Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Neurosci ; 39(7): 1139-1149, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30559153

RESUMEN

Clinical studies indicate that psychosocial stress contributes to adverse chronic pain outcomes in patients, but it is unclear how this is initiated or amplified by stress. Repeated social defeat (RSD) is a mouse model of psychosocial stress that activates microglia, increases neuroinflammatory signaling, and augments pain and anxiety-like behaviors. We hypothesized that activated microglia within the spinal cord facilitate increased pain sensitivity following RSD. Here we show that mechanical allodynia in male mice was increased with exposure to RSD. This stress-induced behavior corresponded with increased mRNA expression of several inflammatory genes, including IL-1ß, TNF-α, CCL2, and TLR4 in the lumbar spinal cord. While there were several adhesion and chemokine-related genes increased in the lumbar spinal cord after RSD, there was no accumulation of monocytes or neutrophils. Notably, there was evidence of microglial activation selectively within the nociceptive neurocircuitry of the dorsal horn of the lumbar cord. Elimination of microglia using the colony stimulating factor 1 receptor antagonist PLX5622 from the brain and spinal cord prevented the development of mechanical allodynia in RSD-exposed mice. Microglial elimination also attenuated RSD-induced IL-1ß, CCR2, and TLR4 mRNA expression in the lumbar spinal cord. Together, RSD-induced allodynia was associated with microglia-mediated inflammation within the dorsal horn of the lumbar spinal cord.SIGNIFICANCE STATEMENT Mounting evidence indicates that psychological stress contributes to the onset and progression of adverse nociceptive conditions. We show here that repeated social defeat stress causes increased pain sensitivity due to inflammatory signaling within the nociceptive circuits of the spinal cord. Studies here mechanistically tested the role of microglia in the development of pain by stress. Pharmacological ablation of microglia prevented stress-induced pain sensitivity. These findings demonstrate that microglia are critical mediators in the induction of pain conditions by stress. Moreover, these studies provide a proof of principle that microglia can be targeted as a therapeutic strategy to mitigate adverse pain conditions.


Asunto(s)
Dolor Crónico/fisiopatología , Dolor Crónico/psicología , Inflamación/psicología , Microglía , Medio Social , Enfermedades de la Médula Espinal/psicología , Estrés Psicológico/psicología , Animales , Ansiedad/psicología , Conducta Animal , Antígeno CD11b/biosíntesis , Antígeno CD11b/genética , Dolor Crónico/genética , Regulación de la Expresión Génica/genética , Hiperalgesia/fisiopatología , Hiperalgesia/psicología , Inflamación/genética , Inflamación/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Compuestos Orgánicos/farmacología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Médula Espinal , Enfermedades de la Médula Espinal/genética , Enfermedades de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal , Estrés Psicológico/genética
2.
Brain Behav Immun ; 69: 113-123, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29126979

RESUMEN

OBJECTIVE: Mounting evidence indicates that stress influences the experience of pain. Exposure to psychosocial stress disrupts bi-directional communication pathways between the central nervous system and peripheral immune system, and can exacerbate the frequency and severity of pain experienced by stressed subjects. Repeated social defeat (RSD) is a murine model of psychosocial stress that recapitulates the immune and behavioral responses to stress observed in humans, including activation of stress-reactive neurocircuitry and increased pro-inflammatory cytokine production. It is unclear, however, how these stress-induced neuroimmune responses contribute to increased pain sensitivity in mice exposed to RSD. Here we used a technique of regional analgesia with local anesthetics in mice to block the development of mechanical allodynia during RSD. We next investigated the degree to which pain blockade altered stress-induced neuroimmune activation and depressive-like behavior. METHODS: Following development of a mouse model of regional analgesia with discrete sensory blockade over the dorsal-caudal aspect of the spine, C57BL/6 mice were divided into experimental groups and treated with Ropivacaine (0.08%), Liposomal Bupivacaine (0.08%), or Vehicle (0.9% NaCl) prior to exposure to stress. This specific region was selected for analgesia because it is the most frequent location for aggression-associated pain due to biting during RSD. Mechanical allodynia was assessed 12 h after the first, third, and sixth day of RSD after resolution of the sensory blockade. In a separate experiment, social avoidance behavior was determined after the sixth day of RSD. Blood, bone marrow, brain, and spinal cord were collected for immunological analyses after the last day of RSD in both experiments following behavioral assessments. RESULTS: RSD increased mechanical allodynia in an exposure-dependent manner that persisted for at least one week following cessation of the stressor. Mice treated with either Ropivacaine or Liposomal Bupivacaine did not develop mechanical allodynia following exposure to stress, but did develop social avoidance behavior. Neither drug affected stress-induced activation of monocytes in the bone marrow, blood, or brain. Neuroinflammatory responses developed in all treatment groups, as evidenced by elevated IL-1ß mRNA levels in the brain and spinal cord after RSD. CONCLUSIONS: In this study, psychosocial stress was associated with increased pain sensitivity in mice. Development of mechanical allodynia with RSD was blocked by regional analgesia with local anesthetics, Ropivacaine or Liposomal Bupivacaine. Despite blocking mechanical allodynia, these anesthetic interventions did not prevent neuroimmune activation or social avoidance associated with RSD. These data suggest that stress-induced neuroinflammatory changes are not associated with increased sensitivity to pain following RSD. Thus, blocking peripheral nociception was effective in inhibiting enhanced pain signaling without altering stress-induced immune or behavioral responses.


Asunto(s)
Anestésicos Locales/uso terapéutico , Bupivacaína/uso terapéutico , Umbral del Dolor/efectos de los fármacos , Dolor/prevención & control , Ropivacaína/uso terapéutico , Conducta Social , Estrés Psicológico/complicaciones , Anestésicos Locales/farmacología , Animales , Conducta Animal/fisiología , Bupivacaína/farmacología , Modelos Animales de Enfermedad , Ratones , Dolor/etiología , Dolor/inmunología , Dimensión del Dolor , Ropivacaína/farmacología , Estrés Psicológico/inmunología
3.
J Neurosci ; 36(30): 7946-56, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27466339

RESUMEN

UNLABELLED: Amplified neuroinflammatory responses following an immune challenge occur with normal aging and can elicit or exacerbate neuropathology. The mechanisms mediating this sensitized or "primed" immune response in the aged brain are not fully understood. The alarmin high mobility group box 1 (HMGB1) can be released under chronic pathological conditions and initiate inflammatory cascades. This led us to investigate whether HMGB1 regulates age-related priming of the neuroinflammatory response. Here, we show that HMGB1 protein and mRNA were elevated in the hippocampus of unmanipulated aged rats (24-month-old F344XBN rats). Furthermore, aged rats had increased HMGB1 in the CSF, suggesting increased HMGB1 release. We demonstrate that blocking HMGB1 signaling with an intracisterna magna (ICM) injection of the competitive antagonist to HMGB1, Box-A, downregulates basal expression of several inflammatory pathway genes in the hippocampus of aged rats. This indicates that blocking the actions of HMGB1 might reduce age-associated inflammatory priming. To test this hypothesis, we evaluated whether HMGB1 antagonism blocks the protracted neuroinflammatory and sickness response to peripheral Escherichia coli (E. coli) infection in aged rats. ICM pretreatment of aged rats with Box-A 24 h before E. coli infection prevented the extended hippocampal cytokine response and associated cognitive and affective behavioral changes. ICM pretreatment with Box-A also inhibited aging-induced potentiation of the microglial proinflammatory response to lipopolysaccharide ex vivo Together, these results suggest that HMGB1 mediates neuroinflammatory priming in the aged brain. Blocking the actions of HMGB1 appears to "desensitize" aged microglia to an immune challenge, thereby preventing exaggerated behavioral and neuroinflammatory responses following infection. SIGNIFICANCE STATEMENT: The world's population is aging, highlighting a need to develop treatments that promote quality of life in aged individuals. Normal aging is associated with precipitous drops in cognition, typically following events that induce peripheral inflammation (e.g., infection, surgery, heart attack). Peripheral immune stimuli cause exaggerated immune responses in the aged brain, which likely underlie these behavioral deficits. Here, we investigated whether the alarmin high mobility group box 1 (HMGB1) mediates age-associated "priming" of the neuroinflammatory response. HMGB1 is elevated in aged rodent brain and CSF. Blocking HMGB1 signaling downregulated expression of inflammatory pathway genes in aged rat brain. Further, HMGB1 antagonism prevented prolonged infection-induced neuroinflammatory and sickness responses in aged rats. Overall, blocking HMGB1 "desensitized" microglia in the aged brain, thereby preventing pathological infection-elicited neuroinflammatory responses.


Asunto(s)
Envejecimiento/inmunología , Encefalitis/inmunología , Proteína HMGB1/inmunología , Hipocampo/inmunología , Mediadores de Inflamación/inmunología , Alarminas/inmunología , Animales , Masculino , Ratas , Distribución Tisular
4.
Chemistry ; 23(64): 16328-16337, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-28872717

RESUMEN

We report on the benefits of changing the bridging group X of bis-pyridyl ligands, that is, Py-X-Py where X is NH, CH2 , C(CH3 )2 , or PPh, on the photo- and electroluminescent properties of a new family of luminescent cationic H-heterocyclic carbene (NHC) copper(I) complexes. A joint experimental and theoretical study demonstrates that the bridging group affects the molecular conformation from a planar-like structure (X is NH and CH2 ) to a boat-like structure (X is C(CH3 )2 and PPh), leading to i) four-fold enhancement of the photoluminescence quantum yield (ϕem ) without affecting the thermally activated delayed fluorescence mechanism, and ii) one order of magnitude reduction of the ionic conductivity (σ) of thin films. This leads to an overall enhancement of the device efficacy and luminance owing to the increased ϕem and the use of low applied driving currents.

5.
J Neurosci ; 35(1): 316-24, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25568124

RESUMEN

Exposure to acute and chronic stressors sensitizes the proinflammatory response of microglia to a subsequent immune challenge. However, the proximal signal by which stressors prime microglia remains unclear. Here, high mobility group box-1 (HMGB-1) protein was explored as a potential mediator of stress-induced microglial priming and whether HMGB-1 does so via the nucleotide-binding domain, leucine-rich repeat, pyrin domain containing protein 3 (NLRP3) inflammasome. Exposure to 100 inescapable tail shocks (ISs) increased HMGB-1 and NLRP3 protein in the hippocampus and led isolated microglia to release HMGB-1 ex vivo. To determine whether HMGB-1 signaling is necessary for stress-induced sensitization of microglia, the HMGB-1 antagonist BoxA was injected into the cisterna magna before IS. Hippocampal microglia were isolated 24 h later and stimulated with LPS ex vivo to probe for stress-induced sensitization of proinflammatory responses. Previous IS potentiated gene expression of NLRP3 and proinflammatory cytokines to LPS, that is, microglia were sensitized. Treatment with BoxA abolished this effect. To determine whether HMGB-1 is sufficient to prime microglia, IS was replaced with intracerebral administration of disulfide or fully reduced HMGB-1. Intracerebral disulfide HMGB-1 mimicked the effect of the stressor, because microglia isolated from HMGB-1-treated rats expressed exaggerated NLRP3 and proinflammatory cytokine expression after LPS treatment, whereas fully reduced HMGB-1 had no effect. The present results suggest that the CNS innate immune system can respond to an acute stressor as if it were cellular damage, thereby releasing the danger signal HMGB-1 in the brain to prime microglia by acting on the NLRP3 inflammasome, in preparation for a later immune challenge.


Asunto(s)
Proteína HMGB1/biosíntesis , Hipocampo/metabolismo , Inflamasomas/biosíntesis , Microglía/metabolismo , Receptores Citoplasmáticos y Nucleares/biosíntesis , Estrés Psicológico/metabolismo , Animales , Proteínas Portadoras , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/psicología
6.
Brain Behav Immun ; 55: 215-224, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26482581

RESUMEN

The alarmin high mobility group box-1 (HMGB1) has been implicated as a key factor mediating neuroinflammatory processes. Recent findings suggest that the redox state of HMGB1 is a critical molecular feature of HMGB1 such that the reduced form (fr-HMGB1) is chemotactic, while the disulfide form (ds-HMGB1) is pro-inflammatory. The present study examined the neuroinflammatory effects of these molecular forms as well as the ability of these forms to prime the neuroinflammatory and microglial response to an immune challenge. To examine the neuroinflammatory effects of these molecular forms in vivo, animals were administered intra-cisterna magna (ICM) a single dose of fr-HMGB1 (10µg), ds-HMGB1 (10µg) or vehicle and basal pro-inflammatory effects were measured 2 and 24h post-injection in hippocampus. Results of this initial experiment demonstrated that ds-HMGB1 increased hippocampal pro-inflammatory mediators at 2h (NF-κBIα mRNA, NLRP3 mRNA and IL-1ß protein) and 24h (NF-κBIα mRNA, TNFα mRNA, and NLRP3 protein) after injection. fr-HMGB1 had no effect on these mediators. These neuroinflammatory effects of ds-HMGB1 suggested that ds-HMGB1 may function to prime the neuroinflammatory response to a subsequent immune challenge. To assess the neuroinflammatory priming effects of these molecular forms, animals were administered ICM a single dose of fr-HMGB1 (10µg), ds-HMGB1 (10µg) or vehicle and 24h after injection, animals were challenged with LPS (10µg/kg IP) or vehicle. Neuroinflammatory mediators and the sickness response (3, 8 and 24h after injection) were measured 2h after immune challenge. We found that ds-HMGB1 potentiated the neuroinflammatory (NF-κBIα mRNA, TNFα mRNA, IL-1ß mRNA, IL-6 mRNA, NLRP3 mRNA and IL-1ß protein) and sickness response (reduced social exploration) to LPS challenge. fr-HMGB1 failed to potentiate the neuroinflammatory response to LPS. To examine whether these molecular forms of HMGB1 directly induce neuroinflammatory effects in isolated microglia, whole brain microglia were isolated and treated with fr-HMGB1 (0, 1, 10, 100, or 1000ng/ml) or ds-HMGB1 (0, 1, 10, 100, or 1000ng/ml) for 4h and pro-inflammatory mediators measured. To assess the effects of these molecular forms on microglia priming, whole brain microglia were pre-exposed to these forms of HMGB1 (0, 1, 10, 100, or 1000ng/ml) and subsequently challenged with LPS (10ng/ml). We found that ds-HMGB1 increased expression of NF-κBIα mRNA and NLRP3 mRNA in isolated microglia, and potentiated the microglial pro-inflammatory response (TNFα mRNA, IL-1ß mRNA and IL-1ß protein) to LPS. fr-HMGB1 failed to potentiate the microglial pro-inflammatory response to LPS. Consistent with prior reports, the present findings demonstrate that the disulfide form of HMGB1 not only potentiates the neuroinflammatory response to a subsequent immune challenge in vivo, but also potentiates the sickness response to that challenge. Moreover, the present findings demonstrate for the first time that ds-HMGB1 directly potentiates the microglia pro-inflammatory response to an immune challenge, a finding that parallels the effects of ds-HMGB1 in vivo. In addition, ds-HMGB1 induced expression of NLRP3 and NF-κBIα in vivo and in vitro suggesting that the NLRP3 inflammasome may play role in the priming effects of ds-HMGB1. Taken together, the present results suggest that the redox state of HMGB1 is a critical determinant of the priming properties of HMGB1 such that the disulfide form of HMGB1 induces a primed immunophenotype in the CNS, which may result in an exacerbated neuroinflammatory response upon exposure to a subsequent pro-inflammatory stimulus.


