Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Brain Behav Immun ; 123: 11-27, 2024 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-39218234

RESUMEN

Neuroimmune signaling is a key process underlying neuropathic pain. Clinical studies have demonstrated that 18 kDa translocator protein (TSPO), a putative marker of neuroinflammation, is upregulated in discrete brain regions of patients with chronic pain. However, no preclinical studies have investigated TSPO dynamics in the brain in the context of neuropathic pain and in response to analgesic treatments. We used positron emission tomography-computed tomography (PET-CT) and [18F]-PBR06 radioligand to measure TSPO levels in the brain across time after chronic constriction injury (CCI) of the sciatic nerve in both male and female rats. Up to 10 weeks post-CCI, TSPO expression was increased in discrete brain regions, including medial prefrontal cortex, somatosensory cortex, insular cortex, anterior cingulate cortex, motor cortex, ventral tegmental area, amygdala, midbrain, pons, medulla, and nucleus accumbens. TSPO was broadly upregulated across these regions at 4 weeks post CCI in males, and 10 weeks in females, though there were regional differences between the sexes. Using immunohistochemistry, we confirmed TSPO expression in these regions. We further demonstrated that TSPO was upregulated principally in microglia in the nucleus accumbens core, and astrocytes and endothelial cells in the nucleus accumbens shell. Finally, we tested whether TSPO upregulation was sensitive to diroximel fumarate, a drug that induces endogenous antioxidants via nuclear factor E2-related factor 2 (Nrf2). Diroximel fumarate alleviated neuropathic pain and reduced TSPO upregulation. Our findings indicate that TSPO is upregulated over the course of neuropathic pain development and is resolved by an antinociceptive intervention in animals with peripheral nerve injury.

2.
Brain Behav Immun ; 115: 157-168, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37838078

RESUMEN

Females represent a majority of chronic pain patients and show greater inflammatory immune responses in human chronic pain patient populations as well as in animal models of neuropathic pain. Recent discoveries in chronic pain research have revealed sex differences in inflammatory signaling, a key component of sensory pathology in chronic neuropathic pain, inviting more research into the nuances of these sex differences. Here we use the chronic constriction injury (CCI) model to explore similarities and differences in expression and production of Inflammatory cytokine IL-1beta in the lumbar spinal cord, as well as its role in chronic pain. We have discovered that intrathecal IL-1 receptor antagonist reverses established pain in both sexes, and increased gene expression of inflammasome NLRP3 is specific to microglia and astrocytes rather than neurons, while IL-1beta is specific to microglia in both sexes. We report several sex differences in the expression level of the genes coding for IL-1beta, as well as the four inflammasomes responsible for IL-1beta release: NLRP3, AIM2, NLRP1, and NLRC4 in the spinal cord. Total mRNA, but not protein expression of IL-1beta is greater in females than males after CCI. Also, while CCI increases all four inflammasomes in both sexes, there are sex differences in relative levels of inflammasome expression. NLRP3 and AIM2 are more highly expressed in females, whereas NLRP1 expression is greater in males.


Asunto(s)
Dolor Crónico , Inflamasomas , Interleucina-1beta , Neuralgia , Animales , Femenino , Humanos , Masculino , Ratas , Dolor Crónico/metabolismo , Constricción , Proteínas de Unión al ADN/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Neuralgia/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Médula Espinal/metabolismo
3.
Brain Behav Immun ; 115: 419-431, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37924957

RESUMEN

Regular aerobic activity is associated with a reduced risk of chronic pain in humans and rodents. Our previous studies in rodents have shown that prior voluntary wheel running can normalize redox signaling at the site of peripheral nerve injury, attenuating subsequent neuropathic pain. However, the full extent of neuroprotection offered by voluntary wheel running after peripheral nerve injury is unknown. Here, we show that six weeks of voluntary wheel running prior to chronic constriction injury (CCI) reduced the terminal complement membrane attack complex (MAC) at the sciatic nerve injury site. This was associated with increased expression of the MAC inhibitor CD59. The levels of upstream complement components (C3) and their inhibitors (CD55, CR1 and CFH) were altered by CCI, but not increased by voluntary wheel running. Since MAC can degrade myelin, which in turn contributes to neuropathic pain, we evaluated myelin integrity at the sciatic nerve injury site. We found that the loss of myelinated fibers and decreased myelin protein which occurs in sedentary rats following CCI was not observed in rats with prior running. Substitution of prior voluntary wheel running with exogenous CD59 also attenuated mechanical allodynia and reduced MAC deposition at the nerve injury site, pointing to CD59 as a critical effector of the neuroprotective and antinociceptive actions of prior voluntary wheel running. This study links attenuation of neuropathic pain by prior voluntary wheel running with inhibition of MAC and preservation of myelin integrity at the sciatic nerve injury site.


Asunto(s)
Neuralgia , Traumatismos de los Nervios Periféricos , Neuropatía Ciática , Humanos , Ratas , Animales , Vaina de Mielina/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento , Actividad Motora/fisiología , Traumatismos de los Nervios Periféricos/complicaciones , Hiperalgesia/metabolismo , Neuralgia/complicaciones , Nervio Ciático/lesiones
4.
J Neurosci ; 42(42): 7862-7874, 2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-36096670

RESUMEN

Peripheral neuropathic pain induced by the chemotherapeutic cisplatin can persist for months to years after treatment. Histone deacetylase 6 (HDAC6) inhibitors have therapeutic potential for cisplatin-induced neuropathic pain since they persistently reverse mechanical hypersensitivity and spontaneous pain in rodent models. Here, we investigated the mechanisms underlying reversal of mechanical hypersensitivity in male and female mice by a 2 week treatment with an HDAC6 inhibitor, administered 3 d after the last dose of cisplatin. Mechanical hypersensitivity in animals of both sexes treated with the HDAC6 inhibitor was temporarily reinstated by a single injection of the neutral opioid receptor antagonist 6ß-naltrexol or the peripherally restricted opioid receptor antagonist naloxone methiodide. These results suggest that tonic peripheral opioid ligand-receptor signaling mediates reversal of cisplatin-induced mechanical hypersensitivity after treatment with an HDAC6 inhibitor. Pointing to a specific role for δ opioid receptors (DORs), Oprd1 expression was decreased in DRG neurons following cisplatin administration, but normalized after treatment with an HDAC6 inhibitor. Mechanical hypersensitivity was temporarily reinstated in both sexes by a single injection of the DOR antagonist naltrindole. Consistently, HDAC6 inhibition failed to reverse cisplatin-induced hypersensitivity when DORs were genetically deleted from advillin+ neurons. Mechanical hypersensitivity was also temporarily reinstated in both sexes by a single injection of a neutralizing antibody against the DOR ligand met-enkephalin. In conclusion, we reveal that treatment with an HDAC6 inhibitor induces tonic enkephalin-DOR signaling in peripheral sensory neurons to suppress mechanical hypersensitivity.SIGNIFICANCE STATEMENT Over one-fourth of cancer survivors suffer from intractable painful chemotherapy-induced peripheral neuropathy (CIPN), which can last for months to years after treatment ends. HDAC6 inhibition is a novel strategy to reverse CIPN without negatively interfering with tumor growth, but the mechanisms responsible for persistent reversal are not well understood. We built on evidence that the endogenous opioid system contributes to the spontaneous, apparent resolution of pain caused by nerve damage or inflammation, referred to as latent sensitization. We show that blocking the δ opioid receptor or its ligand enkephalin unmasks CIPN in mice treated with an HDAC6 inhibitor (latent sensitization). Our work provides insight into the mechanisms by which treatment with an HDAC6 inhibitor apparently reverses CIPN.


Asunto(s)
Antineoplásicos , Neuralgia , Ratones , Masculino , Femenino , Animales , Histona Desacetilasa 6/metabolismo , Cisplatino/toxicidad , Receptores Opioides delta , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Antagonistas de Narcóticos/farmacología , Ligandos , Analgésicos Opioides/efectos adversos , Ratones Endogámicos C57BL , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Inhibidores de Histona Desacetilasas , Niacinamida , Antineoplásicos/toxicidad , Encefalina Metionina , Encefalinas , Anticuerpos Neutralizantes
5.
J Neurosci ; 41(5): 855-865, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33239404

RESUMEN

Chronic pain, encompassing conditions, such as low back pain, arthritis, persistent post-surgical pain, fibromyalgia, and neuropathic pain disorders, is highly prevalent but remains poorly treated. The vast majority of therapeutics are directed solely at neurons, despite the fact that signaling between immune cells, glia, and neurons is now recognized as indispensable for the initiation and maintenance of chronic pain. This review highlights recent advances in understanding fundamental neuroimmune signaling mechanisms and novel therapeutic targets in rodent models of chronic pain. We further discuss new technological developments to study, diagnose, and quantify neuroimmune contributions to chronic pain in patient populations.


Asunto(s)
Autoanticuerpos/inmunología , Dolor Crónico/inmunología , Modelos Animales de Enfermedad , Neuroglía/inmunología , Neuroinmunomodulación/fisiología , Neuronas/inmunología , Animales , Autoanticuerpos/metabolismo , Dolor Crónico/metabolismo , Humanos , Factor 2 Relacionado con NF-E2/inmunología , Factor 2 Relacionado con NF-E2/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Roedores
6.
J Neurosci Res ; 100(1): 265-277, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-32533604

RESUMEN

The present series of studies examine the impact of systemically administered therapeutics on peripheral nerve injury (males; unilateral sciatic chronic constriction injury [CCI])-induced suppression of voluntary wheel running, across weeks after dosing cessation. Following CCI, active phase running distance and speed are suppressed throughout the 7-week observation period. A brief course of morphine, however, increased active phase running distance and speed throughout this same period, an effect apparent only in sham rats. For CCI rats, systemic co-administration of morphine with antagonists of either P2X7 (A438079) or TLR4 ((+)-naloxone) (receptors critical to the activation of NLRP3 inflammasomes and consequent inflammatory cascades) returned running behavior of CCI rats to that of shams through 5+ weeks after dosing ceased. This is a striking difference in effect compared to our prior CCI allodynia results using systemic morphine plus intrathecal delivery of these same antagonists, wherein a sustained albeit partial suppression of neuropathic pain was observed. This may point to actions of the systemic drugs at multiple sites along the neuraxis, modulating injury-induced, inflammasome-mediated effects at the injured sciatic nerve and/or dorsal root ganglia, spinal cord, and potentially higher levels. Given that our data to date point to morphine amplifying neuroinflammatory processes put into motion by nerve injury, it is intriguing to speculate that co-administration of TLR4 and/or P2X7 antagonists can intervene in these inflammatory processes in a beneficial way. That is, that systemic administration of such compounds may suppress inflammatory damage at multiple sites, rapidly and persistently returning neuropathic animals to sham levels of response.


Asunto(s)
Morfina , Neuralgia , Animales , Constricción , Intervención en la Crisis (Psiquiatría) , Hiperalgesia/tratamiento farmacológico , Masculino , Morfina/farmacología , Actividad Motora , Neuralgia/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Nervio Ciático , Receptor Toll-Like 4
7.
Brain Behav Immun ; 91: 418-428, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33127584

RESUMEN

More than a quarter of veterans of the 1990-1991 Persian Gulf War suffer from Gulf War Illness (GWI), a chronic, multi-symptom illness that commonly includes musculoskeletal pain. Exposure to a range of toxic chemicals, including sarin nerve agent, are a suspected root cause of GWI. Moreover, such chemical exposures induce a neuroinflammatory response in rodents, which has been linked to several GWI symptoms in rodents and veterans with GWI. To date, a neuroinflammatory basis for pain associated with GWI has not been investigated. Here, we evaluated development of nociceptive hypersensitivity in a model of GWI. Male Sprague Dawley rats were treated with corticosterone in the drinking water for 7 days, to mimic high physiological stress, followed by a single injection of the sarin nerve agent surrogate, diisopropyl fluorophosphate. These exposures alone were insufficient to induce allodynia. However, an additional sub-threshold challenge (a single intramuscular injection of pH 4 saline) induced long-lasting, bilateral allodynia. Such allodynia was associated with elevation of markers for activated microglia/macrophages (CD11b) and astrocytes/satellite glia (GFAP) in the lumbar dorsal spinal cord and dorsal root ganglia (DRG). Additionally, Toll-like receptor 4 (TLR4) mRNA was elevated in the lumbar dorsal spinal cord, while IL-1ß and IL-6 were elevated in the lumbar dorsal spinal cord, DRG, and gastrocnemius muscle. Demonstrating a casual role for such neuroinflammatory signaling, allodynia was reversed by treatment with either minocycline, the TLR4 inhibitor (+)-naltrexone, or IL-10 plasmid DNA. Together, these results point to a role for neuroinflammation in male rats in the model of musculoskeletal pain related to GWI. Therapies that alleviate persistent immune dysregulation may be a strategy to treat pain and other symptoms of GWI.


Asunto(s)
Síndrome del Golfo Pérsico , Animales , Modelos Animales de Enfermedad , Guerra del Golfo , Masculino , Dolor , Síndrome del Golfo Pérsico/inducido químicamente , Ratas , Ratas Sprague-Dawley
8.
FASEB J ; 33(8): 9577-9587, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31162938

RESUMEN

Deregulation of innate immune TLR4 signaling contributes to various diseases including neuropathic pain and drug addiction. Naltrexone is one of the rare TLR4 antagonists with good blood-brain barrier permeability and showing no stereoselectivity for TLR4. By linking 2 naltrexone units through a rigid pyrrole spacer, the bivalent ligand norbinaltorphimine was formed. Interestingly, (+)-norbinaltorphimine [(+)-1] showed ∼25 times better TLR4 antagonist activity than naltrexone in microglial BV-2 cell line, whereas (-)-norbinaltorphimine [(-)-1] lost TLR4 activity. The enantioselectivity of norbinaltorphimine was further confirmed in primary microglia, astrocytes, and macrophages. The activities of meso isomer of norbinaltorphimine and the molecular dynamic simulation results demonstrate that the stereochemistry of (+)-1 is derived from the (+)-naltrexone pharmacophore. Moreover, (+)-1 significantly increased and prolonged morphine analgesia in vivo. The efficacy of (+)-1 is long lasting. This is the first report showing enantioselective modulation of the innate immune TLR signaling.-Zhang, X., Peng, Y., Grace, P. M., Metcalf, M. D., Kwilasz, A. J., Wang, Y., Zhang, T., Wu, S., Selfridge, B. R., Portoghese, P. S., Rice, K. C., Watkins, L. R., Hutchinson, M. R., Wang, X. Stereochemistry and innate immune recognition: (+)-norbinaltorphimine targets myeloid differentiation protein 2 and inhibits toll-like receptor 4 signaling.


Asunto(s)
Antígeno 96 de los Linfocitos/metabolismo , Naltrexona/análogos & derivados , Receptor Toll-Like 4/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Interleucina-1beta/metabolismo , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Naltrexona/química , Naltrexona/farmacología , Estructura Secundaria de Proteína , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Estereoisomerismo , Relación Estructura-Actividad , Receptor Toll-Like 4/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
9.
Anesthesiology ; 132(2): 343-356, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31939850

RESUMEN

BACKGROUND: Available treatments for neuropathic pain have modest efficacy and significant adverse effects, including abuse potential. Because oxidative stress is a key mechanistic node for neuropathic pain, the authors focused on the master regulator of the antioxidant response-nuclear factor erythroid 2-related factor 2 (NFE2L2; Nrf2)-as an alternative target for neuropathic pain. The authors tested whether dimethyl fumarate (U.S. Food and Drug Administration-approved treatment for multiple sclerosis) would activate NFE2L2 and promote antioxidant activity to reverse neuropathic pain behaviors and oxidative stress-dependent mechanisms. METHODS: Male Sprague Dawley rats, and male and female wild type and Nfe2l2 mice were treated with oral dimethyl fumarate/vehicle for 5 days (300 mg/kg; daily) after spared nerve injury/sham surgery (n = 5 to 8 per group). Allodynia was measured in von Frey reflex tests and hyperalgesia in operant conflict-avoidance tests. Ipsilateral L4/5 dorsal root ganglia were assayed for antioxidant and cytokine/chemokine levels, and mitochondrial bioenergetic capacity. RESULTS: Dimethyl fumarate treatment reversed mechanical allodynia (injury-vehicle, 0.45 ± 0.06 g [mean ± SD]; injury-dimethyl fumarate, 8.2 ± 0.16 g; P < 0.001) and hyperalgesia induced by nerve injury (injury-vehicle, 2 of 6 crossed noxious probes; injury-dimethyl fumarate, 6 of 6 crossed; P = 0.013). The antiallodynic effect of dimethyl fumarate was lost in nerve-injured Nfe2l2 mice, but retained in nerve-injured male and female wild type mice (wild type, 0.94 ± 0.25 g; Nfe2l2, 0.02 ± 0.01 g; P < 0.001). Superoxide dismutase activity was increased by dimethyl fumarate after nerve injury (injury-vehicle, 3.96 ± 1.28 mU/mg; injury-dimethyl fumarate, 7.97 ± 0.47 mU/mg; P < 0.001). Treatment reduced the injury-dependent increases in cytokines and chemokines, including interleukin-1ß (injury-vehicle, 13.30 ± 2.95 pg/mg; injury-dimethyl fumarate, 6.33 ± 1.97 pg/mg; P = 0.022). Injury-impaired mitochondrial bioenergetics, including basal respiratory capacity, were restored by dimethyl fumarate treatment (P = 0.025). CONCLUSIONS: Dimethyl fumarate, a nonopioid and orally-bioavailable drug, alleviated nociceptive hypersensitivity induced by peripheral nerve injury via activation of NFE2L2 antioxidant signaling. Dimethyl fumarate also resolved neuroinflammation and mitochondrial dysfunction-oxidative stress-dependent mechanisms that drive nociceptive hypersensitivity after nerve injury.


Asunto(s)
Antioxidantes/metabolismo , Dimetilfumarato/uso terapéutico , Factor 2 Relacionado con NF-E2/metabolismo , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Animales , Dimetilfumarato/farmacología , Femenino , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Masculino , Ratones , Ratones Noqueados , Ratas , Ratas Sprague-Dawley , Roedores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
10.
Brain Behav Immun ; 82: 432-444, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31542403

RESUMEN

There is growing interest in drug repositioning to find new therapeutic indications for drugs already approved for use in people. Lovastatin is an FDA approved drug that has been used clinically for over a decade as a lipid-lowering medication. While lovastatin is classically considered to act as a hydroxymethylglutaryl (HMG)-CoA reductase inhibitor, the present series of studies reveal a novel lovastatin effect, that being as a Toll-like receptor 4 (TLR4) antagonist. Lovastatin selectively inhibits lipopolysaccharide (LPS)-induced TLR4-NF-κB activation without affecting signaling by other homologous TLRs. In vitro biophysical binding and cellular thermal shift assay (CETSA) show that lovastatin is recognized by TLR4's coreceptor myeloid differentiation protein 2 (MD-2). This finding is supported by molecular dynamics simulations that lovastatin targets the LPS binding pocket of MD-2 and lovastatin binding stabilizes the MD-2 conformation. In vitro studies of BV-2 microglial cells revealed that lovastatin inhibits multiple effects of LPS, including activation of NFkB; mRNA expression of tumor necrosis factor-a, interleukin-6 and cyclo-oxygenase 2; production of nitric oxide and reactive oxygen species; as well as phagocytic activity. Furthermore, intrathecal delivery of lovastatin over lumbosacral spinal cord of rats attenuated both neuropathic pain from sciatic nerve injury and expression of the microglial activation marker CD11 in lumbar spinal cord dorsal horn. Given the well-established role of microglia and proinflammatory signaling in neuropathic pain, these data are supportive that lovastatin, as a TLR4 antagonist, may be productively repurposed for treating chronic pain.


Asunto(s)
Lovastatina/farmacología , Neuralgia/metabolismo , Receptor Toll-Like 4/efectos de los fármacos , Animales , Ciclooxigenasa 2/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Interleucina-1beta/metabolismo , Lovastatina/metabolismo , Antígeno 96 de los Linfocitos/metabolismo , Antígeno 96 de los Linfocitos/fisiología , Masculino , Ratones , Microglía/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Cultivo Primario de Células , Células RAW 264.7 , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Asta Dorsal de la Médula Espinal/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Anesth Analg ; 128(1): 161-167, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29596097

RESUMEN

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.


Asunto(s)
Analgésicos Opioides/toxicidad , Hiperalgesia/inducido químicamente , Morfina/toxicidad , Umbral del Dolor/efectos de los fármacos , Dolor Postoperatorio/inducido químicamente , Células del Asta Posterior/efectos de los fármacos , Analgésicos Opioides/administración & dosificación , Animales , Modelos Animales de Enfermedad , Esquema de Medicación , Hiperalgesia/diagnóstico por imagen , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Mediadores de Inflamación/metabolismo , Laparotomía , Masculino , Morfina/administración & dosificación , Dimensión del Dolor , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/metabolismo , Dolor Postoperatorio/fisiopatología , Células del Asta Posterior/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
12.
Proc Natl Acad Sci U S A ; 113(24): E3441-50, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27247388

RESUMEN

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.


Asunto(s)
Dolor Crónico/metabolismo , Inflamasomas/metabolismo , Microglía/metabolismo , Morfina/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuralgia/metabolismo , Asta Dorsal de la Médula Espinal/metabolismo , Animales , Dolor Crónico/patología , Dolor Crónico/fisiopatología , Clozapina/análogos & derivados , Clozapina/farmacología , Interleucina-1beta/metabolismo , Masculino , Microglía/patología , Neuralgia/patología , Neuralgia/fisiopatología , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Asta Dorsal de la Médula Espinal/patología , Asta Dorsal de la Médula Espinal/fisiopatología
13.
Brain Behav Immun ; 72: 45-50, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28860068

RESUMEN

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.


Asunto(s)
Alarminas/fisiología , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Traumatismos Vertebrales/inmunología , Alarminas/efectos de los fármacos , Animales , Caspasa 1/metabolismo , Proteína HMGB1/metabolismo , Hiperalgesia/metabolismo , Inflamasomas/metabolismo , Inyecciones Espinales , Interleucina-1beta/metabolismo , Masculino , Morfina/metabolismo , Morfina/uso terapéutico , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuralgia/metabolismo , Ratas , Ratas Endogámicas F344 , Receptores Purinérgicos P2X7/metabolismo , Traumatismos Vertebrales/tratamiento farmacológico , Receptor Toll-Like 4/metabolismo
14.
Brain Behav Immun ; 64: 59-64, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27993689

RESUMEN

Despite pain being a sensory experience, studies of spinal cord ventral root damage have demonstrated that motor neuron injury can induce neuropathic pain. Whether injury of cranial motor nerves can also produce nociceptive hypersensitivity has not been addressed. Herein, we demonstrate that chronic constriction injury (CCI) of the buccal branch of the facial nerve results in long-lasting, unilateral allodynia in the rat. An anterograde and retrograde tracer (3000MW tetramethylrhodamine-conjugated dextran) was not transported to the trigeminal ganglion when applied to the injury site, but was transported to the facial nucleus, indicating that this nerve branch is not composed of trigeminal sensory neurons. Finally, intracisterna magna injection of interleukin-1 (IL-1) receptor antagonist reversed allodynia, implicating the pro-inflammatory cytokine IL-1 in the maintenance of neuropathic pain induced by facial nerve CCI. These data extend the prior evidence that selective injury to motor axons can enhance pain to supraspinal circuits by demonstrating that injury of a facial nerve with predominantly motor axons is sufficient for neuropathic pain, and that the resultant pain has a neuroimmune component.


Asunto(s)
Traumatismos del Nervio Facial/fisiopatología , Nervio Facial/fisiopatología , Hiperalgesia/fisiopatología , Neuralgia/fisiopatología , Animales , Nervio Facial/citología , Traumatismos del Nervio Facial/complicaciones , Núcleo Motor del Nervio Facial/citología , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Neuralgia/etiología , Neuralgia/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Ratas Sprague-Dawley , Receptores de Interleucina-1/antagonistas & inhibidores
15.
Brain Behav Immun ; 59: 49-54, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27189037

RESUMEN

Relapsing-remitting multiple sclerosis is commonly associated with motor impairments, neuropathic pain, fatigue, mood disorders, and decreased life expectancy. However, preclinical pharmacological studies predominantly rely on clinical scoring of motor deficit as the sole behavioral endpoint. Thus, the translational potential of these studies is limited. Here, we have assessed the therapeutic potential of a novel anti-inflammatory interleukin-10 (IL-10) non-viral gene therapy formulation (XT-101-R) in a rat relapsing remitting experimental autoimmune encephalomyelitis (EAE) model. EAE induced motor deficits and neuropathic pain as reflected by induction of low-threshold mechanical allodynia, suppressed voluntary wheel running, decreased social exploration, and was associated with markedly enhanced mortality. We also noted that voluntary wheel running was depressed prior to the onset of motor deficit, and may therefore serve as a predictor of clinical symptoms onset. XT-101-R was intrathecally dosed only once at the onset of motor deficits, and attenuated each of the EAE-induced symptoms and improved survival, relative to vehicle control. This is the first pharmacological assessment of such a broad range of EAE symptoms, and provides support for IL-10 gene therapy as a clinical strategy for the treatment of multiple sclerosis.


Asunto(s)
Ansiedad/psicología , Ansiedad/terapia , Conducta Animal/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/psicología , Encefalomielitis Autoinmune Experimental/terapia , Fatiga/psicología , Fatiga/terapia , Interleucina-10/genética , Neuralgia/psicología , Neuralgia/terapia , Animales , Conducta Exploratoria , Terapia Genética , Hiperalgesia/psicología , Hiperalgesia/terapia , Inyecciones Espinales , Relaciones Interpersonales , Esperanza de Vida , Masculino , Actividad Motora , Ratas
16.
Cell Mol Life Sci ; 73(22): 4249-4264, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27314883

RESUMEN

Substance P is a peptide mainly secreted by neurons and is involved in many biological processes, including nociception and inflammation. Animal models have provided insights into the biology of this peptide and offered compelling evidence for the importance of substance P in cell-to-cell communication by either paracrine or endocrine signaling. Substance P mediates interactions between neurons and immune cells, with nerve-derived substance P modulating immune cell proliferation rates and cytokine production. Intriguingly, some immune cells have also been found to secrete substance P, which hints at an integral role of substance P in the immune response. These communications play important functional roles in immunity including mobilization, proliferation and modulation of the activity of immune cells. This review summarizes current knowledge of substance P and its receptors, as well as its physiological and pathological roles. We focus on recent developments in the immunobiology of substance P and discuss the clinical implications of its ability to modulate the immune response.


Asunto(s)
Inmunidad , Sustancia P/metabolismo , Secuencia de Aminoácidos , Animales , Enfermedad , Humanos , Inmunomodulación , Transducción de Señal , Sustancia P/química
17.
Brain Behav Immun ; 58: 348-356, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27519154

RESUMEN

Central neuropathic pain (CNP) is a pervasive, debilitating problem that impacts thousands of people living with central nervous system disorders, including spinal cord injury (SCI). Current therapies for treating this type of pain are ineffective and often have dose-limiting side effects. Although opioids are one of the most commonly used CNP treatments, recent animal literature has indicated that administering opioids shortly after a traumatic injury can actually have deleterious effects on long-term health and recovery. In order to study the deleterious effects of administering morphine shortly after trauma, we employed our low thoracic (T13) dorsal root avulsion model (Spinal Neuropathic Avulsion Pain, SNAP). Administering a weeklong course of 10mg/kg/day morphine beginning 24h after SNAP resulted in amplified mechanical allodynia. Co-administering the non-opioid toll-like receptor 4 (TLR4) antagonist (+)-naltrexone throughout the morphine regimen prevented morphine-induced amplification of SNAP. Exploration of changes induced by early post-trauma morphine revealed that this elevated gene expression of TLR4, TNF, IL-1ß, and NLRP3, as well as IL-1ß protein at the site of spinal cord injury. These data suggest that a short course of morphine administered early after spinal trauma can exacerbate CNP in the long term. TLR4 initiates this phenomenon and, as such, may be potential therapeutic targets for preventing the deleterious effects of administering opioids after traumatic injury.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Hiperalgesia/metabolismo , Morfina/administración & dosificación , Neuralgia/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Hiperalgesia/complicaciones , Masculino , Naltrexona/administración & dosificación , Neuralgia/complicaciones , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/complicaciones , Receptor Toll-Like 4/antagonistas & inhibidores
18.
Brain Behav Immun ; 48: 115-22, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25774010

RESUMEN

Despite dramatic improvement in cardiopulmonary resuscitation (CPR) and other techniques for cardiac arrest (CA), the majority of survivors continue to show signs of decreased memory or executive cognitive function. Such memory impairment may be due to hippocampal CA1 neuronal death, which is delayed by several days after CA/CPR. Classical microgliosis in the CA1 region may contribute to neuronal death, yet the role of a key activation receptor Toll Like Receptor 4 (TLR4) has not been previously investigated for such neuronal death after CA/CPR. We show that (+)-naltrexone was neuroprotective after CA/CPR. TLR4 blockade was associated with decreased expression of markers for microglial/macrophage activation and T cell and B cell infiltration, as well as decreased pro-inflammatory cytokine levels. Notably, IL-10 expression was elevated in response to CA/CPR, but was not attenuated by (+)-naltrexone, suggesting that the local monocyte/microglial phenotype had shifted towards alternative activation. This was confirmed by elevated expression of Arginase-1, and decreased expression of NFκB p65 subunit. Thus, (+)-naltrexone and other TLR4 antagonists may represent a novel therapeutic strategy to alleviate the substantial burden of memory or executive cognitive function impairment after CA/CPR.


Asunto(s)
Muerte Celular/efectos de los fármacos , Paro Cardíaco/patología , Hipocampo/efectos de los fármacos , Naltrexona/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Reanimación Cardiopulmonar , Paro Cardíaco/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Interleucina-10/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología
19.
Headache ; 55(4): 475-89, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25833219

RESUMEN

Despite being the mainstay of pain management, opioids are limited in their clinical utility by adverse effects, such as tolerance and paradoxical hyperalgesia. Research of the past 15 years has extended beyond neurons, to implicate central nervous system immune signaling in these adverse effects. This article will provide an overview of these central immune mechanisms in opioid tolerance and paradoxical hyperalgesia, including those mediated by Toll-like receptor 4, purinergic, ceramide, and chemokine signaling. Challenges for the future, as well as new lines of investigation will be highlighted.


Asunto(s)
Analgésicos Opioides/farmacología , Sistema Nervioso Central/inmunología , Tolerancia a Medicamentos/inmunología , Transducción de Señal/inmunología , Analgesia/efectos adversos , Analgésicos Opioides/efectos adversos , Animales , Sistema Nervioso Central/efectos de los fármacos , Humanos , Hiperalgesia/inducido químicamente , Hiperalgesia/inmunología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/inmunología
20.
FASEB J ; 27(7): 2713-22, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23568774

RESUMEN

Rifampin has been used for the treatment of bacterial infections for many years. Clinically, rifampin has been found to possess immunomodulatory effects. However, the molecular target responsible for the immunosuppressive effects of rifampin is not known. Herein, we show that rifampin binds to myeloid differentiation protein 2 (MD-2), the key coreceptor for innate immune TLR4. Rifampin blocked TLR4 signaling induced by LPS, including NF-κB activation and the overproduction of proinflammatory mediators nitric oxide, interleukin 1ß, and tumor necrosis factor α in mouse microglia BV-2 cells and macrophage RAW 264.7 cells. Rifampin's inhibition of TLR4 signaling was also observed in immunocompetent rat primary macrophage, microglia, and astrocytes. Further, we show that rifampin (75 or 100 mg/kg b.i.d. for 3 d, intraperitoneal) suppressed allodynia induced by chronic constriction injury of the sciatic nerve and suppressed nerve injury-induced activation of microglia. Our findings indicate that MD-2 is a important target of rifampin in its inhibition of innate immune function and contributes to its clinically observed immune-suppressive effect. The results also suggest that rifampin may be repositioned as an agent for the treatment of neuropathic pain.


Asunto(s)
Antígeno 96 de los Linfocitos/metabolismo , Neuralgia/prevención & control , Rifampin/farmacología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Línea Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Hiperalgesia/prevención & control , Interleucina-1beta/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Microglía/citología , Microglía/efectos de los fármacos , Microglía/metabolismo , FN-kappa B/metabolismo , Neuralgia/metabolismo , Neuralgia/fisiopatología , Óxido Nítrico/metabolismo , Unión Proteica , Ratas , Ratas Sprague-Dawley , Rifampin/administración & dosificación , Rifampin/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA