Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Ann Neurol ; 85(3): 406-420, 2019 03.
Article in English | MEDLINE | ID: mdl-30635946

ABSTRACT

OBJECTIVE: The two related tumor necrosis factor members a proliferation-inducing ligand (APRIL) and B-cell activation factor (BAFF) are currently targeted in autoimmune diseases as B-cell regulators. In multiple sclerosis (MS), combined APRIL/BAFF blockade led to unexpected exacerbated inflammation in the central nervous system (CNS) of patients. Here, we investigate the role of the APRIL/BAFF axis in the CNS. METHODS: APRIL expression was analyzed in MS lesions by immunohistochemistry. The in vivo role of APRIL was assessed in the murine MS model, experimental autoimmune encephalitis (EAE). Functional in vitro studies were performed with human and mouse astrocytes. RESULTS: APRIL was expressed in lesions from EAE. In its absence, the disease was worst. Lesions from MS patients also showed APRIL expression upon infiltration of macrophages. Notably, all the APRIL secreted by these macrophages specifically targeted astrocytes. The upregulation of chondroitin sulfate proteoglycan, sometimes bearing chondroitin sulfate of type E sugar moieties, binding APRIL, in reactive astrocytes explained the latter selectivity. Astrocytes responded to APRIL by producing a sufficient amount of IL-10 to dampen antigen-specific T-cell proliferation and pathogenic cytokine secretion. Finally, an intraspinal delivery of recombinant APRIL before disease onset, shortly reduced EAE symptoms. Repeated intravenous injections of recombinant APRIL before and even at disease onset also had an effect. INTERPRETATION: Our data show that APRIL mediates an anti-inflammatory response from astrocytes in MS lesions. This protective activity is not shared with BAFF. ANN NEUROL 2019;85:406-420.


Subject(s)
Astrocytes/metabolism , B-Cell Activating Factor/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Multiple Sclerosis/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Adult , Aged , Animals , Astrocytes/immunology , Astrocytes/pathology , Cell Proliferation , Chondroitin Sulfate Proteoglycans/metabolism , Chondroitin Sulfates/metabolism , Cytokines/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Immunohistochemistry , Interleukin-10/immunology , Macrophages/pathology , Male , Mice , Mice, Knockout , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
2.
Nat Rev Neurosci ; 16(2): 69-78, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25588373

ABSTRACT

Chemokines and opioids are important regulators of immune, inflammatory and neuronal responses in peripheral and central pain pathways. Recent studies have provided insights into the functional interactions between chemokine receptors and opioid receptors, and their role in pain modulation. In this Progress article, we discuss how crosstalk between these two systems might provide a molecular and cellular framework for the development of novel analgesic therapies for the management of acute and/or chronic pain.


Subject(s)
Pain Management , Pain/metabolism , Receptor Cross-Talk/physiology , Receptors, Chemokine/metabolism , Receptors, Opioid/metabolism , Humans
3.
Anesthesiology ; 129(3): 590-607, 2018 09.
Article in English | MEDLINE | ID: mdl-29738328

ABSTRACT

The development of chronic pain is considered a major complication after surgery. Basic science research in animal models helps us understand the transition from acute to chronic pain by identifying the numerous molecular and cellular changes that occur in the peripheral and central nervous systems. It is now well recognized that inflammation and nerve injury lead to long-term synaptic plasticity that amplifies and also maintains pain signaling, a phenomenon referred to as pain sensitization. In the context of surgery in humans, pain sensitization is both responsible for an increase in postoperative pain via the expression of wound hyperalgesia and considered a critical factor for the development of persistent postsurgical pain. Using specific drugs that block the processes of pain sensitization reduces postoperative pain and prevents the development of persistent postoperative pain. This narrative review of the literature describes clinical investigations evaluating different preventative pharmacologic strategies that are routinely used by anesthesiologists in their daily clinical practices for preventing persistent postoperative pain. Nevertheless, further efforts are needed in both basic and clinical science research to identify preclinical models and novel therapeutics targets. There remains a need for more patient numbers in clinical research, for more reliable data, and for the development of the safest and the most effective strategies to limit the incidence of persistent postoperative pain.


Subject(s)
Analgesics/therapeutic use , Chronic Pain/drug therapy , Chronic Pain/physiopathology , Pain, Postoperative/drug therapy , Pain, Postoperative/physiopathology , Adrenergic alpha-2 Receptor Agonists/therapeutic use , Analgesics, Opioid/therapeutic use , Anesthetics, Local/therapeutic use , Animals , Chronic Pain/prevention & control , Humans , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Pain, Postoperative/prevention & control
4.
J Cardiothorac Vasc Anesth ; 30(4): 997-1004, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27521969

ABSTRACT

OBJECTIVES: The authors hypothesized that intravenous acetaminophen as an adjunct analgesic would significantly decrease 24-hour postoperative opioid consumption. DESIGN: Double-blind, randomized, placebo-controlled trial. SETTING: A single academic medical center. PARTICIPANTS: The study was comprised of 68 adult patients undergoing cardiac surgery. INTERVENTIONS: Patients were assigned randomly to receive either 1,000 mg of intravenous acetaminophen or placebo immediately after anesthesia induction, at the end of surgery, and then every 6 hours for the first 24 hours in the intensive care unit, for a total of 6-1,000 mg doses. MEASUREMENTS AND MAIN RESULTS: The primary outcome was 24-hour postoperative opioid consumption. The secondary outcomes included 48-hour postoperative opioid consumption, incisional pain scores, opioid-related adverse effects, length of mechanical ventilation, length of intensive care unit stay, and the extent of wound hyperalgesia assessed at 24 and 48 hours postoperatively. The mean±standard deviation postoperative 24-hour opioid consumption expressed in morphine equivalents was significantly less in the acetaminophen group (45.6±29.5 mg) than in the placebo group (62.3±29.5 mg), representing a 27% reduction in opioid consumption (95% CI, 2.3-31.1 mg; p = 0.024). There were no differences in pain scores and opioid-related adverse effects between the 2 groups. A significantly greater number of patients in the acetaminophen group responded "very much" and "extremely well" when asked how their overall pain experience met their expectation (p = 0.038). CONCLUSIONS: The administration of intravenous acetaminophen during cardiac surgery and for the first 24 hours postoperatively reduced opioid consumption and improved patient satisfaction with their overall pain experience but did not reduce opioid side effects.


Subject(s)
Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Cardiac Surgical Procedures , Pain, Postoperative/drug therapy , Acetaminophen/administration & dosage , Administration, Intravenous , Adolescent , Adult , Aged , Analgesia/methods , Analgesics, Non-Narcotic/administration & dosage , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Hyperalgesia , Length of Stay/statistics & numerical data , Male , Middle Aged , Prospective Studies , Respiration, Artificial/statistics & numerical data , Young Adult
5.
J Clin Monit Comput ; 29(2): 291-5, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25062948

ABSTRACT

The PhysioDoloris™ analgesia monitor assesses nociception effects on the autonomic nervous system by analyzing changes in heart rate variability (HRV). This non-invasive device analyses ECG signals and determines the analgesia nociception index (ANI), allowing for quantitative assessment of the analgesia/nociception balance in anesthetized patients. Ketamine, an analgesic adjuvant with sympathomimetic properties, has been shown to improve perioperative pain management. The purpose of this pilot study was to evaluate whether low-dose ketamine, due to its intrinsic effect on the sino-atrial node, affects HRV and, therefore, interferes with ANI measurements. This pilot study included 20 women undergoing abdominal hysterectomies. Anesthesia and analgesia were maintained with sevoflurane and fentanyl respectively, in a standardized manner. Five minutes after intubation, 0.5 µg kg(-1) of intravenous (i.v.) ketamine was administered. ANI, bispectral index (BIS), heart rate and blood pressure were recorded from the induction of anesthesia until 5 min after skin incision. There was not any significant decrease in mean (±SD) ANI values after intubation (2.11±20.11, p=0.35) or i.v. ketamine administration (1.31±15.26, p=0.28). The mean (±SD) reduction in ANI values after skin incision was statistically significant (13.65±15.44, p=0.01), which is consistent with increased nociception. A single i.v. bolus of 0.5 µg kg(-1) ketamine did not influence the ANI values of 20 women under standardized general anesthesia conditions and absent noxious stimulation. These results suggest that the ANI derived from the PhysioDoloris™ analgesia monitor is feasible under such clinical conditions.


Subject(s)
Electrocardiography/instrumentation , Ketamine/administration & dosage , Monitoring, Intraoperative/instrumentation , Nociceptive Pain/diagnosis , Pain Measurement/drug effects , Pain Measurement/instrumentation , Algorithms , Dose-Response Relationship, Drug , Electrocardiography/drug effects , Equipment Design , Equipment Failure Analysis , Female , Humans , Middle Aged , Nociceptive Pain/prevention & control , Pilot Projects , Reproducibility of Results , Sensitivity and Specificity
6.
Brain Behav Immun ; 38: 38-52, 2014 May.
Article in English | MEDLINE | ID: mdl-24263070

ABSTRACT

Functional interactions between the chemokine receptor CXCR4 and opioid receptors have been reported in the brain, leading to a decreased morphine analgesic activity. However the cellular mechanisms responsible for this loss of opioid analgesia are largely unknown. Here we examined whether Src family-kinases (SFK)-linked mechanisms induced by CXCR4 contributed to the loss of acute morphine analgesia and could represent a new physiological anti-opioid signaling pathway. In this way, we showed by immunohistochemistry and western blot that CXCL12 rapidly activated SFK phosphorylation in vitro in primary cultured lumbar rat dorsal root ganglia (DRG) but also in vivo in the DRG and the spinal cord. We showed that SFK activation occurred in a sub population of sensory neurons, in spinal microglia but also in spinal nerve terminals expressing mu-(MOR) and delta-opioid (DOR) receptor. In addition we described that CXCR4 is detected in MOR- and DOR-immunoreactive neurons in the DRG and spinal cord. In vivo, we demonstrated that an intrathecal administration of CXCL12 (1µg) significantly attenuated the subcutaneous morphine (4mg/kg) analgesia. Conversely, pretreatment with a potent CXCR4 antagonist (5µg) significantly enhanced morphine analgesia. Similar effects were obtained after an intrathecal injection of a specific SFK inhibitor, PP2 (10µg). Furthermore, PP2 abrogated CXCL12-induced decrease in morphine analgesia by suppressing SFK activation in the spinal cord. In conclusion, our data highlight that CXCL12-induced loss of acute morphine analgesia is linked to Src family kinases activation.


Subject(s)
Analgesics, Opioid/pharmacology , Chemokine CXCL12/pharmacology , Ganglia, Spinal/enzymology , Morphine/pharmacology , Receptors, CXCR4/metabolism , src-Family Kinases/metabolism , Animals , Drug Tolerance , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Male , Microglia/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley
7.
Article in English | MEDLINE | ID: mdl-37858736

ABSTRACT

The selection and optimization of appropriate adaptive responses depends on interoceptive and exteroceptive stimuli as well as on the animal's ability to switch from one behavioral strategy to another. Although growing evidence indicate that dopamine D2R-mediated signaling events ensure the selection of the appropriate strategy for each specific situation, the underlying neural circuits through which they mediate these effects are poorly characterized. Here, we investigated the role of D2R signaling in a mesolimbic neuronal subpopulation expressing the Wolfram syndrome 1 (Wfs1) gene. This subpopulation is located within the nucleus accumbens, the central amygdala, the bed nucleus of the stria terminalis, and the tail of the striatum, all brain regions critical for the regulation of emotions and motivated behaviors. Using a mouse model carrying a temporally controlled deletion of D2R in WFS1-neurons, we demonstrate that intact D2R signaling in this neuronal population is necessary to regulate homeostasis-dependent food-seeking behaviors in both male and female mice. In addition, we found that reduced D2R signaling in WFS1-neurons impaired active avoidance learning and innate escape responses. Collectively, these findings identify a yet undocumented role for D2R signaling in WFS1-neurons as a novel effector through which dopamine optimizes appetitive behaviors and regulates defensive behaviors.


Subject(s)
Dopamine , Wolfram Syndrome , Animals , Female , Male , Avoidance Learning , Neurons/physiology , Receptors, Dopamine D1 , Receptors, Dopamine D2/genetics
8.
J Cardiothorac Vasc Anesth ; 27(3): 427-35, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23063945

ABSTRACT

OBJECTIVE: The purpose of this randomized, double-blind placebo-controlled study was to evaluate the effect of nefopam, a centrally acting antinociceptive compound, on the development of hyperalgesia after sternotomy. Preventive strategy giving nefopam from the early stage of anesthesia was compared with a postoperative strategy only and placebo. DESIGN: This study was double-blinded and randomized. SETTING: It was conducted in a single university hospital. PARTICIPANTS: Ninety American Society of Anesthesiologists II to III patients scheduled for elective cardiac surgery. INTERVENTIONS: Patients were assigned randomly to receive a 0.3-mg/kg bolus of nefopam at the induction of anesthesia followed by a continuous infusion of 0.065 mg/kg/h for 48 hours (G1), a 0.3-mg/kg bolus of nefopam at the end of surgery followed by a continuous infusion of 0.065 mg/kg/h for 48 hours (G2), or a placebo (G3). Postoperative analgesia was based on morphine patient-controlled analgesia and rescue analgesia when necessary. Postoperative hyperalgesia, pain scores, morphine consumption, and postoperative cognitive dysfunction were assessed for the first 48 hours and thereafter on postoperative days 4 and 7. MEASUREMENTS AND MAIN RESULTS: The postoperative extent of dynamic hyperalgesia and the decrease of the nociceptive threshold evaluated by von Frey filaments at the sternal midline were smaller in group 1 and group 2 compared with the placebo group at the 24th hour. The primary objective was the extent of hyperalgesia at the midline given as the mean (standard deviation [SD]) (4.4 [2.5] cm for G1, 4.1 [2.7] for G2, and 6.1 [2.7] cm for G3. The punctuate is given as mean (SD) (64 [43] g for G1, 68 [40.8] g for G2, and 32 [27] g for G3; with p < 0.05 for the comparisons of extent and punctuate hyperalgesia between G1 and G3 and G2 and G3). The extent of hyperalgesia was not significantly different among the 3 groups on days 2, 4, and 7 after surgery. There were no significant differences in pain scores, morphine consumption, or postoperative cognitive dysfunctions. CONCLUSIONS: Nefopam administered during the perioperative period slightly reduced acute hyperalgesia after cardiac surgery, but this was not associated with improved analgesic efficacy.


Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Cardiac Surgical Procedures/adverse effects , Hyperalgesia/drug therapy , Nefopam/therapeutic use , Pain, Postoperative/drug therapy , Aged , Anesthesia/adverse effects , Coronary Artery Bypass , Double-Blind Method , Female , Follow-Up Studies , Heart Valve Prosthesis Implantation , Humans , Male , Middle Aged , Pain Management/methods , Pain Measurement/methods , Pain Threshold , Perioperative Care , Postoperative Complications/epidemiology , Postoperative Nausea and Vomiting/epidemiology
9.
Curr Opin Anaesthesiol ; 26(5): 621-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23995064

ABSTRACT

PURPOSE OF REVIEW: The aim of the present review is to describe how regional anaesthesia might oppose neuronal changes that surgery and opioids cause in the central nervous system to block both pain sensitization and chronicization following surgery. This might help anaesthesiologists to better understand the impact of their practice on the development of postoperative chronic pain. RECENT FINDINGS: Even though there are more evidences from animals and clinical trials showing that regional anaesthesia might impact the acute pain/hyperalgesia and chronic postsurgical pain, the controversy on how and when to use regional anesthesia to avoid chronic pain persists. Animal studies brought only a very partial answer on what to do in our daily clinical practice. Clinical studies were performed in different types of surgery with various protocols so that a strong conclusion on whether or not using regional anesthesia might benefit the patient to decrease the risk of postoperative chronic pain remains unclear. Studies performed with neuraxial anaesthesia seem to bring better evidences than those with nerve blocks. Future studies will have to specifically evaluate acute hyperalgesia and postoperative chronic pain and not only the classical pain scores and analgesic consumption to bring us the answer we all need. SUMMARY: Regional anaesthesia is able to reduce postoperative acute hyperalgesia and long-term chronic pain by decreasing pain sensitization induced by the surgery itself, and intraoperative use and opioid-induced hyperalgesia. Nevertheless, clinical studies on neuraxial anaesthesia and nerve blocks did not bring so far a strong conclusion to this question, and further better designed studies are necessary.


Subject(s)
Anesthesia, Conduction , Hyperalgesia/prevention & control , Pain, Postoperative/prevention & control , Animals , Chronic Pain/prevention & control , Disease Models, Animal , Humans
10.
JCI Insight ; 8(9)2023 05 08.
Article in English | MEDLINE | ID: mdl-37154155

ABSTRACT

Chronic pain, whether of inflammatory or neuropathic origin, affects about 18% of the population of developed countries, and most current treatments are only moderately effective and/or cause serious side effects. Therefore, the development of novel therapeutic approaches still represents a major challenge. The Na,K-ATPase modulator FXYD2 is critically required for the maintenance of neuropathic pain in rodents. Here, we set up a therapeutic protocol based on the use of chemically modified antisense oligonucleotides (ASOs) to inhibit FXYD2 expression and treat chronic pain. We identified an ASO targeting a 20-nucleotide stretch in the FXYD2 mRNA that is evolutionarily conserved between rats and humans and is a potent inhibitor of FXYD2 expression. We used this sequence to synthesize lipid-modified forms of ASO (FXYD2-LASO) to facilitate their entry into dorsal root ganglia neurons. We established that intrathecal or intravenous injections of FXYD2-LASO in rat models of neuropathic or inflammatory pain led to a virtually complete alleviation of their pain symptoms, without causing obvious side effects. Remarkably, by using 2'-O-2-methoxyethyl chemical stabilization of the ASO (FXYD2-LASO-Gapmer), we could significantly prolong the therapeutic action of a single treatment up to 10 days. This study establishes FXYD2-LASO-Gapmer administration as a promising and efficient therapeutic strategy for long-lasting relief of chronic pain conditions in human patients.


Subject(s)
Chronic Pain , Neuralgia , Rats , Humans , Animals , Oligonucleotides, Antisense/pharmacology , Chronic Pain/drug therapy , Chronic Pain/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Oligonucleotides , Neuralgia/drug therapy , Chronic Disease
11.
Prog Neurobiol ; 222: 102405, 2023 03.
Article in English | MEDLINE | ID: mdl-36646299

ABSTRACT

Acute pain has been associated with persistent pain sensitization of nociceptive pathways increasing the risk of transition from acute to chronic pain. We demonstrated the critical role of the FLT3- tyrosine kinase receptor, expressed in sensory neurons, in pain chronification after peripheral nerve injury. However, it is unclear whether injury-induced pain sensitization can also promote long-term mood disorders. Here, we evaluated the emotional and sensorial components of pain after a single (SI) or double paw incision (DI) and the implication of FLT3. DI mice showed an anxiodepressive-like phenotype associated with extended mechanical pain hypersensitivity and spontaneous pain when compared to SI mice. Behavioral exaggeration was associated with peripheral and spinal changes including increased microglia activation after DI versus SI. Intrathecal microglial inhibitors not only eliminated the exaggerated pain hypersensitivity produced by DI but also prevented anxiodepressive-related behaviors. Behavioral and cellular changes produced by DI were blocked in Flt3 knock-out animals and recapitulated by repeated intrathecal FL injections in naive animals. Finally, humanized antibodies against FLT3 reduced DI-induced behavioral and microglia changes. Altogether our results show that the repetition of peripheral lesions facilitate not only exaggerated nociceptive behaviors but also induced anxiodepressive disorders supported by spinal central changes that can be blocked by targeting peripheral FLT3.


Subject(s)
Chronic Pain , Peripheral Nerve Injuries , Animals , Mice , Chronic Pain/metabolism , Emotions , Hyperalgesia/metabolism , Microglia/metabolism , Neurons/metabolism , Peripheral Nerve Injuries/metabolism
12.
Elife ; 122023 05 31.
Article in English | MEDLINE | ID: mdl-37254842

ABSTRACT

Resident macrophages are distributed across all tissues and are highly heterogeneous due to adaptation to different tissue-specific environments. The resident macrophages of the sensory ganglia (sensory neuron-associated macrophages, sNAMs) are in close contact with the cell body of primary sensory neurons and might play physiological and pathophysiological roles. After peripheral nerve injury, there is an increase in the population of macrophages in the sensory ganglia, which have been implicated in different conditions, including neuropathic pain development. However, it is still under debate whether macrophage accumulation in the sensory ganglia after peripheral nerve injury is due to the local proliferation of resident macrophages or a result of blood monocyte infiltration. Here, we confirmed that the number of macrophages increased in the sensory ganglia after the spared nerve injury (SNI) model in mice. Using different approaches, we found that the increase in the number of macrophages in the sensory ganglia after SNI is a consequence of the proliferation of resident CX3CR1+ macrophages, which participate in the development of neuropathic pain, but not due to infiltration of peripheral blood monocytes. These proliferating macrophages are the source of pro-inflammatory cytokines such as TNF and IL-1b. In addition, we found that CX3CR1 signaling is involved in the sNAMs proliferation and neuropathic pain development after peripheral nerve injury. In summary, these results indicated that peripheral nerve injury leads to sNAMs proliferation in the sensory ganglia in a CX3CR1-dependent manner accounting for neuropathic pain development. In conclusion, sNAMs proliferation could be modulated to change pathophysiological conditions such as chronic neuropathic pain.


Subject(s)
Neuralgia , Peripheral Nerve Injuries , Mice , Animals , Peripheral Nerve Injuries/complications , Ganglia, Spinal , Macrophages , Ganglia, Sensory , Sensory Receptor Cells , Cell Proliferation , Hyperalgesia
13.
J Neurosci ; 31(36): 12889-99, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21900567

ABSTRACT

A stressful event increases the risk of developing depression later in life, but the possible predisposing factors remain unknown. Our study aims to characterize latent vulnerability traits underlying the development of depressive disorders in adult animals. Four weeks after a priming stressful event, serum corticosterone concentration returned to control values in all animals, whereas the other biological parameters returned to basal level in only 58% of animals (called nonvulnerable). In contrast, 42% of animals displayed persistent decreased serum and hippocampus BDNF concentrations, reduced hippocampal volume and neurogenesis, CA3 dendritic retraction and decrease in spine density, as well as amygdala neuron hypertrophy, constituting latent vulnerability traits to depression. In this group, called vulnerable, a subsequent mild stress evoked a rise of serum corticosterone levels and a "depressive" phenotype, in contrast to nonvulnerable animals. Intracerebroventricular administration of 7,8-dihydroxyflavone, a selective TrkB receptor agonist, dampened the development of the "depressive" phenotype. Our results thus characterize the presence of latent vulnerability traits that underlie the emergence of depression and identify the association of low BDNF with normal corticosterone serum concentrations as a predictive biomarker of vulnerability to depression.


Subject(s)
Biomarkers/metabolism , Brain/physiology , Depressive Disorder/pathology , Neuronal Plasticity/physiology , Animals , Antidepressive Agents, Tricyclic/pharmacology , Biomarkers/analysis , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation , Competitive Behavior , Corticosterone/blood , Depressive Disorder/physiopathology , Hippocampus/metabolism , Hippocampus/pathology , Hypothalamo-Hypophyseal System/physiopathology , Imipramine/pharmacology , Longitudinal Studies , Male , Neurons/pathology , Pituitary-Adrenal System/physiopathology , Rats , Rats, Sprague-Dawley , Social Environment , Stress, Psychological/metabolism , Swimming/psychology , Taste/physiology
14.
Eur J Neurosci ; 36(5): 2619-31, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22694179

ABSTRACT

Initial studies implicated the chemokine CXC motif ligand 12 (CXCL12) and its cognate CXC motif receptor 4 (CXCR4) in pain modulation. However, there has been no description of the distribution, transport and axonal sorting of CXCL12 and CXCR4 in rat nociceptive structures, and their direct participation in nociception modulation has not been demonstrated. Here, we report that acute intrathecal administration of CXCL12 induced mechanical hypersensitivity in naive rats. This effect was prevented by a CXCR4-neutralizing antibody. To determine the morphological basis of this behavioural response, we used light and electron microscopic immunohistochemistry to map CXCL12- and CXCR4-immunoreactive elements in dorsal root ganglia, lumbar spinal cord, sciatic nerve and skin. Light microscopy analysis revealed CXCL12 and CXCR4 immunoreactivity in calcitonin gene related peptide-containing peptidergic primary sensory neurons, which were both conveyed to central and peripheral sensory nerve terminals. Electron microscopy clearly demonstrated CXCL12 and CXCR4 immunoreactivity in primary sensory nerve terminals in the dorsal horn; both were sorted into small clear vesicles and large dense-core vesicles. This suggests that CXCL12 and CXCR4 are trafficked from nerve cell bodies to the dorsal horn. Double immunogold labelling for CXCL12 and calcitonin gene related peptide revealed partial vesicular colocalization in axonal terminals. We report, for the first time, that CXCR4 receptors are mainly located on the neuronal plasma membrane, where they are present at pre-synaptic and post-synaptic sites of central terminals. Receptor inactivation experiments, behavioural studies and morphological analyses provide strong evidence that the CXCL12/CXCR4 system is involved in modulation of nociceptive signalling.


Subject(s)
Chemokine CXCL12/analysis , Nociceptors/chemistry , Receptors, CXCR4/analysis , Animals , Male , Nociceptors/ultrastructure , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Sciatic Nerve/chemistry , Spinal Cord/chemistry
15.
J Neuroinflammation ; 9: 36, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22353423

ABSTRACT

BACKGROUND: Sustained neuroinflammation strongly contributes to the pathogenesis of pain. The clinical challenge of chronic pain relief led to the identification of molecules such as cytokines, chemokines and more recently matrix metalloproteinases (MMPs) as putative therapeutic targets. Evidence points to a founder member of the matricial CCN family, NOV/CCN3, as a modulator of these inflammatory mediators. We thus investigated the possible involvement of NOV in a preclinical model of persistent inflammatory pain. METHODS: We used the complete Freund's adjuvant (CFA)-induced model of persistent inflammatory pain and cultured primary sensory neurons for in vitro experiments. The mRNA expression of NOV and pro-inflammatory factors were measured with real-time quantitative PCR, CCL2 protein expression was assessed using ELISA, MMP-2 and -9 activities using zymography. The effect of drugs on tactile allodynia was evaluated by the von Frey test. RESULTS: NOV was expressed in neurons of both dorsal root ganglia (DRG) and dorsal horn of the spinal cord (DHSC). After intraplantar CFA injection, NOV levels were transiently and persistently down-regulated in the DRG and DHSC, respectively, occurring at the maintenance phase of pain (15 days). NOV-reduced expression was restored after treatment of CFA rats with dexamethasone. In vitro, results based on cultured DRG neurons showed that siRNA-mediated inhibition of NOV enhanced IL-1ß- and TNF-α-induced MMP-2, MMP-9 and CCL2 expression whereas NOV addition inhibited TNF-α-induced MMP-9 expression through ß1 integrin engagement. In vivo, the intrathecal delivery of MMP-9 inhibitor attenuated mechanical allodynia of CFA rats. Importantly, intrathecal administration of NOV siRNA specifically led to an up-regulation of MMP-9 in the DRG and MMP-2 in the DHSC concomitant with increased mechanical allodynia. Finally, NOV intrathecal treatment specifically abolished the induction of MMP-9 in the DRG and, MMP-9 and MMP-2 in the DHSC of CFA rats. This inhibitory effect on MMP is associated with reduced mechanical allodynia. CONCLUSIONS: This study identifies NOV as a new actor against inflammatory pain through regulation of MMPs thus uncovering NOV as an attractive candidate for therapeutic improvement in pain relief.


Subject(s)
Immediate-Early Proteins/metabolism , Inflammation/complications , Intercellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinase 2/metabolism , Pain/etiology , Pain/metabolism , Analysis of Variance , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cells, Cultured , Chemokine CCL2/metabolism , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Freund's Adjuvant , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Immediate-Early Proteins/genetics , Inflammation/chemically induced , Intercellular Signaling Peptides and Proteins/genetics , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/metabolism , Nerve Tissue Proteins/metabolism , Pain/drug therapy , Pain Measurement , Pain Threshold/drug effects , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Spinal Cord/pathology , Time Factors , Transfection , Up-Regulation/drug effects
16.
17.
J Cardiothorac Vasc Anesth ; 25(6): 917-25, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21641819

ABSTRACT

OBJECTIVE: One of the strategies to attenuate opioid-induced hyperalgesia (OIH) may be to decrease intraoperative doses of opioids by using target-controlled infusion (TCI). DESIGN: Double-blind and randomized study. SETTING: A single university hospital. PARTICIPANTS: Forty American Society of Anesthesiologists II to III patients scheduled for elective cardiac surgery. INTERVENTIONS: patients were randomized to 1 of the 2 groups: 1 group received an infusion of intraoperative remifentanil using TCI (target: 7 ng/mL), and the 2nd one was given an intraoperative continuous infusion (CI) (0.3 µg/kg/min). The anesthestic protocol and postoperative pain management were the same in both groups. The extent of mechanical dynamic hyperalgesia on the middle line perpendicular to the wound was considered the primary endpoint. The secondary endpoints were other results of dynamic and punctuate hyperalgesia until postoperative day 7, visual analog scale (VAS) and verbal rating scale (VRS) scores, and total morphine consumption until postoperative day 2. MEASUREMENTS AND MAIN RESULTS: Morphometric and demographic characteristics and duration of surgery were comparable in both groups. Intraoperative remifentanil consumption was greater in CI than in TCI group (5,329 [1,833] v 3,662 [1,160] µg, p = 0.003). During the first 44 hours, there were no differences in morphine consumption, VAS, and VRS. The extent of hyperalgesia was significantly lower on postoperative days 1, 2, and 4 in the TCI group than in the CI group on the 3 evaluated lines (p < 0.05). Punctuate hyperalgesia evaluating 3 different points was lower in the TCI than in the CI group from postoperative day 1 until postoperative day 7 (p < 0.05). CONCLUSIONS: The intraoperative decrease of opioid consumption when comparing the CI versus TCI mode of administration of remifentanil led to less OIH after cardiac surgery.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/therapeutic use , Cardiac Surgical Procedures/adverse effects , Hyperalgesia/prevention & control , Pain, Postoperative/prevention & control , Piperidines/administration & dosage , Piperidines/therapeutic use , Acetaminophen/therapeutic use , Aged , Anesthesia, General , Anesthesia, Intravenous , Blood Pressure/drug effects , Double-Blind Method , Drug Delivery Systems , Electroencephalography , Female , Heart Rate/drug effects , Humans , Male , Middle Aged , Morphine/therapeutic use , Pain Measurement/drug effects , Pain Threshold/drug effects , Physical Stimulation , Postoperative Nausea and Vomiting/epidemiology , Preanesthetic Medication , Remifentanil , Sternotomy
18.
Nat Commun ; 12(1): 2356, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33883545

ABSTRACT

Charcot-Marie-Tooth disease 1 A (CMT1A) results from a duplication of the PMP22 gene in Schwann cells and a deficit of myelination in peripheral nerves. Patients with CMT1A have reduced nerve conduction velocity, muscle wasting, hand and foot deformations and foot drop walking. Here, we evaluate the safety and efficacy of recombinant adeno-associated viral vector serotype 9 (AAV2/9) expressing GFP and shRNAs targeting Pmp22 mRNA in animal models of Charcot-Marie-Tooth disease 1 A. Intra-nerve delivery of AAV2/9 in the sciatic nerve allowed widespread transgene expression in resident myelinating Schwann cells in mice, rats and non-human primates. A bilateral treatment restore expression levels of PMP22 comparable to wild-type conditions, resulting in increased myelination and prevention of motor and sensory impairments over a twelve-months period in a rat model of CMT1A. We observed limited off-target transduction and immune response using the intra-nerve delivery route. A combination of previously characterized human skin biomarkers is able to discriminate between treated and untreated animals, indicating their potential use as part of outcome measures.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/therapy , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/genetics , Animals , Charcot-Marie-Tooth Disease/pathology , Dependovirus/genetics , Disease Models, Animal , Female , Gene Silencing , Genetic Therapy/methods , Genetic Vectors , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Rats , Rats, Mutant Strains , Schwann Cells/metabolism , Schwann Cells/pathology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology
19.
Eur J Neurosci ; 29(4): 727-37, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19200067

ABSTRACT

The clinically important opioid fentanyl, administered acutely, enhances mechanical hypersensitivity in a model of surgical pain induced by plantar incision. Activity of neurokinin-1 (NK-1) receptor-expressing ascending spinal neurons, descending pathways originating in the rostral ventromedial medulla (RVM), and spinal dynorphin are necessary for the development and maintenance of hyperalgesia during sustained morphine exposure, suggesting that these mechanisms may also be important in opioid enhancement of surgical pain. Therefore, we examined the roles of these mechanisms in sensory hypersensitivity produced by acute fentanyl administration in rats not undergoing surgical incision and in rats undergoing plantar incision. In non-operated rats, fentanyl induced analgesia followed by immediate and long-lasting sensory hypersensitivity, as previously described. Fentanyl also enhanced pain sensitivity induced by plantar incision. Ablation of NK-1-expressing spinal neurons by pre-treatment with substance P-Saporin reduced sensory hypersensitivity in fentanyl-treated rats and, to a lesser extent, in fentanyl-treated rats with a surgical incision. Microinjection of lidocaine into the RVM completely reversed fentanyl-induced sensory hypersensitivity and fentanyl enhancement of incision-induced sensory hypersensitivity. RVM lidocaine injection resulted in a slight reduction of incision-induced sensory hypersensitivity in the absence of fentanyl pre-treatment. Spinal dynorphin content increased by 30 +/- 7% and 66 +/- 17% in fentanyl- and fentanyl/incision-treated rats. Spinal administration of antiserum to dynorphin attenuated sensory hypersensitivity in fentanyl-treated rats. These data support a partial role of NK-1 receptor-containing ascending pathways and a crucial role of descending facilitatory pathways in fentanyl-induced hyperalgesia and in the enhanced hyperalgesia produced by fentanyl treatment following surgical incision.


Subject(s)
Medulla Oblongata/physiopathology , Neurons/metabolism , Pain, Postoperative/physiopathology , Receptors, Neurokinin-1/metabolism , Spinal Cord/physiopathology , Analgesics, Opioid/pharmacology , Anesthetics, Local/pharmacology , Animals , Disease Models, Animal , Dynorphins/metabolism , Fentanyl/pharmacology , Immunohistochemistry , Lidocaine/pharmacology , Male , Neural Pathways/physiopathology , Pain Threshold/drug effects , Pain, Postoperative/chemically induced , Pain, Postoperative/drug therapy , Rats , Rats, Sprague-Dawley
20.
J Neurochem ; 107(1): 50-60, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18636982

ABSTRACT

Peripheral nerve lesion leads to the production of interleukin 6 (IL-6)-related neuropoietic cytokines involved in nerve protection and regeneration. This family of cytokines mainly signal through the signal transducer and activator of transcription (STAT) pathway that is locally activated in injured nerves. IL-6 is also involved in pain that frequently arises from peripheral nerve lesion. We investigated the possible activation of this major IL-6 signaling system in the spinal cord after peripheral nerve injury and its role in neuropathic pain. Ligation of L5-L6 spinal nerves (SNL) evoked an accumulation of active, phosphorylated form of STAT3 in microglial cells of dorsal spinal cord mostly in projection areas of injured nerves. SNL resulted also in a massive induction of IL-6 mRNA expression in dorsal root ganglia and increased concentration of IL-6 in dorsal spinal cord. Intrathecal injection of anti-rat IL-6 antibodies prevented the SNL-induced accumulation of phospho-STAT3 in the spinal cord. STAT3 pathway blockade with Janus kinase 2 inhibitor AG490 attenuated both mechanical allodynia and thermal hyperalgesia in SNL rats. These data show that in response to SNL injury Janus kinase/STAT3 system is activated mainly through IL-6 signaling in spinal microglia and that this transduction pathway participates in development of pain associated with nerve alteration.


Subject(s)
Interleukin-6/metabolism , Janus Kinase 1/metabolism , Microglia/metabolism , Peripheral Nervous System Diseases/physiopathology , STAT3 Transcription Factor/metabolism , Spinal Cord/physiopathology , Animals , Antibodies/pharmacology , Disease Models, Animal , Enzyme Activation/physiology , Hyperalgesia/enzymology , Hyperalgesia/physiopathology , Interleukin-6/genetics , Interleukin-6/immunology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Ligation , Male , Microglia/enzymology , Peripheral Nerves/enzymology , Peripheral Nerves/physiopathology , Peripheral Nervous System Diseases/enzymology , Peripheral Nervous System Diseases/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Spinal Cord/enzymology , Spinal Cord/metabolism , Spinal Nerves/injuries , Spinal Nerves/physiopathology , Spinal Nerves/surgery , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL