Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 43(3): 419-432, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36639888

ABSTRACT

We tested the hypothesis that dorsal cervical epidural electrical stimulation (CEES) increases respiratory activity in male and female anesthetized rats. Respiratory frequency and minute ventilation were significantly increased when CEES was applied dorsally to the C2-C6 region of the cervical spinal cord. By injecting pseudorabies virus into the diaphragm and using c-Fos activity to identify neurons activated during CEES, we found neurons in the dorsal horn of the cervical spinal cord in which c-Fos and pseudorabies were co-localized, and these neurons expressed somatostatin (SST). Using dual viral infection to express the inhibitory Designer Receptors Exclusively Activated by Designer Drugs (DREADD), hM4D(Gi), selectively in SST-positive cells, we inhibited SST-expressing neurons by administering Clozapine N-oxide (CNO). During CNO-mediated inhibition of SST-expressing cervical spinal neurons, the respiratory excitation elicited by CEES was diminished. Thus, dorsal cervical epidural stimulation activated SST-expressing neurons in the cervical spinal cord, likely interneurons, that communicated with the respiratory pattern generating network to effect changes in ventilation.SIGNIFICANCE STATEMENT A network of pontomedullary neurons within the brainstem generates respiratory behaviors that are susceptible to modulation by a variety of inputs; spinal sensory and motor circuits modulate and adapt this output to meet the demands placed on the respiratory system. We explored dorsal cervical epidural electrical stimulation (CEES) excitation of spinal circuits to increase ventilation in rats. We identified dorsal somatostatin (SST)-expressing neurons in the cervical spinal cord that were activated (c-Fos-positive) by CEES. CEES no longer stimulated ventilation during inhibition of SST-expressing spinal neuronal activity, thereby demonstrating that spinal SST neurons participate in the activation of respiratory circuits affected by CEES. This work establishes a mechanistic foundation to repurpose a clinically accessible neuromodulatory therapy to activate respiratory circuits and stimulate ventilation.


Subject(s)
Cervical Cord , Neurons , Respiratory Rate , Animals , Female , Male , Rats , Cervical Cord/physiology , Electric Stimulation/methods , Neurons/physiology , Proto-Oncogene Proteins c-fos , Somatostatin/metabolism , Somatostatin/pharmacology , Spinal Cord/physiology , Respiratory Rate/physiology
2.
J Physiol ; 600(12): 2973-2999, 2022 06.
Article in English | MEDLINE | ID: mdl-35639046

ABSTRACT

Opioid overdose suppresses brainstem respiratory circuits, causes apnoea and may result in death. Epidural electrical stimulation (EES) at the cervical spinal cord facilitated motor activity in rodents and humans, and we hypothesized that EES of the cervical spinal cord could antagonize opioid-induced respiratory depression in humans. Eighteen patients requiring surgical access to the dorsal surface of the spinal cord between C2 and C7 received EES or sham stimulation for up to 90 s at 5 or 30 Hz during complete (OFF-State) or partial suppression (ON-State) of respiration induced by remifentanil. During the ON-State, 30 Hz EES at C4 and 5 Hz EES at C3/4 increased tidal volume and decreased the end-tidal carbon dioxide level compared to pre-stimulation control levels. EES of 5 Hz at C5 and C7 increased respiratory frequency compared to pre-stimulation control levels. In the OFF-State, 30 Hz cervical EES at C3/4 terminated apnoea and induced rhythmic breathing. In cadaveric tissue obtained from a brain bank, more neurons expressed both the neurokinin 1 receptor (NK1R) and somatostatin (SST) in the cervical spinal levels responsive to EES (C3/4, C6 and C7) compared to a region non-responsive to EES (C2). Thus, the capacity of cervical EES to oppose opioid depression of respiration may be mediated by NK1R+/SST+ neurons in the dorsal cervical spinal cord. This study provides proof of principle that cervical EES may provide a novel therapeutic approach to augment respiratory activity when the neural function of the central respiratory circuits is compromised by opioids or other pathological conditions. KEY POINTS: Epidural electrical stimulation (EES) using an implanted spinal cord stimulator (SCS) is an FDA-approved method to manage chronic pain. We tested the hypothesis that cervical EES facilitates respiration during administration of opioids in 18 human subjects who were treated with low-dose remifentanil that suppressed respiration (ON-State) or high-dose remifentanil that completely inhibited breathing (OFF-State) during the course of cervical surgery. Dorsal cervical EES of the spinal cord augmented the respiratory tidal volume or increased the respiratory frequency, and the response to EES varied as a function of the stimulation frequency (5 or 30 Hz) and the cervical level stimulated (C2-C7). Short, continuous cervical EES restored a cyclic breathing pattern (eupnoea) in the OFF-State, suggesting that cervical EES reversed the opioid-induced respiratory depression. These findings add to our understanding of respiratory pattern modulation and suggest a novel mechanism to oppose the respiratory depression caused by opioids.


Subject(s)
Cervical Cord , Respiratory Insufficiency , Spinal Cord Injuries , Analgesics, Opioid/adverse effects , Apnea , Electric Stimulation/methods , Humans , Remifentanil , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/therapy , Spinal Cord/physiology
3.
Anesth Analg ; 128(1): 161-167, 2019 01.
Article in English | MEDLINE | ID: mdl-29596097

ABSTRACT

BACKGROUND: Opioids are effective postoperative analgesics. Disturbingly, we have previously reported that opioids such as morphine can worsen inflammatory pain and peripheral and central neuropathic pain. These deleterious effects are mediated by immune mediators that promote neuronal hyperexcitability in the spinal dorsal horn. Herein, we tested whether perioperative morphine could similarly prolong postoperative pain in male rats. METHODS: Rats were treated with morphine for 7 days, beginning immediately after laparotomy, while the morphine was tapered in a second group. Expression of genes for inflammatory mediators was quantified in the spinal dorsal horn. In the final experiment, morphine was administered before laparotomy for 7 days. RESULTS: We found that morphine treatment after laparotomy extended postoperative pain by more than 3 weeks (time × treatment: P < .001; time: P < .001; treatment: P < .05). Extension of postoperative pain was not related to morphine withdrawal, as it was not prevented by dose tapering (time × treatment: P = .8; time: P < .001; treatment: P = .9). Prolonged postsurgical pain was associated with increased expression of inflammatory genes, including those encoding Toll-like receptor 4, NOD like receptor protein 3 (NLRP3), nuclear factor kappa B (NFκB), caspase-1, interleukin-1ß, and tumor necrosis factor (P < .05). Finally, we showed that of preoperative morphine, concluding immediately before laparotomy, similarly prolonged postoperative pain (time × treatment: P < .001; time: P < .001; treatment: P < .001). There is a critical window for morphine potentiation of pain, as a 7-day course of morphine that concluded 1 week before laparotomy did not prolong postsurgical pain. CONCLUSIONS: These studies indicate the morphine can have a deleterious effect on postoperative pain. These studies further suggest that longitudinal studies could be performed to test whether opioids similarly prolong postoperative pain in the clinic.


Subject(s)
Analgesics, Opioid/toxicity , Hyperalgesia/chemically induced , Morphine/toxicity , Pain Threshold/drug effects , Pain, Postoperative/chemically induced , Posterior Horn Cells/drug effects , Analgesics, Opioid/administration & dosage , Animals , Disease Models, Animal , Drug Administration Schedule , Hyperalgesia/diagnostic imaging , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Inflammation Mediators/metabolism , Laparotomy , Male , Morphine/administration & dosage , Pain Measurement , Pain, Postoperative/drug therapy , Pain, Postoperative/metabolism , Pain, Postoperative/physiopathology , Posterior Horn Cells/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors
4.
Brain Res ; 1692: 9-11, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29723521

ABSTRACT

We have recently reported that a short course of morphine, starting 10 days after sciatic chronic constriction injury (CCI), prolonged the duration of mechanical allodynia for months after morphine ceased. Maintenance of this morphine-induced persistent sensitization was dependent on microglial reactivity and Toll-like receptor 4 signaling. Given that microRNAs (miRNAs) such as miR-124 and miR-146a possess the ability to modulate such signaling, we directly compared their function in this model. We found that both miRNAs reversed established allodynia in our model of morphine-induced persistent sensitization. The efficacy of miR-124 and miR-146a were comparable, and in both cases allodynia returned within hours to days of miRNA dosing conclusion. Our findings demonstrate that miRNAs targeting Toll-like receptor signaling are effective in reversing neuropathic pain, which underscores the clinical potential of these non-coding RNAs.


Subject(s)
Analgesics, Opioid/adverse effects , Anti-Inflammatory Agents/therapeutic use , MicroRNAs/therapeutic use , Morphine/adverse effects , Neuralgia/physiopathology , Analgesics, Opioid/therapeutic use , Animals , Disease Models, Animal , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Morphine/therapeutic use , Neuralgia/drug therapy , Pain Threshold/drug effects , Rats , Rats, Inbred F344
5.
Exp Neurol ; 304: 125-131, 2018 06.
Article in English | MEDLINE | ID: mdl-29530713

ABSTRACT

The absence of selective pharmacological tools is a major barrier to the in vivo study of microglia. To address this issue, we developed a Gq- and Gi-coupled Designer Receptor Exclusively Activated by a Designer Drug (DREADD) to enable selective stimulation or inhibition of microglia, respectively. DREADDs under a CD68 (microglia/macrophage) promoter were intrathecally transfected via an AAV9 vector. Naïve male rats intrathecally transfected with Gq (stimulatory) DREADDs exhibited significant allodynia following intrathecal administration of the DREADD-selective ligand clozapine-N-oxide (CNO), which was abolished by intrathecal interleukin-1 receptor antagonist. Chronic constriction injury-induced allodynia was attenuated by intrathecal CNO in male rats intrathecally transfected with Gi (inhibitory) DREADDs. To explore mechanisms, BV2 cells were stably transfected with Gq or Gi DREADDs in vitro. CNO treatment induced pro-inflammatory mediator production per se from cells expressing Gq-DREADDs, and inhibited lipopolysaccharide- and CCL2-induced inflammatory signaling from cells expressing Gi-DREADDs. These studies are the first to manipulate microglia function using DREADDs, which allow the role of glia in pain to be conclusively demonstrated, unconfounded by neuronal off-target effects that exist for all other drugs that also inhibit glia. Hence, these studies are the first to conclusively demonstrate that in vivo stimulation of resident spinal microglia in intact spinal cord is a) sufficient for allodynia, and b) necessary for allodynia induced by peripheral nerve injury. DREADDs are a unique tool to selectively explore the physiological and pathological role of microglia in vivo.


Subject(s)
Microglia/metabolism , Neuralgia/physiopathology , Receptors, G-Protein-Coupled/metabolism , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Designer Drugs/pharmacology , Disease Models, Animal , Inflammation/metabolism , Inflammation/physiopathology , Male , Microglia/drug effects , Neuralgia/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/physiopathology , Transfection
6.
Brain Behav Immun ; 72: 45-50, 2018 08.
Article in English | MEDLINE | ID: mdl-28860068

ABSTRACT

We have recently reported that a short course of morphine, starting 10days after sciatic chronic constriction injury (CCI), prolonged the duration of mechanical allodynia for months after morphine ceased. Maintenance of this morphine-induced persistent sensitization was dependent on spinal NOD-like receptor protein 3 (NLRP3) inflammasomes-protein complexes that proteolytically activate interleukin-1ß (IL-1ß) via caspase-1. However, it is still unclear how NLRP3 inflammasome signaling is maintained long after morphine is cleared. Here, we demonstrate that spinal levels of the damage associated molecular patterns (DAMPs) high mobility group box 1 (HMGB1) and biglycan are elevated during morphine-induced persistent sensitization in male rats; that is, 5weeks after cessation of morphine dosing. We also show that HMGB1 and biglycan levels are at least partly dependent on the initial activation of caspase-1, as well as Toll like receptor 4 (TLR4) and the purinergic receptor P2X7R-receptors responsible for priming and activation of NLRP3 inflammasomes. Finally, pharmacological attenuation of the DAMPs HMGB1, biglycan, heat shock protein 90 and fibronectin persistently reversed morphine-prolonged allodynia. We conclude that after peripheral nerve injury, morphine treatment results in persistent DAMP release via TLR4, P2X7R and caspase-1, which are involved in formation/activation of NLRP3 inflammasomes. These DAMPs are responsible for maintaining persistent allodynia, which may be due to engagement of a positive feedback loop, in which NLRP3 inflammasomes are persistently activated by DAMPs signaling at TLR4 and P2X7R.


Subject(s)
Alarmins/physiology , Peripheral Nerve Injuries/drug therapy , Spinal Injuries/immunology , Alarmins/drug effects , Animals , Caspase 1/metabolism , HMGB1 Protein/metabolism , Hyperalgesia/metabolism , Inflammasomes/metabolism , Injections, Spinal , Interleukin-1beta/metabolism , Male , Morphine/metabolism , Morphine/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuralgia/metabolism , Rats , Rats, Inbred F344 , Receptors, Purinergic P2X7/metabolism , Spinal Injuries/drug therapy , Toll-Like Receptor 4/metabolism
7.
Pain ; 157(9): 2012-23, 2016 09.
Article in English | MEDLINE | ID: mdl-27355182

ABSTRACT

Exercise is known to exert a systemic anti-inflammatory influence, but whether its effects are sufficient to protect against subsequent neuropathic pain is underinvestigated. We report that 6 weeks of voluntary wheel running terminating before chronic constriction injury (CCI) prevented the full development of allodynia for the ∼3-month duration of the injury. Neuroimmune signaling was assessed at 3 and 14 days after CCI. Prior exercise normalized ipsilateral dorsal spinal cord expression of neuroexcitatory interleukin (IL)-1ß production and the attendant glutamate transporter GLT-1 decrease, as well as expression of the disinhibitory P2X4R-BDNF axis. The expression of the macrophage marker Iba1 and the chemokine CCL2 (MCP-1), and a neuronal injury marker (activating transcription factor 3), was attenuated by prior running in the ipsilateral lumbar dorsal root ganglia. Prior exercise suppressed macrophage infiltration and/or injury site proliferation, given decreased presence of macrophage markers Iba1, iNOS (M1), and Arg-1 (M2; expression was time dependent). Chronic constriction injury-driven increases in serum proinflammatory chemokines were suppressed by prior running, whereas IL-10 was increased. Peripheral blood mononuclear cells were also stimulated with lipopolysaccharide ex vivo, wherein CCI-induced increases in IL-1ß, nitrite, and IL-10 were suppressed by prior exercise. Last, unrestricted voluntary wheel running, beginning either the day of, or 2 weeks after, CCI, progressively reversed neuropathic pain. This study is the first to investigate the behavioral and neuroimmune consequences of regular exercise terminating before nerve injury. This study suggests that chronic pain should be considered a component of "the diseasome of physical inactivity," and that an active lifestyle may prevent neuropathic pain.


Subject(s)
Exercise Movement Techniques/methods , Neuralgia/prevention & control , Activating Transcription Factor 3/metabolism , Animals , Calcium-Binding Proteins/metabolism , Constriction, Pathologic/complications , Cytokines/metabolism , Disease Models, Animal , Excitatory Amino Acid Transporter 2/metabolism , Functional Laterality , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Hyperalgesia/rehabilitation , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male , Microfilament Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuralgia/etiology , Neuralgia/pathology , Nitrites/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X5/metabolism , Sciatic Neuropathy/prevention & control , p21-Activated Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Proc Natl Acad Sci U S A ; 113(24): E3441-50, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27247388

ABSTRACT

Opioid use for pain management has dramatically increased, with little assessment of potential pathophysiological consequences for the primary pain condition. Here, a short course of morphine, starting 10 d after injury in male rats, paradoxically and remarkably doubled the duration of chronic constriction injury (CCI)-allodynia, months after morphine ceased. No such effect of opioids on neuropathic pain has previously been reported. Using pharmacologic and genetic approaches, we discovered that the initiation and maintenance of this multimonth prolongation of neuropathic pain was mediated by a previously unidentified mechanism for spinal cord and pain-namely, morphine-induced spinal NOD-like receptor protein 3 (NLRP3) inflammasomes and associated release of interleukin-1ß (IL-1ß). As spinal dorsal horn microglia expressed this signaling platform, these cells were selectively inhibited in vivo after transfection with a novel Designer Receptor Exclusively Activated by Designer Drugs (DREADD). Multiday treatment with the DREADD-specific ligand clozapine-N-oxide prevented and enduringly reversed morphine-induced persistent sensitization for weeks to months after cessation of clozapine-N-oxide. These data demonstrate both the critical importance of microglia and that maintenance of chronic pain created by early exposure to opioids can be disrupted, resetting pain to normal. These data also provide strong support for the recent "two-hit hypothesis" of microglial priming, leading to exaggerated reactivity after the second challenge, documented here in the context of nerve injury followed by morphine. This study predicts that prolonged pain is an unrealized and clinically concerning consequence of the abundant use of opioids in chronic pain.


Subject(s)
Chronic Pain/metabolism , Inflammasomes/metabolism , Microglia/metabolism , Morphine/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuralgia/metabolism , Spinal Cord Dorsal Horn/metabolism , Animals , Chronic Pain/pathology , Chronic Pain/physiopathology , Clozapine/analogs & derivatives , Clozapine/pharmacology , Interleukin-1beta/metabolism , Male , Microglia/pathology , Neuralgia/pathology , Neuralgia/physiopathology , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/pathology , Spinal Cord Dorsal Horn/physiopathology
9.
Ann Transl Med ; 3(Suppl 1): S23, 2015 May.
Article in English | MEDLINE | ID: mdl-26046069
SELECTION OF CITATIONS
SEARCH DETAIL
...