage
14.
Anesthesiology ; 116(1): 170-81, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22139590

ABSTRACT

BACKGROUND: Intrathecal methylprednisolone acetate (MPA) has been used in patients with chronic pain syndromes. Its safety has been debated after reports of adverse events. No systematic preclinical evaluation of MPA has been reported. In the current study, the acute and long-term effects of intrathecal MPA on dog spinal tissue was studied with the injectate reformulated to include minimal adjuvants. METHODS: Seventeen dogs were implanted with intrathecal catheters and randomized to three groups: vehicle (lidocaine; 4 dogs), MPA 20 mg/ml (human dose; 7 dogs), and MPA 80 mg/ml (maximum deliverable dose; 6 dogs). In parallel with the human protocols, dogs received four injections at 7-day intervals. Clinical observations and plasma methylprednisolone measurements were done before and at intervals after intrathecal delivery. One week (acute) or 6 weeks (long-term) after the last injection, animals were sacrificed and spinal tissues harvested for histopathology. RESULTS: Other than a brief motor block, no adverse clinical event occurred in any animal. Group A (vehicle) showed minimal histologic changes (median histology-score; acute: 1.3, long-term: 1.0). Group B (MPA 20 mg/ml) had a diffuse inflammatory reaction (acute: 2.0, long-term: 3.0), group C (MPA 80 mg/ml) a severe inflammatory response, with large inflammatory masses (acute: 4.0, long-term: 7.0) The severity of the inflammatory reaction increased significantly with increasing dose at long-term sacrifice (acute P = 0.167, long-term P = 0.014). No neuronal injury, demyelination, or gliosis was seen in any animal. CONCLUSION: These results, showing dose-dependent intrathecal inflammatory reactions at MPA doses and injectate concentrations comparable to those used in humans, indicate that the continued use of this modality in humans is not recommended.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/toxicity , Methylprednisolone/pharmacokinetics , Methylprednisolone/toxicity , Animals , Anti-Inflammatory Agents/administration & dosage , Body Weight/drug effects , Delayed-Action Preparations , Dogs , Dose-Response Relationship, Drug , Female , Inflammation/pathology , Injections, Spinal , Male , Meningitis/chemically induced , Meningitis/pathology , Methylprednisolone/administration & dosage , Neuralgia, Postherpetic/drug therapy , Pain Measurement/drug effects , Paraffin Embedding , Preservatives, Pharmaceutical , Safety
15.
J Neurosci ; 31(6): 2113-24, 2011 Feb 09.
Article in English | MEDLINE | ID: mdl-21307248

ABSTRACT

Phosphinositide 3-kinase (PI3K), Akt, and their downstream kinase, mammalian target of rapamycin (mTOR), are implicated in neural plasticity. The functional linkages of this signaling cascade in spinal dorsal horn and their role in inflammatory hyperalgesia have not been elucidated. In the present work, we identified the following characteristics of this cascade. (1) Local inflammation led to increase in rat dorsal horn phosphorylation (activation) of Akt (pAkt) and mTOR (pmTOR), as assessed by Western blotting and immunocytochemistry. (2) Increased pAkt and pmTOR were prominent in neurons in laminae I, III, and IV, whereas pmTOR and its downstream targets (pS6, p4EBP) were also observed in glial cells. (3) Intrathecal treatment with inhibitors to PI3K or Akt attenuated Formalin-induced second-phase flinching behavior, as well as carrageenan-induced thermal hyperalgesia and tactile allodynia. (4) Intrathecal rapamycin (an mTORC1 inhibitor) displayed anti-hyperalgesic effect in both inflammatory pain models. Importantly, intrathecal wortmannin at anti-hyperalgesic doses reversed the evoked increase not only in Akt but also in mTORC1 signaling (pS6/p4EBP). (5) pAkt and pmTOR are expressed in neurokinin 1 receptor-positive neurons in laminae I-III after peripheral inflammation. Intrathecal injection of Substance P activated this cascade (increased phosphorylation) and resulted in hyperalgesia, both of which effects were blocked by intrathecal wortmannin and rapamycin. Together, these findings reveal that afferent inputs trigged by peripheral inflammation initiate spinal activation of PI3K-Akt-mTOR signaling pathway, a component of which participates in neuronal circuits of facilitated pain processing.


Subject(s)
Hyperalgesia/enzymology , Hyperalgesia/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Spinal Cord/enzymology , TOR Serine-Threonine Kinases/metabolism , Androstadienes/pharmacology , Androstadienes/therapeutic use , Animals , Carrageenan/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Formaldehyde/adverse effects , Gene Expression Regulation, Enzymologic/drug effects , Hyperalgesia/drug therapy , Inflammation/chemically induced , Inflammation/complications , Male , Nerve Tissue Proteins/metabolism , Pain Measurement , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Signal Transduction/drug effects , Sirolimus/metabolism , Sirolimus/pharmacology , Spinal Cord/pathology , Statistics, Nonparametric , Substance P/pharmacology , Time Factors , Wortmannin
16.
Neuroreport ; 21(4): 313-7, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20134354

ABSTRACT

Pharmacological studies indicate that spinal p38 mitogen-activated protein kinase plays a role in the development of hyperalgesia. We investigated whether either the spinal isoform p38alpha or p38beta is involved in peripheral inflammation evoked pain state and increased expression of spinal COX-2. Using intrathecal antisense oligonucleotides, we show that hyperalgesia is prevented by downregulation of p38beta but not p38alpha, whereas increases in spinal COX-2 protein expression at 8 hours are mediated by both p38alpha and beta isoforms. These data suggest that early activation of spinal p38beta isoform may affect acute facilitatory processing, and both p38beta and alpha isoforms mediate temporally delayed upregulation of spinal COX-2.


Subject(s)
Cyclooxygenase 2/metabolism , Hyperalgesia/prevention & control , Pain/metabolism , Spinal Cord/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Carrageenan/administration & dosage , Carrageenan/pharmacology , Cyclooxygenase 1/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Hyperalgesia/chemically induced , Inflammation , Injections, Spinal , MAP Kinase Signaling System/drug effects , Male , Membrane Proteins/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/pharmacology , Pain/chemically induced , Pain/physiopathology , Pain Measurement/drug effects , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/physiopathology , Time Factors
17.
Gastroenterology ; 136(4): 1339-48, e1-2, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19249394

ABSTRACT

BACKGROUND & AIMS: Chronic psychological stress is associated with visceral hyperalgesia and increased expression of spinal NK1 receptors (NK1Rs). We aimed to identify the role of spinal microglia in this process. METHODS: Male Wistar rats were exposed to water avoidance (WA) or sham stress 1 hour each day for 10 days and given daily injections of minocycline, the p38 inhibitor SB203580, or saline. Phosphorylation levels of the kinase p38 (P-p38), the microglia marker OX42, NK1R, and IkappaBalpha were assessed by immunoblotting and/or immunostaining of spinal samples collected at day 11. The visceromotor response to colorectal distention at baseline and following WA were also assayed in rats given injections of minocycline, SB203580, or vehicle. The effects of fractalkine were assessed on the visceromotor response in rats exposed to minocycline or vehicle. RESULTS: P-p38 protein levels and immunoreactivity were increased in stressed rats and colocalized with OX42-positive cells and neurons in the dorsal horn. This increase was reversed by minocycline or SB203580 exposure. Stress-induced increased NK1R expression was blocked by minocycline but not SB203580. WA-induced decreased IkappaBalpha expression was blocked by minocycline and SB203580. WA-induced hyperalgesia was blocked by minocycline and SB203580 intrathecally. Fractalkine-induced hyperalgesia was blocked by minocycline. CONCLUSIONS: This is the first demonstration that stress-induced activation of spinal microglia has a key role in visceral hyperalgesia and associated spinal NK1R up-regulation.


Subject(s)
Hyperalgesia/physiopathology , Microglia/physiology , Receptors, Neurokinin-1/metabolism , Spinal Nerves/physiology , Stress, Physiological/physiology , Up-Regulation/physiology , Viscera/innervation , Animals , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hyperalgesia/metabolism , Imidazoles/pharmacology , Male , Microglia/drug effects , Minocycline/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Receptors, Neurokinin-1/genetics , Spinal Nerves/drug effects , Viscera/metabolism , Viscera/physiopathology , Water Deprivation/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Rehabil Res Dev ; 41(6A): 757-66, 2004.
Article in English | MEDLINE | ID: mdl-15685464

ABSTRACT

Implanted pure titanium fixtures are able to completely integrate with bone, in part because of the formation of a strong extracellular matrix (ECM) bond at the titanium-bone interface. In this study, we used a rodent femur model of intramedullary osseointegration to analyze the changes in immunoreactivity of ECM-controlling matrix metalloproteinases (MMPs), tissue inhibitor of metalloproteinase-3 (TIMP-3), and tumor necrosis factor alpha (TNF-alpha) during osseointegration. We observed dramatic increases in MMP-2, MMP-9, MMP-7, TIMP-3, and TNF-alpha in osteocytes, osteoclasts, haversian canals, and the interface matrix in bone ipsilateral to the titanium implant. An increase in TIMP-3, MMP-9, and MMP-7 in hypertrophied chondrocytes and the vascular component of the epiphysial growth plate was also observed in experimental bone. These findings were not seen in contralateral or sham-operated bone, where the titanium fixtures were threaded into the femur and immediately removed. Our data link titanium-induced bone remodeling to changes in expression and distribution of MMPs.


Subject(s)
Matrix Metalloproteinases/biosynthesis , Osseointegration , Prostheses and Implants , Titanium/pharmacology , Animals , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...