dynorphins has been researched along with Spinal-Cord-Injuries* in 33 studies
7 review(s) available for dynorphins and Spinal-Cord-Injuries
Article | Year |
---|---|
Monoclonal antibodies as novel neurotherapeutic agents in CNS injury and repair.
Central nervous system (CNS) injury is a complex in which numerous neurochemicals and other vasoactive agents actively contribute towards the development of posttraumatic brain pathology and/or repair mechanisms. A focal trauma to the brain or spinal cord releases several endogenous neurodestructive agents within the CNS, resulting in adverse cellular reactions. Our laboratory is engaged in identifying these endogenous neurodestructive signals in the CNS following injury caused by trauma or hyperthermia. Our observations show that serotonin (5-HT), dynorphin A (Dyn A 1-17), nitric oxide synthase (NOS), and tumor necrosis factor-α (TNF-α) could be potential neurodestructive signals in the CNS injury. Thus, neutralization of these agents using monoclonal antibodies directed against 5-HT, NOS, Dyn A (1-17), and TNF-α in vivo will result in marked neuroprotection and enhance neurorepair after trauma. In addition, a suitable combination of monoclonal antibodies, for example, NOS and TNF-α, when applied 60-90 min after trauma, is capable to enhance neuroprotective ability and thwart cell and tissue injury after spinal cord insult. Taken together, our novel observations suggest a potential use of monoclonal antibodies as suitable therapeutic agents in CNS injuries to achieve neuroprotection and/or neurorepair. Topics: Animals; Antibodies, Monoclonal; Antigen-Antibody Reactions; Brain Injuries; Disease Models, Animal; Dynorphins; History, 18th Century; History, 19th Century; History, 20th Century; Humans; Nerve Regeneration; Neuroprotective Agents; Nitric Oxide Synthase Type I; Serotonin; Spinal Cord Injuries; Tumor Necrosis Factor-alpha; Wound Healing | 2012 |
Pathobiology of dynorphins in trauma and disease.
Dynorphins, endogenous opioid neuropeptides derived from the prodynorphin gene, are involved in a variety of normative physiologic functions including antinociception and neuroendocrine signaling, and may be protective to neurons and oligodendroglia via their opioid receptor-mediated effects. However, under experimental or pathophysiological conditions in which dynorphin levels are substantially elevated, these peptides are excitotoxic largely through actions at glutamate receptors. Because the excitotoxic actions of dynorphins require supraphysiological concentrations or prolonged tissue exposure, there has likely been little evolutionary pressure to ameliorate the maladaptive, non-opioid receptor mediated consequences of dynorphins. Thus, dynorphins can have protective and/or proapoptotic actions in neurons and glia, and the net effect may depend upon the distribution of receptors in a particular region and the amount of dynorphin released. Increased prodynorphin gene expression is observed in several disease states and disruptions in dynorphin processing can accompany pathophysiological situations. Aberrant processing may contribute to the net negative effects of dysregulated dynorphin production by tilting the balance towards dynorphin derivatives that are toxic to neurons and/or oligodendroglia. Evidence outlined in this review suggests that a variety of CNS pathologies alter dynorphin biogenesis. Such alterations are likely maladaptive and contribute to secondary injury and the pathogenesis of disease. Topics: Animals; Apoptosis; Dynorphins; Evolution, Molecular; Gene Expression Regulation; Humans; Neuroglia; Neurons; Neuropeptides; Receptors, Opioid; Spinal Cord Injuries | 2005 |
Influence of dynorphin A antibodies on the formation of edema and cell changes in spinal cord trauma.
Topics: Animals; Dynorphins; Edema; Neurons; Rats; Spinal Cord Injuries | 1995 |
Central non-opioid physiological and pathophysiological effects of dynorphin A and related peptides.
Dynorphin A (Dyn A) and related opioid peptides derived from prodynorphin possess a high affinity for kappa opioid receptors, but they also bind to other opioid receptors (mu and delta) as well as to some non-opioid receptor sites. Although the physiological role of these peptides is not well established, recent experimental data pinpoint their particular involvement in physiological and pathophysiological conditions that relate to algesia, spinal cord injury and epilepsy. In this paper, we review data which support the concept that the non-opioid behavioral effects of Dyn A and related endogenous peptides which are observed under these conditions are physiologically and pathophysiologically relevant. Topics: Brain; Dynorphins; Epilepsy; Humans; Neuropeptides; Nociceptors; Receptors, N-Methyl-D-Aspartate; Receptors, Opioid; Spinal Cord; Spinal Cord Injuries | 1992 |
[Opioid peptides in spinal injury].
Topics: Animals; Calcium Channels; Dynorphins; Humans; Naloxone; Receptors, N-Methyl-D-Aspartate; Spinal Cord Injuries | 1992 |
Neuropeptides and central nervous system injury. Clinical implications.
It has been proposed that endogenous opioids play a pathophysiologic role in the secondary injury that follows spinal trauma, brain trauma, and cerebral ischemia. Opiate antagonists, at high doses, have been found to improve outcome in various experimental models of central nervous system injury. Thyrotropin-releasing hormone, which appears to act in part as a functional antagonist of opioid systems, has proved effective in the treatment of experimental spinal cord and brain trauma. The literature relating to these developments is reviewed, with emphasis on the potential clinical application of these classes of substances. Topics: Animals; Cats; Central Nervous System Diseases; Dynorphins; Humans; Ischemic Attack, Transient; Naloxone; Narcotic Antagonists; Narcotics; Rabbits; Spinal Cord Injuries; Thyrotropin-Releasing Hormone | 1986 |
Endogenous opioids in spinal cord injury: a critical evaluation.
Based upon evidence that opioid antagonists improve neurological outcome following either traumatic or ischemic spinal cord injury, endogenous opioids have been implicated in the pathophysiology of these disorders. Naloxone improved both spinal cord perfusion and neurological function following traumatic spinal cord injury in cats, and was subsequently observed to improve neurological outcome following ischemic spinal cord injury in rabbits. Using several opioid antagonists with varied selectivities for different types of opioid receptors, it was suggested that kappa opioid receptors are involved in both these models of spinal cord injury. In addition, spinal cord trauma in rats is associated with increased concentrations of the endogenous kappa agonist dynorphin A, and increased kappa opioid receptor binding capacity localized to the injury site. Furthermore, dynorphin A induces hindlimb and tail flaccidity following intrathecal injection in rats. Thus, the pathophysiological effects of endogenous opioids in spinal cord injury have been proposed to involve dynorphin A interactions with kappa opioid receptors. However, disparities between the actions of intrathecally injected dynorphin A in rats and the presumed actions of endogenous dynorphin A in cat and rabbit spinal cord injury have been revealed in recent experiments. Paralysis resulting from intrathecal dynorphin A is not altered by opioid receptor antagonists or TRH, produced by non-opioid dynorphin A fragments but not by other selective kappa opioid agonists, and associated with non-opioid mediated reductions in spinal cord blood flow. Furthermore, despite reports of endogenous opioid changes following rat spinal cord trauma, in contrast to cats and rabbits, naloxone failed to improve neurological outcome following traumatic rat spinal cord injury. Thus, the specific endogenous opioids and opioid receptor types involved in spinal cord injury remain to be resolved, and do not appear to be universal among different models of spinal cord injury in different species. Additionally, dynorphin A may participate in spinal cord injury mechanisms in the rat through non-opioid actions. Topics: Animals; Blood Pressure; Dynorphins; Endorphins; Injections, Spinal; Naloxone; Nerve Degeneration; Receptors, Opioid; Spinal Cord; Spinal Cord Injuries | 1986 |
26 other study(ies) available for dynorphins and Spinal-Cord-Injuries
Article | Year |
---|---|
Morphine-induced changes in the function of microglia and macrophages after acute spinal cord injury.
Opioids are among the most effective and commonly prescribed analgesics for the treatment of acute pain after spinal cord injury (SCI). However, morphine administration in the early phase of SCI undermines locomotor recovery, increases cell death, and decreases overall health in a rodent contusion model. Based on our previous studies we hypothesize that morphine acts on classic opioid receptors to alter the immune response. Indeed, we found that a single dose of intrathecal morphine increases the expression of activated microglia and macrophages at the injury site. Whether similar effects of morphine would be seen with repeated intravenous administration, more closely simulating clinical treatment, is not known.. To address this, we used flow cytometry to examine changes in the temporal expression of microglia and macrophages after SCI and intravenous morphine. Next, we explored whether morphine changed the function of these cells through the engagement of cell-signaling pathways linked to neurotoxicity using Western blot analysis.. Our flow cytometry studies showed that 3 consecutive days of morphine administration after an SCI significantly increased the number of microglia and macrophages around the lesion. Using Western blot analysis, we also found that repeated administration of morphine increases β-arrestin, ERK-1 and dynorphin (an endogenous kappa opioid receptor agonist) production by microglia and macrophages.. These results suggest that morphine administered immediately after an SCI changes the innate immune response by increasing the number of immune cells and altering neuropeptide synthesis by these cells. Topics: Analgesics; Analgesics, Opioid; Animals; beta-Arrestins; Dynorphins; Macrophages; Microglia; Morphine; Rats; Rats, Sprague-Dawley; Receptors, Opioid, kappa; Recovery of Function; Spinal Cord; Spinal Cord Injuries | 2022 |
Neuropathic pain and temporal expression of preprodynorphin, protein kinase C and N-methyl-D-aspartate receptor subunits after spinal cord injury.
Central neuropathic pain is refractory to conventional treatment and thus remains a therapeutic challenge. In this work, we used a well-recognized model of central neuropathic pain to evaluate time-dependent expression of preprodynorphin (ppD), protein kinase C gamma (PKCgamma) and NMDA receptor (NMDAR) subunits NR1, NR2A and NR2B, all critical players in nociceptive processing at the spinal level. Male Sprague-Dawley rats were subjected to spinal hemisection at T13 level and sham-operated rats were included as control animals. The development of hindpaw mechanical allodynia was assessed using the von Frey filaments test. Real time RT-PCR was employed to determine the relative mRNA levels of NMDAR subunits, ppD and PKCgamma in the dorsal spinal cord 1, 14 and 28 days after injury. Our results show that, coincident with the allodynic phase after injury, there was a strong up-regulation of the mRNAs coding for ppD, PKCgamma and NMDAR subunits in the dorsal spinal cord caudal to the injury site. The present study provides further evidence that these molecules are involved in the development/maintenance of central neuropathic pain and thus could be the target of therapeutic approaches. Topics: Analysis of Variance; Animals; Disease Models, Animal; Dynorphins; Functional Laterality; Hyperalgesia; Male; Pain Measurement; Pain Threshold; Protein Kinase C; Protein Precursors; Protein Subunits; Rats; Rats, Sprague-Dawley; Receptors, N-Methyl-D-Aspartate; RNA, Messenger; Spinal Cord Injuries; Time Factors | 2008 |
Caspase-3 activity is reduced after spinal cord injury in mice lacking dynorphin: differential effects on glia and neurons.
Dynorphins are endogenous opioid peptide products of the prodynorphin gene. An extensive literature suggests that dynorphins have deleterious effects on CNS injury outcome. We thus examined whether a deficiency of dynorphin would protect against tissue damage after spinal cord injury (SCI), and if individual cell types would be specifically affected. Wild-type and prodynorphin(-/-) mice received a moderate contusion injury at 10th thoracic vertebrae (T10). Caspase-3 activity at the injury site was significantly decreased in tissue homogenates from prodynorphin(-/-) mice after 4 h. We examined frozen sections at 4 h post-injury by immunostaining for active caspase-3. At 3-4 mm rostral or caudal to the injury, >90% of all neurons, astrocytes and oligodendrocytes expressed active caspase-3 in both wild-type and knockout mice. At 6-7 mm, there were fewer caspase-3(+) oligodendrocytes and astrocytes than at 3-4 mm. Importantly, caspase-3 activation was significantly lower in prodynorphin(-/-) oligodendrocytes and astrocytes, as compared with wild-type mice. In contrast, while caspase-3 expression in neurons also declined with further distance from the injury, there was no effect of genotype. Radioimmunoassay showed that dynorphin A(1-17) was regionally increased in wild-type injured versus sham-injured tissues, although levels of the prodynorphin processing product Arg(6)-Leu-enkephalin were unchanged. Our results indicate that dynorphin peptides affect the extent of post-injury caspase-3 activation, and that glia are especially sensitive to these effects. By promoting caspase-3 activation, dynorphin peptides likely increase the probability of glial apoptosis after SCI. While normally beneficial, our findings suggest that prodynorphin or its peptide products become maladaptive following SCI and contribute to secondary injury. Topics: Animals; Apoptosis; Caspase 3; Down-Regulation; Dynorphins; Enzyme Activation; Female; Gene Expression Regulation, Enzymologic; Gliosis; Mice; Mice, Inbred C57BL; Mice, Knockout; Nerve Degeneration; Nerve Regeneration; Neuroglia; Neurons; Recovery of Function; Spinal Cord Injuries | 2007 |
Topical application of dynorphin A (1-17) antibodies attenuates neuronal nitric oxide synthase up-regulation, edema formation, and cell injury following focal trauma to the rat spinal cord.
Previous investigations from our laboratory show that up-regulation of neuronal nitric oxide synthase (NOS) following spinal cord injury (SCI) is injurious to the cord. Antiserum to dynorphin A (1-17) induces marked neuroprotection in our model of SCI, indicating an interaction between dynorphin and NOS regulation. The present investigation was undertaken to find out whether topical application of dynorphin A (1-17) antiserum has some influence on neuronal NOS up-regulation in the traumatized spinal cord. SCI was produced in anesthetized animals by making a unilateral incision into the right dorsal horn of the T10-11 segments. The antiserum to dynorphin A (1-17) was applied (1 : 20, 20 microL in 10 seconds) 5 minutes after trauma over the injured spinal cord and the rats were allowed to survive 5 hours after SCI. Topical application of dynorphin A (1-17) antiserum significantly attenuated neuronal NOS up-regulation in the adjacent T9 and T12 segments. In the antiserum-treated group, spinal cord edema and cell injury were also less marked. These observations provide new evidence that the opioid active peptide dynorphin A may be involved in the mechanisms underlying NOS regulation in the spinal cord after injury, and confirms our hypothesis that up-regulation of neuronal NOS is injurious to the cord. Topics: Animals; Antibodies; Dynorphins; Edema; Male; Neuroprotective Agents; Nitric Oxide Synthase Type I; Rats; Rats, Sprague-Dawley; Spinal Cord Injuries; Treatment Outcome; Up-Regulation | 2006 |
Topical application of dynorphin A (1-17) antiserum attenuates trauma induced alterations in spinal cord evoked potentials, microvascular permeability disturbances, edema formation and cell injury: an experimental study in the rat using electrophysiologic
Dynorphin is a neuropeptide that is present in high quantities in the dorsal horn of the spinal cord. The peptide is actively involved in pain processing pathways. However, its involvement in spinal cord injury is not well known. Alteration in dynorphin immunoreactivity occurs following a focal trauma to the rat spinal cord. Infusion of dynorphin into the intrathecal space of the cord results in ischemia, cell damage and abnormal motor function. Antibodies to dynorphin when injected into the intrathecal space of the spinal cord following trauma improve motor recovery, reduce edema and cell changes. However, influence of dynorphin on trauma induced alteration in spinal cord bioelectrical activity is still not known. Spinal cord evoked potentials (SCEP) are good indicator of spinal cord pathology following trauma. Therefore, in present investigation, influence of dynorphin antibodies on trauma induced changes in SCEP were examined in our rat model. In addition, spinal cord edema formation, microvascular permeability disturbances and cell injury were also investigated. Our results show that topical application of dynorphin antiserum (1 : 200) two min before injury markedly attenuated the SCEP changes immediately after injury. In the antiserum treated animals, a significant reduction in the microvascular permeability, edema formation and cell injury was observed in the traumatised spinal cord. These observations suggest that (i). dynorphin is involved in the altered bioelectrical activity of the spinal cord following trauma, (ii). the peptide actively participates in the pathophysiological processes of cell injury in the spinal cord trauma, and (iii). the dynorphin antiserum has potential therapeutic value for the treatment of spinal cord injuries. Topics: Animals; Antibodies; Blood-Brain Barrier; Capillary Permeability; Dynorphins; Edema; Evoked Potentials; Male; Neurons; Rats; Rats, Sprague-Dawley; Spinal Cord; Spinal Cord Injuries | 2002 |
Spinal and supraspinal changes in opioid mRNA expression are related to the onset of pain behaviors following excitotoxic spinal cord injury.
Excitotoxic spinal cord injury (SCI) causes anatomic, physiologic and molecular changes within the spinal cord and brain. Intraspinal injection of quisqualic acid (QUIS) produces an excitotoxic injury that leads to the onset of behavioral syndromes, believed to be related to the clinical condition of chronic pain. The opioid system, classically involved in the suppression of pain transmission, has been associated with the onset of pain-related behaviors and changes in spinal opioid peptide expression have been demonstrated in various models of SCI and chronic pain. Recently, changes in opioid peptide expression have been demonstrated in both spinal and supraspinal areas following excitotoxic SCI. Therefore, the purpose of this study was to examine changes in opioid peptide gene expression as they relate to the onset of pain behaviors following excitotoxic SCI. Male, Long-Evans rats were given an intraspinal injection of 1.2 microl of 125 mM QUIS and allowed to survive for 10 days, a duration sufficient for the development of pain-related behaviors. Animals were assessed daily for the presence of excessive grooming behavior, i.e. self-directed biting and scratching resulting in damage to superficial and deeper layers of the skin. Animals were also tested for thermal hypersensitivity using a cold plate apparatus on days 5, 7, and 10 following QUIS injection. After sacrifice, quantitative in situ hybridization was performed on regions of the spinal cord surrounding the lesion site as well as whole brain sections through various levels of the thalamus and cortex. Spinal preproenkephalin (PPE) and preprodynorphin (PPD) expression was significantly increased in animals that developed excessive grooming behaviors vs. those that did not. For PPE, this difference was seen bilaterally, in areas of cord caudal to the site of injury. For PPD, this difference was seen only ipsilateral to the site of injection, rostral to the site of injury. In addition, PPE expression in the anterior cingulate cortex and PPD expression in the contralateral parietal cortex were significantly higher in grooming vs. non-grooming animals. These results support previous conclusions that both spinal and supraspinal regulation of endogenous opioid peptide expression plays a role in the response to or onset of post-SCI pain. These results also suggest that the opioid peptides are regulated independently and serve different functions in response to SCI. Topics: Animals; Brain; Dynorphins; Enkephalins; Excitatory Amino Acid Agonists; Gene Expression; Grooming; Male; Opioid Peptides; Pain; Protein Precursors; Quisqualic Acid; Rats; Rats, Long-Evans; RNA, Messenger; Spinal Cord; Spinal Cord Injuries | 2001 |
Expression of c-fos mRNA is increased and related to dynorphin mRNA expression following excitotoxic spinal cord injury in the rat.
Previous studies have demonstrated that excitotoxic spinal cord injury (SCI) created by the intraspinal injection of quisqualic acid (QUIS) is capable of inducing opioid peptide gene expression within the spinal cord and cortex. The opioids are classically involved in the suppression of pain transmission but specifically, dynorphin, has been implicated in the secondary pathophysiologic response to SCI. Activation of the immediate early gene, c-fos, has been implicated in the induction of preprodynorphin (PPD) gene expression and therefore, may be an important intermediate step in the generation of the opioid response to SCI. The purpose of this study was to investigate whether intraspinal QUIS injection induces c-fos expression within the spinal cord. Male, Long-Evans, adult rats (n=5) received an intraspinal injection of 1.2 microl of 125 mM QUIS directed at spinal segments T12-L2. Four hours post-injection brain and spinal cord tissues were removed and processed for in situ hybridization. Integrated density of c-fos and PPD mRNA expression was increased in the spinal dorsal horn following QUIS injection as compared to sham-injected animals. This indicates that SCI rapidly induces c-fos and PPD expression and suggests that c-fos plays a role in the induction of PPD expression. Topics: Animals; Dynorphins; Excitatory Amino Acid Agonists; Gene Expression Regulation; Male; Neurotoxins; Nociceptors; Pain; Posterior Horn Cells; Protein Precursors; Proto-Oncogene Proteins c-fos; Quisqualic Acid; Rats; Rats, Long-Evans; RNA, Messenger; Spinal Cord Injuries; Up-Regulation | 2001 |
The role of kainic acid/AMPA and metabotropic glutamate receptors in the regulation of opioid mRNA expression and the onset of pain-related behavior following excitotoxic spinal cord injury.
Intraspinal injection of quisqualic acid, a mixed kainic acid/2-amino-3(3-hydroxy-5-methylisoxazol-4-yl)propionic acid and metabotropic glutamate receptor agonist, produces an excitotoxic injury that leads to the onset of both spontaneous and evoked pain behavior as well as changes in spinal and cortical expression of opioid peptide mRNA, preprodynorphin and preproenkephalin. What characteristics of the quisqualic acid-induced injury are attributable to activation of each receptor subtype is unknown. This study attempted to define the role of activation of the kainic acid/2-amino-3(3-hydroxy-5-methylisoxazol-4-yl)propionic acid (AMPA) and metabotropic glutamate receptor subtypes in the regulation of opioid peptide expression and the onset of spontaneous and evoked pain-related behavior following excitotoxic spinal cord injury by comparing quisqualic acid-induced changes with those created by co-injection of quisqualic acid and the kainic acid/AMPA antagonist, 2,3-dihydroxy-6-nitro-7-sulfamoylbenzo[f]quinoxaline, (NBQX) or the metabotropic antagonist, (RS)-1-aminoindan-1,5-dicarboxylic acid (AIDA). Therefore, 42 male Long-Evans adult rats were divided into seven treatment groups and received intraspinal microinjections of saline (sham), 0.5% dimethylsulphoxide (sham), quisqualic acid (1.2 microl, 125 mM), NBQX (1.2 microl, 60 microM), AIDA (1.2 microl, 250 microM), quisqualic acid/NBQX (1.2 microl, 125 mM/60 microM), or quisqualic acid/AIDA (1.2 microl, 125 mM/250 microM) directed at spinal levels thoracic 12-lumbar 2. Behavioral observations of spontaneous and evoked pain responses were completed following surgery. After a 10-day survival period, animals were killed and brain and spinal cord tissues were removed and processed for histologic analysis and in situ hybridization. Both AIDA and NBQX affected the quisqualic acid-induced total lesion volume but only AIDA caused a decrease in the percent tissue damage at the lesion epicenter. Preprodynorphin and preproenkephalin expression is increased in both spinal and cortical areas in quisqualic acid-injected animals versus sham-, NBQX or AIDA-injected animals. NBQX did not affect quisqualic acid-induced spinal or cortical expression of preprodynorphin or preproenkephalin except for a significant decrease in preproenkephalin expression in the spinal cord. In contrast, AIDA significantly decreases quisqualic acid-induced preprodynorphin and preproenkephalin expression within the spinal cord and cortex. AIDA, b Topics: Animals; Behavior, Animal; Dynorphins; Enkephalins; Excitatory Amino Acid Agonists; Excitatory Amino Acid Antagonists; Grooming; Indans; Male; Nerve Degeneration; Neurons; Neurotoxins; Opioid Peptides; Pain; Pain Measurement; Protein Precursors; Quinoxalines; Rats; Rats, Long-Evans; Receptors, AMPA; Receptors, Glutamate; Receptors, Kainic Acid; Receptors, Metabotropic Glutamate; RNA, Messenger; Spinal Cord; Spinal Cord Injuries | 2001 |
[Effects of high dose of dynorphin on NMDA receptor and NOS activities in spinal cord of rats].
To elucidate the effects of N-methyl-D-aspartate(NMDA) receptor and nitric oxide synthase (NOS) activity in dynorphin (Dyn)-induced spinal cord injury.. The NMDA receptor activity was measured by radio-ligand of 3H-MK801. The constitutive and inducible NOS (cNOS and iNOS) activities were assayed by 3H-arginine conversion.. In ventral samples, both 3H-MK801 binding and cNOS activity increased at 0.5 h and persisted for 48 h while iNOS activity enhanced at 4 h after intratheacal injection (i.t.) Dyn A(1-17) at dose of 20 nmol/L. However, the 3H-MK801 binding activity reduced significantly from 4 h to 24 h and cNOS activity did not change at the same time in dorsal samples. 7-nitroindozol (7-NI) and aminoguanidine (AG) inhibited the effects of Dyn A(1-17) (20 nmol/L) on 3H-MK801 binding and NOS activities in ventral samples. N-nitro-L-arginine methyl ester (L-NAME) did not affect the elevation of Dyn A(1-17) on NOS activities but caused 3H-MK801 binding activity reduction in ventral samples.. NMDA-NOS pathway might play important role in Dyn spinal neurotoxicity. NOS inhibitors and Dyn might produce cooperative down-regulation on the function of NMDA-NOS pathway in dorsal cord. Topics: Animals; Dose-Response Relationship, Drug; Dynorphins; Enzyme Inhibitors; Female; Male; Nitric Oxide Synthase; Rats; Rats, Wistar; Receptors, N-Methyl-D-Aspartate; Spinal Cord; Spinal Cord Injuries | 2000 |
Constitutive and inducible nitric oxide synthases after dynorphin-induced spinal cord injury.
It has recently been demonstrated that selective inhibition of both neuronal constitutive and inducible nitric oxide synthases (ncNOS and iNOS) is neuroprotective in a model of dynorphin (Dyn) A(1-17)-induced spinal cord injury. In the present study, various methods including the conversion of 3H-L-arginine to 3H-citrulline, immunohistochemistry and in situ hybridization are employed to determine the temporal profiles of the enzymatic activities, immunoreactivities, and mRNA expression for both ncNOS and iNOS after intrathecal injection of a neurotoxic dose (20 nmol) of Dyn A(1-17). The expression of ncNOS immunoreactivity and mRNA increased as early as 30 min after injection and persisted for 1-4 h. At 24-48 h, the number of ncNOS positive cells remained elevated while most neurons died. The cNOS enzymatic activity in the ventral spinal cord also significantly increased at 30 min 48 h, but no significant changes in the dorsal spinal cord were observed. However, iNOS mRNA expression increased later at 2 h, iNOS immunoreactivity and enzymatic activity increased later at 4 h and persisted for 24-48 h after injection of 20 nmol Dyn A(1-17). These results indicate that both ncNOS and iNOS are associated with Dyn-induced spinal cord injury, with ncNOS predominantly involved at an early stage and iNOS at a later stage. Topics: Animals; Dynorphins; Gene Expression Regulation, Enzymologic; Immunohistochemistry; Nitric Oxide Synthase; Nitric Oxide Synthase Type I; Nitric Oxide Synthase Type II; Rats; Rats, Wistar; Reference Values; RNA, Messenger; Spinal Cord; Spinal Cord Injuries; Transcription, Genetic | 2000 |
Opioid peptide messenger RNA expression is increased at spinal and supraspinal levels following excitotoxic spinal cord injury.
Spinal cord injury in rats is known to cause anatomical, physiological and molecular changes within the spinal cord. These changes may account for behavioral syndromes that appear following spinal cord injury, syndromes believed to be related to the clinical condition of chronic pain. Intraspinal injection of quisqualic acid produces an excitotoxic injury with pathological characteristics similar to those associated with ischemic and traumatic spinal cord injury. In addition, recent studies have demonstrated changes in blood flow, neuronal excitability and gene expression in the brain following excitotoxic injury, indicating that behavioral changes may result from modification of neuronal substrates at supraspinal levels of the neuraxis. Because changes in spinal opioid peptide expression have been demonstrated in models of traumatic spinal cord injury and chronic pain, the present study investigated messenger RNA expression of the opioid peptides, preproenkephalin and preprodynorphin, at spinal and supraspinal levels following excitotoxic spinal cord injury. Male, Long-Evans rats were given three intraspinal injections of quisqualic acid (total 1.2 microl, 125mM). After one, three, five, seven or 10days, animals were killed and quantitative in situ hybridization performed on regions of the spinal cord surrounding the lesion site, as well as whole-brain sections through various levels of the thalamus. Preproenkephalin and preprodynorphin expression was increased in spinal cord areas adjacent to the site of quisqualic injection and in cortical regions associated with nociceptive function, preproenkephalin in the cingulate cortex and preprodynorphin in the parietal cortex, both ipsilaterally and contralaterally at various time-points following injury. These results further our knowledge of the secondary events that occur following spinal cord injury, specifically implicating supraspinal opioid systems in the CNS response to spinal cord injury. Topics: Animals; Dynorphins; Enkephalins; Excitatory Amino Acid Agonists; Lumbar Vertebrae; Male; Opioid Peptides; Protein Precursors; Quisqualic Acid; Rats; Rats, Long-Evans; RNA, Messenger; Spinal Cord Injuries; Thoracic Vertebrae | 2000 |
Dynorphin mRNA expression in dorsal horn neurons after traumatic spinal cord injury: temporal and spatial analysis using in situ hybridization.
Dynorphin, an endogenous opioid, may contribute to secondary nervous tissue damage following spinal cord injury. The temporal and spatial distribution of preprodynorphin (PPD) mRNA expression in the injured rat spinal cord was examined by in situ hybridization. Rats were subjected to traumatic spinal cord injury at the T13 spinal segment using the weight-drop method. Motor function of these rats was evaluated by their ability to maintain their position on an inclined plane. Two double-labeling experiments revealed that increased PPD mRNA and dynorphin peptide expression were found exclusively in dorsal horn neurons. Neurons exhibiting an increase in the level of PPD mRNA were concentrated in the superficial laminae and the neck of dorsal horn within several spinal segments from the epicenter of the injury at 24 and 48 h after injury. A number of neurons showing increased PPD mRNA were found in gray matter adjacent to the injury areas. Segments caudal to the injury site exhibited a long-lasting elevation of PPD mRNA in neurons, compared to the rostral segments. The number of neurons expressing PPD mRNA in each rat was significantly positively correlated with its motor dysfunction. These findings suggest that increased expression of dynorphin mRNA and peptide in dorsal horn neurons occurs after traumatic spinal cord injury. This also supports the hypothesis that the dynorphin has a pathological role in secondary tissue damage and neurological dysfunction after spinal cord injury. Topics: Animals; Autoradiography; Disease Models, Animal; Disease Progression; Dynorphins; Gene Expression; In Situ Hybridization; Male; Microscopy, Confocal; Neurons; Photomicrography; Protein Precursors; Rats; Rats, Sprague-Dawley; RNA, Messenger; Spinal Cord; Spinal Cord Injuries; Time Factors | 1998 |
[Effect of neurotoxin on spinal cord injury in rats].
To elucidate the effect of neurotoxin, excitatory amino acid and neuropeptide on spinal cord injury post trauma.. We detected the changes of concentration of EAA, glutamate(Glu) and aspartate(Asp) at injured segment tissue after spinal cord injury. Changes in dynorphin DynA1-13, an endogenous opioid, immnoreactivity following traumatic spinal cord injury in the rat were examined. We observed the effects of Glu and dynorphin DynA1-13 administered intrathecally on function of spinal cord in rats and the effects of 3-(2-carboxypiperazin-4-yl)propyl-1-phosphonic acid (CPP), an antagonist of EAA receptor, and antisera against dynorphin administered intrathecally on secondary spinal cord injury.. The concentrations of EAA and dynorphin DynA1-13 increased progressively along with the severity of injury, and were related to injury time. Glu exacerbated the extent of spinal cord injury. Dynorphin DynA1-13 administered intrathecally caused dose-related hindlimb paraplysis in rats, whereas CPP and antisera against dynorphin DynA1-13 were found to have protective effect on spinal cord injury.. These findings further support the potential pathophysiological role of EAA and dynorphin DynA1-13 as neurotoxin in spinal cord injury. Topics: Animals; Dynorphins; Excitatory Amino Acids; Female; Male; Neurotoxins; Rats; Rats, Sprague-Dawley; Spinal Cord Injuries | 1998 |
[Effects of dynorphin A1-17 on the activities, immunoreactivities and mRNA expression of cNOS and iNOS in rat spinal cord and their mechanisms].
Intrathecal administration(i.t.) of Dynorphin A1-17(Dyn) 1.25-20 nmol produced dose-dependent paralysis of hindlimbs and tail as well as inhibition of tail flick and foot flinch reflexes. The Dyn spinal neurotoxicity and antinociception involve two differential mechanisms: Enhancement of NMDA-Ca(2+)-NOS/NO pathway and c-fos over-expression in the ventral spinal cord for neurotoxicity, and depression of NMDA receptor and NOS activities in the dorsal spinal cord for antinociception. Both brain-derived constitutive NOS (predominant at early stage) and inducible NOS (at later stage) are detrimental, but endothelial constitutive NOS might be beneficial to Dyn spinal neurotoxicity. Dyn exerts a dualistic modulatory effect on [Ca2+]i of the cultured rat single spinal neurons, inducing sustained overload of intracellular free calcium via both NMDA and kappa receptor activation at higher concentrations, and producing significant inhibition of the depolarization-evoked calcium influx only via kappa receptor activation at lower concentrations. Dyn exposure for 1 h produced direct neurotoxicity in the cultured spinal neurons within an optimal range of concentrations. Topics: Analgesics; Animals; Dynorphins; Nitric Oxide Synthase; Nitric Oxide Synthase Type II; Nitric Oxide Synthase Type III; Rats; RNA, Messenger; Spinal Cord; Spinal Cord Injuries | 1997 |
[Receptor mechanism of dynorphin A in rat secondary spinal cord injuries].
To clarify the effects of dynorphin A on secondary spinal cord injuries.. Spinal cord injured rat model made according to modified Allen's method was used by comparing the effect of intrathecal dynorphin A antiserum and that of its receptor antagonist nor-BNI.. The recovery of the muscle tension and motor function of the hindlimb in dynorphin antiserum group was markedly faster than that in both control and nor-BNI groups. Also, the recovery of motor function in the nor-BNI group was favorable at the early stage of injury, compared to the control group. Pathological observation showed that the residual area of the spinal cord in dynorphin A1-13 antiserum group was larger than that in both control and nor-BNI groups. The residual area of the spinal cord in the nor-BNI group was also larger than that in the control group.. These observations support the hypothesis that dynorphin contributes to certain pathophysiological changes following traumatic SCI through both opiate-receptor mediated and nonopioid mechanisms. Topics: Animals; Dynorphins; Immune Sera; Injections, Spinal; Male; Naltrexone; Peptide Fragments; Rats; Rats, Sprague-Dawley; Receptors, Opioid, kappa; Spinal Cord Injuries | 1996 |
Changes in the content of immunoreactive dynorphin in dorsal and ventral spinal cord of the rat in three different conditions.
Previous study in our group demonstrated that dynorphin exerted a significant analgesic effect in spinal cord, and electroacupuncture (EA) of high frequency (100 Hz) produced analgesia which was mediated by dynorphin released in spinal cord. However, no data are up to nowadays available for demonstrating either from anterior horn or from dorsal horn the dynorphin comes. Using radioimmunoassay, we found in the present study that ir-dynorphin content in dorsal horn was 10 times more than in anterior horn, which were 11.24 +/- 0.91 pmol/10 mg protein and 1.08 +/- 0.20 pmol/10 mg protein, respectively. Stimulation of 100 Hz EA for 30 min produced a significant increase of ir-dynorphin content in dorsal horn, in contrast, no change of ir-dynorphin content was found in anterior horn. For the meanwhile, a significant elevation of ir-dynorphin was induced by 100 Hz EA in cerebrospinal fluid. It is concluded according to these data that EA, as a stimulation in physiological condition, elevates the content of ir-dynorphin only in dorsal horn, and induces release of dynorphin in loci. The released dynorphin acts on dorsal horn to mediate the analgesia of EA. Topics: Animals; Dynorphins; Electroacupuncture; Immunohistochemistry; Injections; Male; Nerve Tissue Proteins; Radioimmunoassay; Rats; Rats, Wistar; Spinal Cord; Spinal Cord Injuries; Subarachnoid Space | 1995 |
[Effects of intrasubarachnoid space injection of anti dynorphin A1-13 serum or nor-BNI on the recovery of spinal cord injury in rats].
A pronounced and reversible spinal cord compression injury was performed by the compression of a metal plate 2.2mm x 5. Omm in size to the exposed spinal dura and loaded with 35g weight for 5 minutes at T7-8. Anti dynorphin serum at 10 microliters (1:30000) or kappa antagonist nor-BNI at 100 ng was administered intrasubarachnoidly shortly after the injury, with half dose applied 1, 2, 3 h after the injury for another three times. The recovery of neurological function was investigated. The results showed that the recovery of the muscle tension and motor function of the hindlimb in anti dynorphin serum group is markedly faster than that in both control and nor-BNI groups. Also the recovery of motor function in nor-BNI group is favourable at the earlier stage of injury, comparing to the control group. There were no significant differences in the change of mean arterial pressure and blood physiological parameters among the three groups. Topics: Animals; Dynorphins; Immune Sera; In Vitro Techniques; Injections, Spinal; Male; Naltrexone; Narcotic Antagonists; Peptide Fragments; Rats; Receptors, Opioid, kappa; Spinal Cord Injuries | 1995 |
[Effect of antiserum against dynorphin A administered intrathecally on spinal cord injury (SCI) of rats and its significance].
The antagonistic effects of antisera against dynorphin A, beta-endorphin, and leu-enkephailin administered intrathecally on secondary SCI were observed and compared after moderate SCI using principle of antigen-antibody neutralization reaction. The protective effect of antiserum against dynorphin A was most prominent on secondary SCI and the effect was more prominent when administered at 24 hours following SCI than when administered at 0 hour four hours, one week or two weeks following SCI. That suggests increase of dynorphin A level in spinal cord tissue may play an active role in the early stage, but its harmful effect on secondary SCI will be more and more prominent after accumulation of excessive dynorphin A. Topics: Animals; Dynorphins; Endorphins; Enkephalin, Leucine; Immune Sera; Immunization, Passive; Injections, Spinal; Male; Motor Activity; Neutralization Tests; Peptide Fragments; Rats; Rats, Sprague-Dawley; Spinal Cord Injuries | 1995 |
Dynorphin A-induced rat spinal cord injury: evidence for excitatory amino acid involvement in a pharmacological model of ischemic spinal cord injury.
Dynorphin A reduced lumbosacral blood flow, elevated cerebrospinal fluid lactic acid concentrations and caused flaccid hindlimb paralysis and striking neuropathological changes after its injection into the spinal subarachnoid space in rats. Coadministration of the vasodilator hydralazine substantially eliminated the paralytic, anaerobic metabolic and neuropathological responses to dynorphin A. In contrast, in concentrations up to 1 mM, dynorphin A did not alter the viability of cultured rat spinal cord neurons. Thus, it appears that this peptide lacks direct neurotoxic effects and that neuronal injuries in vivo result primarily from ischemia associated with dynorphin A-induced blood flow reductions. NMDA receptor antagonists significantly improved recovery from dynorphin A-induced hindlimb paralysis, and substantially eliminated neuropathological changes without attenuating the acute blood flow reductions or lactic acid elevations. Additionally, glutamate and aspartate concentrations were increased significantly in spinal cord cerebrospinal fluid samples removed during the time that peptide-induced spinal cord blood flow reductions were observed. In contrast, neither amino acid concentration was elevated in media removed after 1-hr exposure of spinal cord neuronal cell cultures to 100 microM concentrations of dynorphin A. These results indicate that the paralysis and spinal cord injuries produced in rats after spinal subarachnoid injection of dynorphin A result predominantly from spinal cord ischemia, and further identify excitatory amino acids and N-methyl-D-aspartate receptor mechanisms as important mediators in this injury model. Topics: Amino Acids; Animals; Cells, Cultured; Disease Models, Animal; Dynorphins; Hindlimb; Ischemia; Male; Neurons; Rats; Rats, Sprague-Dawley; Receptors, N-Methyl-D-Aspartate; Spinal Cord; Spinal Cord Injuries | 1994 |
Dynorphin A content in the rat brain and spinal cord after a localized trauma to the spinal cord and its modification with p-chlorophenylalanine. An experimental study using radioimmunoassay technique.
The distribution of dynorphin A in the spinal cord and brain of normal rats and of rats subjected to a focal injury of the spinal cord was examined in a rat model using a radioimmunoassay (RIA) technique. The validity of RIA was checked by high performance liquid chromatography (HPLC). Furthermore, the possibility that the peptide is somehow functionally related with endogenous 5-hydroxytryptamine (5-HT, serotonin), was also evaluated using a pharmacological approach. In normal animals, the peptide content was very similar in the spinal cord segments (T9, T10-11, and T12) examined whereas, the dynorphin content of the whole brain was about two-fold higher compared with that in the spinal cord. A focal injury to the spinal cord in the right dorsal horn (about 1.5 mm deep, 2.5 mm long and 1.5 mm to the right of the midline) of the lower thoracic cord (T10-11) in urethane anaesthetised animals significantly altered the peptide content in the whole brain as well as in the spinal cord. Thus, a decrease in the peptide level in whole brain, T10-11 and in the T12 segments of the spinal cord was observed 1 and 2 h after trauma. At 5 h, the peptide had accumulated markedly in the T9 segment (about a two-fold increase) as compared with the controls. At this time, the peptide content had been restored in the T10-11 and T12 segments. On the other hand, the whole brain dynorphin level continued to remain low (by 55%) as compared to the control group.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Brain Chemistry; Chromatography, High Pressure Liquid; Dynorphins; Fenclonine; Male; Radioimmunoassay; Rats; Rats, Sprague-Dawley; Serotonin; Spinal Cord; Spinal Cord Injuries | 1992 |
Opioid and nonopioid mechanisms may contribute to dynorphin's pathophysiological actions in spinal cord injury.
It has been suggested that the opioid dynorphin, an endogenous agonist for kappa-opiate receptors, contributes to secondary tissue damage after spinal cord injury. To evaluate this hypothesis further, effects of intrathecally administered dynorphin (Dyn) A-(1-17), dynorphin antiserum, or the kappa-selective opiate antagonist nor-binaltorphimine (nor-BNI) were studied in rats subjected to standardized impact trauma to the thoracic spinal cord. Effects of intrathecal Dyn A-(1-17) were also compared to those of Dyn A-(2-17), which is inactive at opiate receptors, in uninjured and injured animals. Both Dyn A-(1-17) and Dyn A-(2-17) produced motor dysfunction in uninjured rats, but Dyn A-(1-17) was approximately 2.5 times more potent. At lower doses of Dyn A-(1-17), paraparesis was markedly attenuated by nor-BNI; nor-BNI was less effective at higher doses of Dyn A-(1-17) and did not modify the motor dysfunction produced by Dyn A-(2-17). Treatment with dynorphin antiserum significantly improved outcome after trauma as compared to control treatment with normal rabbit serum or leucine-enkephalin antiserum. Dyn A-(1-17), but not Dyn A-(2-17) at similar doses, exacerbated neurological dysfunction after spinal cord injury. Pretreatment with nor-BNI attenuated neurological dysfunction after traumatic spinal cord injury to a similar degree in rats administered saline or Dyn A-(1-17). These observations support the hypothesis that dynorphin contributes to certain pathophysiological changes after traumatic spinal cord injury through both opiate-receptor (kappa-receptor)-mediated and nonopioid mechanisms. Topics: Animals; Dynorphins; Injections, Spinal; Male; Naltrexone; Rats; Rats, Inbred Strains; Receptors, Opioid; Spinal Cord Injuries | 1990 |
Dynorphin A-(1-17) induces alterations in free fatty acids, excitatory amino acids, and motor function through an opiate-receptor-mediated mechanism.
The endogenous opioid dynorphin A-(1-17) (Dyn A) has been implicated as a mediator of tissue damage after traumatic spinal cord injury (TSCI) and causes hindlimb paralysis when administered intrathecally. Motor impairment following intrathecal Dyn A is attenuated by antagonists of excitatory amino acids (EAAs); whether opioid receptors mediate such injury has been questioned. TSCI causes various biochemical changes associated with secondary tissue damage, including alterations in tissue amio acids, phospholipids, and fatty acids. Such changes reflect injury severity and correlate with motor dysfunction. The present studies examined whether dynorphin administration causes similar biochemical alterations and whether effects of Dyn A can be modified by treatment with opioid-receptor antagonists. At 24 hr after intrathecal Dyn A, there were significant declines in tissue levels of glutamate, aspartate, and glycine. Increases in total free fatty acids were found at 2 and 24 hr, reflecting changes in both saturated and unsaturated components, which were associated with significant decreases in tissue cholesterol and phospholipid phosphorus at the earlier time point. Each of these neurochemical changes, as well as corresponding motor deficits, were limited by pretreatment with the opioid antagonist nalmefene. In separate experiments, both nalmefene and the selective kappa-opioid antagonist nor-binaltorphimine (nor-BNI) limited dynorphin-induced motor dysfunction; effects of nor-BNI were dose related, and those of nalmefene were stereospecific. Therefore, behavioral and neurochemical consequences of Dyn A administration are mediated in part through opiate receptors, most likely kappa-receptors.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Amino Acids; Animals; Dose-Response Relationship, Drug; Drosophila Proteins; Dynorphins; Fatty Acids, Nonesterified; Injections, Spinal; Male; Membrane Proteins; Motor Activity; Naltrexone; Nerve Tissue Proteins; Peptide Fragments; Rats; Rats, Inbred Strains; Receptors, Opioid; Receptors, Opioid, kappa; Spinal Cord; Spinal Cord Injuries; Stereoisomerism | 1990 |
Prodynorphin gene expression in spinal cord is enhanced after traumatic injury in the rat.
Levels of mRNAs coding for prodynorphin (Pro-Dyn) and proenkephalin (Pro-Enk) as well as the levels of immunoreactive (ir)-dynorphin (Dyn) and (ir)-Met-enkephalin (Met-Enk) were measured in the spinal cord of rats, 65 h following transection or injury of the spinal cord at the T6 spinal segment. Levels of both Pro-Dyn mRNA and of ir-Dyn were significantly increased between 60 and 150%, above control levels in the whole spinal cord, whereas those of Pro-Enk mRNA and of ir-Met-Enk remained unchanged. The increase in spinal levels of Pro-Dyn mRNA were highest in the areas close to the side of transection and indicate an involvement of the Pro-Dyn opioid system in the response to spinal injury and transection. Topics: Animals; Dynorphins; Enkephalin, Methionine; Enkephalins; Gene Expression Regulation; Male; Nucleic Acid Hybridization; Protein Precursors; Rats; Rats, Inbred Strains; RNA; RNA, Complementary; RNA, Messenger; Spinal Cord Injuries | 1988 |
Effects of traumatic injury on dynorphin immunoreactivity in spinal cord.
Traumatic spinal cord injury in rats resulted in a significant eleation of dynorphin A immunoreactivity in spinal cord tissue at the level of, and below, the site of injury. [Leu5]enkephalin levels in the same tissue samples were not significantly altered following severe injury. Dynorphin A immunoreactivity was found in the fraction relatively enriched in synaptosomes after subcellular fractionation of spinal cord tissue. The dynorphin A content of this fraction was not significantly changed following injury, suggesting that dynorphin containing nerve terminals and axons are not severely damaged as a result of the injury. Topics: Animals; Dynorphins; Enkephalin, Leucine; Male; Rats; Rats, Inbred Strains; Spinal Cord; Spinal Cord Injuries; Subcellular Fractions; Synaptosomes | 1985 |
Endogenous opioid immunoreactivity in rat spinal cord following traumatic injury.
It has been postulated that endogenous opioids play a pathophysiological role in spinal cord injury, based on the therapeutic effects of the opiate receptor antagonist naloxone in certain experimental models. The high doses of naloxone required to exert a therapeutic action suggest that naloxone's effects may be mediated by non-mu opiate receptors, such as the kappa receptor. This notion is supported by recent pharmacological studies demonstrating that an opiate antagonist more active at kappa sites is effective and far more potent than naloxone in improving outcome after spinal cord injury. Moreover, dynorphin--postulated to be the endogenous ligand for the kappa receptor--is unique among opioids in producing hindlimb paralysis following intrathecal administration in the rat. In the present studies we have examined changes in endogenous opioid immunoreactivity following traumatic spinal cord injury in the rat. Dynorphin A was found to increase progressively with graded injury; changes were restricted to the injury segment and adjacent areas and were time dependent. Dynorphin A-(1-8) showed no marked changes. Methionine and leucine enkephalin were either unaltered or reduced at the injury site; changes were not well localized and were not clearly related to the injury variables. These findings provide further support for a potential pathophysiological role of prodynorphin-derived peptides in spinal cord injury. Topics: Animals; Dynorphins; Endorphins; Enkephalin, Methionine; Leucine; Peptide Fragments; Radioimmunoassay; Rats; Rats, Inbred Strains; Spinal Cord; Spinal Cord Injuries; Time Factors | 1985 |
Increased dynorphin immunoreactivity in spinal cord after traumatic injury.
Opiate antagonists, at high doses, have been shown to improve physiological variables and outcome after experimental spinal injury. Dynorphin appears to be unique amongst opioids in producing hindlimb paralysis after intrathecal injection. Taken together, these findings suggest a possible pathophysiological role for endogenous opioids, particularly dynorphin, in spinal injury. In the present studies we examined the relationship between changes in dynorphin immunoreactivity (Dyn-ir) in rat spinal cord after traumatic injury and the subsequent motor dysfunction. Trauma was associated with significantly increased Dyn-ir at the injury site, but not distant from the lesion. Dyn-ir was found elevated as early as 2 h and as late as 2 weeks after trauma, and was significantly correlated with the degree of injury. These data are consistent with the hypothesis that dynorphin systems may be involved in the secondary injury that follows spinal trauma. Topics: Animals; Dynorphins; Male; Naloxone; Narcotic Antagonists; Paraplegia; Rats; Rats, Inbred Strains; Receptors, Opioid; Receptors, Opioid, kappa; Spinal Cord; Spinal Cord Injuries | 1985 |