Asunto(s)
Alarminas/inmunología , Proteína HMGB1/farmacología , Hipocampo/inmunología , Inflamasomas/inmunología , Mediadores de Inflamación/inmunología , Inflamación/inmunología , Microglía/inmunología , Animales , Proteína HMGB1/administración & dosificación , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley
7.
Brain Behav Immun ; 51: 99-108, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26254235

RESUMEN

Methamphetamine (METH) induces neuroinflammatory effects, which may contribute to the neurotoxicity of METH. However, the mechanism by which METH induces neuroinflammation has yet to be clarified. A considerable body of evidence suggests that METH induces cellular damage and distress, particularly in dopaminergic neurons. Damaged neurons release danger-associated molecular patterns (DAMPs) such as high mobility group box-1 (HMGB1), which induces pro-inflammatory effects. Therefore, we explored the notion here that METH induces neuroinflammation indirectly through the release of HMGB1 from damaged neurons. Adult male Sprague-Dawley rats were injected IP with METH (10mg/kg) or vehicle (0.9% saline). Neuroinflammatory effects of METH were measured in nucleus accumbens (NAcc), ventral tegmental area (VTA) and prefrontal cortex (PFC) at 2h, 4h and 6h after injection. To assess whether METH directly induces pro-inflammatory effects in microglia, whole brain or striatal microglia were isolated using a Percoll density gradient and exposed to METH (0, 0.1, 1, 10, 100, or 1000µM) for 24h and pro-inflammatory cytokines measured. The effect of METH on HMGB1 and IL-1ß in striatal tissue was then measured. To determine the role of HMGB1 in the neuroinflammatory effects of METH, animals were injected intra-cisterna magna with the HMGB1 antagonist box A (10µg) or vehicle (sterile water). 24h post-injection, animals were injected IP with METH (10mg/kg) or vehicle (0.9% saline) and 4h later neuroinflammatory effects measured in NAcc, VTA, and PFC. METH induced robust pro-inflammatory effects in NAcc, VTA, and PFC as a function of time and pro-inflammatory analyte measured. In particular, METH induced profound effects on IL-1ß in NAcc (2h) and PFC (2h and 4h). Exposure of microglia to METH in vitro failed to induce a pro-inflammatory response, but rather induced significant cell death as well as a decrease in IL-1ß. METH treatment increased HMGB1 in parallel with IL-1ß in striatum. Pre-treatment with the HMGB1 antagonist box A blocked the neuroinflammatory effects (IL-1ß) of METH in NAcc, VTA and PFC. The present results suggest that HMGB1 mediates, in part, the neuroinflammatory effects of METH and thus may alert CNS innate immune cells to the toxic effects of METH.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encefalitis/metabolismo , Proteína HMGB1/metabolismo , Mediadores de Inflamación/metabolismo , Metanfetamina/administración & dosificación , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Encefalitis/inducido químicamente , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
8.
Brain Behav Immun ; 48: 1-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25816800

RESUMEN

High mobility group box-1 (HMGB1) is an endogenous danger signal or alarmin that mediates activation of the innate immune response including chemotaxis and pro-inflammatory cytokine release. HMGB1 has been implicated in the pathophysiology of several neuroinflammatory conditions including ischemia, traumatic brain injury, seizure and chronic ethanol use. In the present review, the unique structural and functional properties of HMGB1 will be explored including its affinity for multiple pattern recognition receptors (TLR2/TLR4), redox sensitivity and adjuvant-like properties. In light of recent evidence suggesting that HMGB1 may also mediate stress-induced sensitization of neuroinflammatory responses, mechanisms of HMGB1 action in neuroinflammatory priming are explored. A model of neuroinflammatory priming is developed wherein glucocorticoids induce synthesis and release of HMGB1 from microglia, which signals through TLR2/TLR4, thereby priming the NLRP3 inflammasome. We propose that if GCs reach a critical threshold as during a fight/flight response, they may thus function as an alarmin by inducing HMGB1, thereby preparing an organism's innate immune system (NLRP3 inflammasome priming) for subsequent immune challenges such as injury, trauma or infection, which are more likely to occur during a fight/flight response. In doing so, GCs may confer a significant survival advantage by enhancing the central innate immune and sickness response to immune challenges.


Asunto(s)
Alarminas/metabolismo , Encéfalo/metabolismo , Proteína HMGB1/metabolismo , Inflamasomas/metabolismo , Inflamación/metabolismo , Estrés Fisiológico/inmunología , Animales , Encéfalo/inmunología , Inmunidad Innata/fisiología , Inflamación/inmunología
9.
Brain Behav Immun ; 42: 22-32, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24998196

RESUMEN

High-fat diet (HFD)-induced obesity is reaching worldwide proportions. In addition to causing obesity, HFDs also induce a variety of health disorders, which includes cognitive decline. Hippocampal function may be particularly vulnerable to the negative consequences of HFD, and it is suspected that 'primed' neuroinflammatory processes may mediate this response. To examine the link between diet, hippocampal function and neuroinflammation, male Wistar rats were fed a medium or HFD. Hippocampal memory function was measured using contextual pre-exposure fear conditioning (CPE-FC). Rats fed a HFD demonstrated impaired memory, an effect that was augmented with longer duration of HFD consumption. HFD-induced memory impairments were linked to potentiated levels of interleukin-1 beta (IL-1ß) protein in the hippocampus 2h after the foot-shock that occurs during CPE-FC. Central IL-1 receptor antagonism, with intracisterna magna (ICM) administration of hIL-1RA prior to the foot-shock prevented the diet-induced memory disruption, suggesting a critical role for IL-1ß in this phenomenon. Additionally, obese animals whose diet regimen was reversed from HFD back to standard chow recovered memory function and did not demonstrate a foot-shock-induced hippocampal IL-1ß increase. Interestingly, dietary reversal neutralized the negative impact of HFD on memory and IL-1ß, yet animals maintained physiological evidence of obesity (increased body mass and serum leptin), indicating that dietary components, not body mass, may mediate the negative effects on memory.


Asunto(s)
Condicionamiento Psicológico/fisiología , Dieta Alta en Grasa , Hipocampo/metabolismo , Interleucina-1beta/metabolismo , Trastornos de la Memoria/etiología , Memoria/fisiología , Receptores de Interleucina-1/antagonistas & inhibidores , Animales , Condicionamiento Psicológico/efectos de los fármacos , Miedo , Hipocampo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria/metabolismo , Ratas , Ratas Wistar , Receptores de Interleucina-1/metabolismo
10.
Brain Behav Immun ; 32: 112-21, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23500798

RESUMEN

Acute and chronic stressors sensitize or prime the neuroinflammatory response to a subsequent peripheral or central immunologic challenge. However, the neuroimmune process(es) by which stressors prime or sensitize subsequent neuroinflammatory responses remains unclear. Prior evidence suggested that toll-like receptors (TLRs) might be involved in the mediation of primed neuroinflammatory responses, but the role of TLRs during a stressor has never been directly tested. Here, a novel TLR2 and TLR4 antagonist, OxPAPC, was used to probe the contribution of TLRs in the stress sensitization phenomenon. OxPAPC has not previously been administered to the brain, and so its action in blocking TLR2 and TLR4 action in brain was first verified. Administration of OxPAPC into the CNS prior to stress prevented the stress-induced potentiation of hippocampal pro-inflammatory response to a subsequent peripheral LPS challenge occurring 24 h later. In addition, in vivo administration of OxPAPC prior to stress prevented the sensitized pro-inflammatory response from isolated microglia following administration of LPS ex vivo, further implicating microglia as a key neuroimmune substrate that mediates stress-induced sensitized neuroinflammation.


Asunto(s)
Inflamación/patología , Estrés Psicológico/inmunología , Estrés Psicológico/patología , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Cisterna Magna , Citocinas/biosíntesis , Citocinas/genética , Electrochoque , Hipocampo/patología , Inmunidad Innata , Inyecciones , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Lipopolisacáridos/farmacología , Masculino , Neuroinmunomodulación/efectos de los fármacos , Fosfatidilcolinas/farmacología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
J Neuroimmunol ; 337: 577073, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31670063

RESUMEN

Traumatic injury in children is known to cause immune suppression. Polytrauma involving a traumatic brain injury (TBI) may increase this degree of immune suppression, which increases the risk of developing nosocomial infections, potentially causing secondary brain injury and worsening patient outcomes. Despite the high prevalence of polytrauma with TBI in children, mechanisms of immune suppression following such injuries remain poorly understood. Here, we used a combined animal injury model of TBI and hemorrhage to assess immune function after polytrauma. Pre-pubescent rats were injured using a prefrontal controlled cortical impact method and a controlled hemorrhage by femoral arteriotomy. Immune function was measured by whole blood ex-vivo tumor necrosis factor alpha production capacity following incubation with lipopolysaccharide, measuring the percentage of monocytes by flow cytometry, and by examining concentrations of plasma cytokines. The degree of brain injury was sufficient to produce deficits in spatial memory testing (Barnes maze). Both hemorrhage and TBI with hemorrhage (combined injury) reduced several of the measured plasma cytokines, as compared with TBI alone. The combined injury correlated with reduced concentration of monocytes and reduced tumor necrosis factor alpha production capacity at post-injury day 1. These results demonstrate that this animal model can be used to study post-injury immune suppression.


Asunto(s)
Lesiones Traumáticas del Encéfalo/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Tolerancia Inmunológica/fisiología , Inmunidad Innata/fisiología , Traumatismo Múltiple/inmunología , Factores de Edad , Animales , Lesiones Traumáticas del Encéfalo/sangre , Lesiones Traumáticas del Encéfalo/complicaciones , Citocinas/sangre , Traumatismo Múltiple/sangre , Traumatismo Múltiple/complicaciones , Ratas , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/inmunología
12.
Biol Psychiatry ; 85(8): 667-678, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30527629

RESUMEN

BACKGROUND: Stress is associated with an increased prevalence of anxiety and depression. Repeated social defeat (RSD) stress in mice increases the release of monocytes from the bone marrow that are recruited to the brain by microglia. These monocytes enhance inflammatory signaling and augment anxiety. Moreover, RSD promotes stress sensitization, in which exposure to acute stress 24 days after cessation of RSD causes anxiety recurrence. The purpose of this study was to determine whether microglia were critical to stress sensitization and exhibited increased reactivity to subsequent acute stress or immune challenge. METHODS: Mice were exposed to RSD, microglia were eliminated by colony-stimulating factor 1 receptor antagonism (PLX5622) and allowed to repopulate, and responses to acute stress or immune challenge (lipopolysaccharide) were determined 24 days after RSD sensitization. RESULTS: Microglia maintained a unique messenger RNA signature 24 days after RSD. Moreover, elimination of RSD-sensitized microglia prevented monocyte accumulation in the brain and blocked anxiety recurrence following acute stress (24 days). When microglia were eliminated prior to RSD and repopulated and mice were subjected to acute stress, there was monocyte accumulation in the brain and anxiety in RSD-sensitized mice. These responses were unaffected by microglial elimination/repopulation. This may be related to neuronal sensitization that persisted 24 days after RSD. Following immune challenge, there was robust microglial reactivity in RSD-sensitized mice associated with prolonged sickness behavior. Here, microglial elimination/repopulation prevented the amplified immune reactivity ex vivo and in vivo in RSD-sensitized mice. CONCLUSIONS: Microglia and neurons remain sensitized weeks after RSD, and only the immune reactivity component of RSD-sensitized microglia was prevented by elimination/repopulation.


Asunto(s)
Microglía/inmunología , Microglía/patología , Conducta Social , Estrés Psicológico/inmunología , Estrés Psicológico/psicología , Animales , Encéfalo/metabolismo , Receptor 1 de Quimiocinas CX3C/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Conducta de Enfermedad , Lipopolisacáridos , Masculino , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/metabolismo , Monocitos/inmunología , Compuestos Orgánicos/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Estrés Psicológico/metabolismo
13.
Antivir Ther ; 23(6): 495-504, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29714167

RESUMEN

BACKGROUND: Raltegravir (RTG) and dolutegravir (DTG) have different pharmacokinetic patterns in the gastrointestinal tract. To determine if this results in pharmacodynamic differences, we compared HIV RNA, HIV DNA and immunological markers in gut-associated lymphoid tissue (GALT) of HIV-infected participants receiving RTG or DTG with tenofovir+emtricitabine (TDF/FTC). METHODS: GALT specimens from the terminal ileum, splenic flexure and rectum were obtained by colonoscopy at a single time point in 20 adults treated with RTG (n=10) or DTG (n=10) with HIV RNA <50 copies/ml. Flow cytometry, drug concentrations, and HIV RNA and DNA were analysed in tissue. CD4/8+ T-cells were tested for γδ TCR, and markers of T-cell activation and exhaustion. Data are reported as median (Q1-Q3). RESULTS: A total of 15 men and 5 women were enrolled. There was no difference in time since HIV diagnosis for those on RTG (9.5 [4-22] years) and DTG (17 [1-24] years; P=0.6), although time on RTG (5.4 [2.3-6.7] years) was greater than DTG (1.0 [0.1-1.5] years; P<0.001). Concentrations of RTG and DTG in rectal tissue were similar to previous reports: median tissue:plasma ratio was 11.25 for RTG and 0.44 for DTG. RNA:DNA ratios were 1.14 (0.18-5.10) for the RTG group and 0.90 (0.30-18.87) for the DTG group (P=0.95). No differences (P≥0.1) between CD4+ and CD8+ T-cell markers were found. CONCLUSIONS: RTG produced higher tissue exposures than DTG, but no significant differences in GALT HIV RNA, DNA or most immunological markers were observed. ClinicalTrials.gov NCT02218320.


Asunto(s)
Fármacos Anti-VIH/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Tejido Linfoide/efectos de los fármacos , Raltegravir Potásico/uso terapéutico , Adulto , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Colon Transverso/efectos de los fármacos , Colon Transverso/patología , Colon Transverso/virología , ADN Viral/antagonistas & inhibidores , ADN Viral/genética , ADN Viral/metabolismo , Emtricitabina/uso terapéutico , Femenino , Expresión Génica , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/inmunología , Humanos , Íleon/efectos de los fármacos , Íleon/patología , Íleon/virología , Inmunidad Innata/efectos de los fármacos , Tejido Linfoide/patología , Tejido Linfoide/virología , Masculino , Persona de Mediana Edad , Oxazinas , Piperazinas , Piridonas , ARN Viral/antagonistas & inhibidores , ARN Viral/genética , ARN Viral/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Recto/efectos de los fármacos , Recto/patología , Recto/virología , Tenofovir/uso terapéutico , Resultado del Tratamiento
14.
Neuropsychopharmacology ; 42(1): 46-61, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27319971

RESUMEN

Mounting evidence indicates that proinflammatory signaling in the brain affects mood, cognition, and behavior and is linked with the etiology of psychiatric disorders, including anxiety and depression. The purpose of this review is to focus on stress-induced bidirectional communication pathways between the central nervous system (CNS) and peripheral immune system that converge to promote a heightened neuroinflammatory environment. These communication pathways involve sympathetic outflow from the brain to the peripheral immune system that biases hematopoietic stem cells to differentiate into a glucocorticoid-resistant and primed myeloid lineage immune cell. In conjunction, microglia-dependent neuroinflammatory events promote myeloid cell trafficking to the brain that reinforces stress-related behavior, and is argued to play a role in stress-related psychiatric disorders. We will discuss evidence implicating a key role for endothelial cells that comprise the blood-brain barrier in propagating peripheral-to-central immune communication. We will also discuss novel neuron-to-glia communication pathways involving endogenous danger signals that have recently been argued to facilitate neuroinflammation under various conditions, including stress. These findings help elucidate the complex communication that occurs in response to stress and highlight novel therapeutic targets against the development of stress-related psychiatric disorders.


Asunto(s)
Conducta/fisiología , Sistema Nervioso Central/inmunología , Sistema Inmunológico/inmunología , Inflamación/inmunología , Trastornos Mentales/inmunología , Monocitos/inmunología , Transducción de Señal/inmunología , Estrés Psicológico/inmunología , Animales , Humanos , Trastornos Mentales/etiología , Estrés Psicológico/complicaciones
15.
Dalton Trans ; 46(19): 6312-6323, 2017 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-28452386

RESUMEN

This work studies the effect of the σ-Hammett parameter (σp) - i.e., the σ-donation effect caused by substitution at the para position of a bipyridine ligand (4,4'-R2bipy, where R is MeO, Me, H, NO2) - on both the photo- and electro-luminescence features of a series of heteroleptic copper(i) complexes - i.e., [Cu(N^N)(P^P)]+ where N^N and P^P ligands are R2bipy and Xantphos, respectively. By virtue of a comprehensive photophysical, theoretical, and thin-film lighting device - i.e., light-emitting electrochemical cells (LECs) - investigation, we note a clear relationship between the σp and the photo- and electro-luminescence parameters, such as photoluminescence quantum yields, excited-state lifetimes, and emission maxima, as well as device brightness, stability, and efficacy, respectively. As the most relevant finding, the substitution with the group featuring the most negative σp - i.e., MeO - provides a ca. five-fold enhancement of all of the aforementioned figures-of-merit upon comparison within the series of complexes. As such, this work provides a new guideline for a device optimization through a rational ligand design for heteroleptic copper(i) complexes.

16.
Neurobiol Stress ; 4: 62-70, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27981190

RESUMEN

Stress and glucocorticoids (GCs) have universally been considered to be anti-inflammatory, however in recent years, stress and GCs have been found to exert permissive effects (immunological priming) on neuroinflammatory processes. This phenomenon of priming is characterized by prior stress or GC exposure potentiating the neuroinflammatory response to a subsequent immune challenge. A considerable body of evidence is discussed here that supports this permissive effect of stress and GCs. In light of this evidence, a mechanism of neuroinflammatory priming is proposed involving a signal cascade in the brain involving danger-associated molecular patterns (HMGB-1) and inflammasomes (NLRP3), which results in an exaggerated or amplified neuroinflammatory response and subsequently, the amplification of the physiological and behavioral sequelae of this response (i.e. sickness). Finally, we explore the notion that stressor-induced sensitization of the neuroimmune microenvironment may predispose individuals to psychiatric disorders, in which exaggerated innate immune/inflammatory responses in the brain are now thought to play a key role.

17.
Dalton Trans ; 45(21): 8984-93, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27160065

RESUMEN

This work provides the synthesis, structural characterization, electrochemical and photophysical features, as well as the application in light-emitting electrochemical cells (LECs) of a novel heteroleptic copper(i) complex - [Cu(impy)(POP)][PF6], where impy is 3-(2-methoxyphenyl)-1-(pyridine-2-yl)imidazo[1,5-a]pyridine and POP is bis{2-(diphenylphosphanyl)phenyl}ether. This compound shows blue photoluminescence (PL, λ = 450 nm) in solution and solid-state and excellent redox stability. Despite these excellent features, the electroluminescence (EL) response is located at ∼550 nm. Although the EL spectrum of LECs is typically red-shifted compared to the PL of the electroluminescent material, a shift of ca. 100 nm represents the largest one reported in LECs. To date, the large shift phenomena have been attributed to (i) a change in the nature of the lowest emitting state due to a concentration effect of the films, (ii) a reversible substitution of the ligands due to the weak coordination to the Cu(i), and (iii) a change in the distribution of the excited states due to polarization effects. After having discarded these along with others like the irreversible degradation of the emitter during device fabrication and/or under operation conditions, driving conditions, active layer composition, and changes in the excited states under different external electrical stimuli, we attribute the origin of this unexpected shift to a lack of a thermally activated delayed fluorescence (TADF) process due to the solely ligand-centered character of the excited states. As such, the lack of a charge transfer character in the excited states leads to a blue-fluorescence and yellow-phosphorescence photo- and electro-responses, respectively. This corroborates recent studies focused on the design of TADF for heteroleptic copper(i) complexes. Overall, this work is a clear insight into the design of new copper(i) complexes towards the preparation of blue LECs, which are still unexplored.

18.
Dalton Trans ; 45(34): 13284-8, 2016 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-27363542

RESUMEN

The syntheses, photophysical/electrochemical characterizations of different metallated porphyrins -i.e., Zn(2+), Pt(2+), Pd(2+), and Sn(4+) porphyrins - as well as their first application in light-emitting electrochemical cells are provided. A direct comparison demonstrates that depending on the metallation either efficient (Pt-por) or stable (Zn-por) devices are achieved, demonstrating that the choice of the metal core is a key aspect for future developments.

19.
Dalton Trans ; 45(8): 3251-5, 2016 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-26827753

RESUMEN

We report two unprecedent alkynyl bridging cyclometalated clusters [Ir2M2(ppy)4(µ-C[triple bond, length as m-dash]CC6H4-OMe3)4] where M is Ag (2) and Cu (3), which display distinctive luminescence properties. While 2 features a green phosphorescence/electroluminescence nature located at the ppy ligands ((3)LC), 3 shows an orange emission confined to the metals and alkynyl groups having a mixed (3)L'C/(3)L'MCT/(3)MMCT (L' = alkynyl) nature.

20.
Psychoneuroendocrinology ; 66: 82-90, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26799851

RESUMEN

Circadian rhythms are endogenous cycles of physiology and behavior that align with the daily rotation of the planet and resulting light-dark cycle. The circadian system ensures homeostatic balance and regulates many aspects of physiology, including the stress response and susceptibility to and/or severity of stress-related sequelae. Both acute and chronic stressors amplify neuroinflammatory responses to a subsequent immune challenge, however it is not known whether circadian timing of the stressor regulates the priming response. Here, we test whether stress-induced neuroinflammatory priming is regulated by the circadian system. As has been previously shown, exposure to 100 inescapable tails shocks (IS) increased hippocampal cytokines following a subsequent inflammatory challenge. However, this effect was limited to animals that experienced the stressor during the light phase. Rats exposed to stress during the dark phase did not alter inflammatory potential following lipopolysaccharide (LPS) challenge. To determine whether microglia might be involved in diurnal differences in neuroinflammatory priming, microglia were isolated 24h after stress that occurred either during the middle of the light or dark phase. Only microglia isolated from animals stressed during the light phase demonstrated an exaggerated inflammatory response when treated ex vivo with LPS. To determine possible circadian dependency of microglia responsiveness to glucocorticoids - the likely proximal mediator for stress associated neuroinflammatory priming - microglia were isolated during the middle of the light or dark phase and treated ex vivo with corticosterone. Glucocorticoids treatment downregulated CX3CR1 and CD200R, two genes involved in microglial inflammatory "off" signaling; however, there was no effect of time of day on expression of either gene. Importantly, while absolute concentrations of corticosterone were comparable following IS during the light and dark phase, the magnitude of change in corticosterone was greater during the light phase. This work highlights the importance of studying circadian rhythms to elucidate biological mechanisms of stress.


Asunto(s)
Ritmo Circadiano/fisiología , Inflamación/etiología , Inflamación Neurogénica/etiología , Estrés Psicológico/complicaciones , Animales , Citocinas/metabolismo , Glucocorticoides/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Hipocampo/metabolismo , Lipopolisacáridos/farmacología , Masculino , Microglía/efectos de los fármacos , Microglía/inmunología , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/inmunología , Estrés Psicológico/fisiopatología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA