Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 118: 368-379, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38471576

RESUMO

Microglia play a central role in the etiology of many neuropathologies. Transgenic tools are a powerful experiment approach to gain reliable and specific control over microglia function. Adeno-associated virus (AAVs) vectors are already an indispensable tool in neuroscience research. Despite ubiquitous use of AAVs and substantial interest in the role of microglia in the study of central nervous system (CNS) function and disease, transduction of microglia using AAVs is seldom reported. This review explores the challenges and advancements made in using AAVs for expressing transgenes in microglia. First, we will examine the functional anatomy of the AAV capsid, which will serve as a basis for subsequent discussions of studies exploring the relationship between capsid mutations and microglia transduction efficacy. After outlining the functional anatomy of AAVs, we will consider the experimental evidence demonstrating AAV-mediated transduction of microglia and microglia-like cell lines followed by an examination of the most promising experimental approaches identified in the literature. Finally, technical limitations will be considered in future applications of AAV experimental approaches.


Assuntos
Dependovirus , Microglia , Animais , Dependovirus/genética , Transdução Genética , Microglia/metabolismo , Animais Geneticamente Modificados , Transgenes , Vetores Genéticos
2.
Int J Mol Sci ; 24(6)2023 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-36982250

RESUMO

Inflammatory conditions, including allergic asthma and conditions in which chronic low-grade inflammation is a risk factor, such as stress-related psychiatric disorders, are prevalent and are a significant cause of disability worldwide. Novel approaches for the prevention and treatment of these disorders are needed. One approach is the use of immunoregulatory microorganisms, such as Mycobacterium vaccae NCTC 11659, which have anti-inflammatory, immunoregulatory, and stress-resilience properties. However, little is known about how M. vaccae NCTC 11659 affects specific immune cell targets, including monocytes, which can traffic to peripheral organs and the central nervous system and differentiate into monocyte-derived macrophages that, in turn, can drive inflammation and neuroinflammation. In this study, we investigated the effects of M. vaccae NCTC 11659 and subsequent lipopolysaccharide (LPS) challenge on gene expression in human monocyte-derived macrophages. THP-1 monocytes were differentiated into macrophages, exposed to M. vaccae NCTC 11659 (0, 10, 30, 100, 300 µg/mL), then, 24 h later, challenged with LPS (0, 0.5, 2.5, 250 ng/mL), and assessed for gene expression 24 h following challenge with LPS. Exposure to M. vaccae NCTC 11659 prior to challenge with higher concentrations of LPS (250 ng/mL) polarized human monocyte-derived macrophages with decreased IL12A, IL12B, and IL23A expression relative to IL10 and TGFB1 mRNA expression. These data identify human monocyte-derived macrophages as a direct target of M. vaccae NCTC 11659 and support the development of M. vaccae NCTC 11659 as a potential intervention to prevent stress-induced inflammation and neuroinflammation implicated in the etiology and pathophysiology of inflammatory conditions and stress-related psychiatric disorders.


Assuntos
Lipopolissacarídeos , Mycobacterium , Humanos , Lipopolissacarídeos/farmacologia , Doenças Neuroinflamatórias , Inflamação , Macrófagos
3.
Int J Mol Sci ; 25(1)2023 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-38203645

RESUMO

Previous studies have shown that the in vivo administration of soil-derived bacteria with anti-inflammatory and immunoregulatory properties, such as Mycobacterium vaccae NCTC 11659, can prevent a stress-induced shift toward an inflammatory M1 microglial immunophenotype and microglial priming in the central nervous system (CNS). It remains unclear whether M. vaccae NCTC 11659 can act directly on microglia to mediate these effects. This study was designed to determine the effects of M. vaccae NCTC 11659 on the polarization of naïve BV-2 cells, a murine microglial cell line, and BV-2 cells subsequently challenged with lipopolysaccharide (LPS). Briefly, murine BV-2 cells were exposed to 100 µg/mL whole-cell, heat-killed M. vaccae NCTC 11659 or sterile borate-buffered saline (BBS) vehicle, followed, 24 h later, by exposure to 0.250 µg/mL LPS (Escherichia coli 0111: B4; n = 3) in cell culture media vehicle (CMV) or a CMV control condition. Twenty-four hours after the LPS or CMV challenge, cells were harvested to isolate total RNA. An analysis using the NanoString platform revealed that, by itself, M. vaccae NCTC 11659 had an "adjuvant-like" effect, while exposure to LPS increased the expression of mRNAs encoding proinflammatory cytokines, chemokine ligands, the C3 component of complement, and components of inflammasome signaling such as Nlrp3. Among LPS-challenged cells, M. vaccae NCTC 11659 had limited effects on differential gene expression using a threshold of 1.5-fold change. A subset of genes was assessed using real-time reverse transcription polymerase chain reaction (real-time RT-PCR), including Arg1, Ccl2, Il1b, Il6, Nlrp3, and Tnf. Based on the analysis using real-time RT-PCR, M. vaccae NCTC 11659 by itself again induced "adjuvant-like" effects, increasing the expression of Il1b, Il6, and Tnf while decreasing the expression of Arg1. LPS by itself increased the expression of Ccl2, Il1b, Il6, Nlrp3, and Tnf while decreasing the expression of Arg1. Among LPS-challenged cells, M. vaccae NCTC 11659 enhanced LPS-induced increases in the expression of Nlrp3 and Tnf, consistent with microglial priming. In contrast, among LPS-challenged cells, although M. vaccae NCTC 11659 did not fully prevent the effects of LPS relative to vehicle-treated control conditions, it increased Arg1 mRNA expression, suggesting that M. vaccae NCTC 11659 induces an atypical microglial phenotype. Thus, M. vaccae NCTC 11659 acutely (within 48 h) induced immune-activating and microglial-priming effects when applied directly to murine BV-2 microglial cells, in contrast to its long-term anti-inflammatory and immunoregulatory effects observed on the CNS when whole-cell, heat-killed preparations of M. vaccae NCTC 11659 were given peripherally in vivo.


Assuntos
Infecções por Citomegalovirus , Microglia , Mycobacteriaceae , Animais , Camundongos , Lipopolissacarídeos/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Interleucina-6 , Adjuvantes Imunológicos , Adjuvantes Farmacêuticos , Anti-Inflamatórios
4.
Neurobiol Aging ; 71: 105-114, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30118926

RESUMO

Aging is a major risk factor for developing postoperative cognitive dysfunction. Neuroinflammatory processes, which can play a causal role in the etiology of postoperative cognitive dysfunction, are potentiated or primed as a function of aging. Here we explored whether exposure to a microorganism with immunoregulatory and anti-inflammatory properties, Mycobacterium vaccae NCTC 11659 (M. vaccae), could ameliorate age-associated neuroinflammatory priming. Aged (24 months) and adult (3 months) male F344XBN rats were immunized with heat-killed M. vaccae (3 injections, once per week) before undergoing a laparotomy or anesthesia control procedure. Aged, but not young rats, showed postoperative learning/memory deficits in a fear-conditioning paradigm. Importantly, M. vaccae immunization protected aged rats from these surgery-induced cognitive impairments. M. vaccae immunization also shifted the aged proinflammatory hippocampal microenvironment toward an anti-inflammatory phenotype. Furthermore, M. vaccae immunization reduced age-related hyperinflammatory responses in isolated hippocampal microglia. Overall, our novel data suggest that M. vaccae can induce an anti-inflammatory milieu in the aged brain and thus mitigate the neuroinflammatory and cognitive impairments induced by surgery.


Assuntos
Vacinas Bacterianas/administração & dosagem , Disfunção Cognitiva/imunologia , Disfunção Cognitiva/cirurgia , Encefalite/imunologia , Encefalite/cirurgia , Mycobacterium/imunologia , Animais , Disfunção Cognitiva/prevenção & controle , Encefalite/prevenção & controle , Hipocampo/imunologia , Imunização , Masculino , Memória , Microglia/imunologia , Ratos Endogâmicos F344 , Vacinas de Produtos Inativados/administração & dosagem
5.
Mol Cell Neurosci ; 89: 71-79, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29678518

RESUMO

Neuroinflammation is a common pathogenic mechanism for a number of neurodegenerative disorders including Alzheimer's and Parkinson's diseases. Microglia, the immune cells of the brain, contribute to the onset and progression of the neuroinflammation observed in these diseases. Microglia become activated and initiate an inflammatory response by interacting with a diverse set of molecules, including the group of endogenous proteins released upon cell damage, termed damage-associated molecular patterns (DAMPs). One of these molecules, mitochondrial transcription factor A (TFAM), has been shown to induce pro-inflammatory and cytotoxic responses of microglia in vitro. Here, we demonstrate that TFAM injected into the cisterna magna of male Sprague-Dawley rats upregulates (i) the expression of monocyte chemotactic protein (MCP)-1, interleukin (IL)-1ß, IL-6, tumor necrosis factor (TNF)-α and nuclear factor-kappa B inhibitor alpha (NF-κBIA) in the hippocampus; (ii) the expression of MCP-1, IL-1ß and TNF-α in the frontal cortex; and (iii) IL-1ß protein concentration in both these brain regions. These same inflammatory mediators are upregulated in isolated rat microglia following their in vitro exposure to extracellular TFAM. Blocking the receptor for advanced glycation endproducts (RAGE) and the macrophage antigen complex (Mac)-1 by specific antibodies inhibited the TFAM-induced secretion of MCP-1 by THP-1 monocytic cells, which were used to model human microglia. Our data support the hypothesis that extracellular TFAM can interact with RAGE and Mac-1 to function as a DAMP that causes pro-inflammatory microglial activation. Blocking this interaction may represent a potential target for attenuating the neuroinflammation observed in neurodegenerative diseases.


Assuntos
Citocinas/metabolismo , Lobo Frontal/metabolismo , Hipocampo/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Inflamação/metabolismo , Masculino , Microglia/metabolismo , Ratos , Ratos Sprague-Dawley
6.
J Neurosci ; 36(30): 7946-56, 2016 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-27466339

RESUMO

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.


Assuntos
Envelhecimento/imunologia , Encefalite/imunologia , Proteína HMGB1/imunologia , Hipocampo/imunologia , Mediadores da Inflamação/imunologia , Alarminas/imunologia , Animais , Masculino , Ratos , Distribuição Tecidual
7.
Brain Behav Immun ; 45: 171-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25433170

RESUMO

The circadian system regulates many physiological functions including inflammatory responses. For example, mortality caused by lipopolysaccharide (LPS) injection varies depending on the time of immunostimulation in mammals. The effects of more subtle challenges on the immune system and cellular mechanisms underlying circadian differences in neuroinflammatory responses are not well understood. Here we show that adult male Sprague-Dawley rats injected with a sub-septic dose of LPS during the light phase displayed elevated sickness behaviors and hippocampal cytokine production compared to rats injected during the dark phase. Microglia are the primary central nervous system (CNS) immune cell type and may mediate diurnal differences in sickness response, thus we explored whether microglia demonstrate temporal variations in inflammatory factors. Hippocampal microglia isolated from adult rats rhythmically expressed inflammatory factors and circadian clock genes. Microglia displayed robust rhythms of TNFα, IL1ß and IL6 mRNA, with peak cytokine gene expression occurring during the middle of the light phase. Microglia isolated during the light phase were also more reactive to immune stimulation; such that, ex vivo LPS treatment induced an exaggerated cytokine response in light phase-isolated microglia. Treating microglia with corticosterone ex vivo induced expression of the circadian clock gene Per1. However, microglia isolated from adrenalectomized rats maintained temporal differences in clock and inflammatory gene expression. This suggests circadian clock gene expression in microglia is entrained by, but oscillates in the absence of, glucocorticoids. Taken together, these findings demonstrate that microglia possess a circadian clock that influences inflammatory responses. These results indicate time-of-day is an important factor to consider when planning inflammatory interventions such as surgeries or immunotherapies.


Assuntos
Relógios Circadianos/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Glucocorticoides/farmacologia , Hipocampo/efeitos dos fármacos , Comportamento de Doença/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Microglia/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , Adrenalectomia , Animais , Relógios Circadianos/imunologia , Ritmo Circadiano/efeitos dos fármacos , Corticosterona/farmacologia , Citocinas/imunologia , Hipocampo/imunologia , Comportamento de Doença/fisiologia , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Masculino , Microglia/imunologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
8.
Brain Behav Immun ; 43: 60-7, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25043992

RESUMO

Bi-directional communication between the peripheral and central nervous systems has been extensively demonstrated. Aged rats exhibit a prolonged proinflammatory response in the hippocampus region of the brain following a peripheral bacterial infection, and this response in turn causes robust memory declines. Here we aimed to determine whether hepatic or splenic macrophages play a role in the maintenance of this central response. Proinflammatory cytokines measured in liver and spleen four days following an Escherichia coli infection revealed a potentiated proinflammatory response in liver, and to a lesser extent in spleen, in aged relative to young rats. To determine whether this potentiated response was caused by impaired bacterial clearance in these organs, E. coli colony forming units in liver and spleen were measured 4 days after infection, and there were no difference between young and aged rats in either organ. No E. coli was detected in the hippocampus, eliminating the possibility that the aged blood brain barrier allowed E. coli to enter the brain. Depletion of hepatic and splenic macrophages with clodronate-encapsulated liposomes effectively eliminated the proinflammatory response to E. coli at four days in both organs. However, this treatment failed to reduce the proinflammatory response in the hippocampus. Moreover, depletion of peripheral macrophages from liver and spleen did not prevent E. coli-induced memory impairment. These data strongly suggest that hepatic and splenic macrophages do not play a major role in the long-lasting maintenance of the proinflammatory response in the hippocampus of aged rats following a bacterial infection, or the memory declines that this response produces.


Assuntos
Infecções por Escherichia coli/complicações , Fígado/patologia , Macrófagos/patologia , Transtornos da Memória/microbiologia , Baço/patologia , Animais , Condicionamento Clássico/fisiologia , Citocinas/metabolismo , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Medo/fisiologia , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Fígado/metabolismo , Macrófagos/metabolismo , Masculino , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Ratos , Ratos Endogâmicos F344 , Baço/metabolismo
9.
Psychoneuroendocrinology ; 40: 191-200, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24485491

RESUMO

Chronic stress as well as chronic treatment with glucocorticoids (GCs) primes the neuroinflammatory response to a subsequent pro-inflammatory challenge. However, it remains unclear whether chronic GCs sensitize the response of key CNS immune substrates (i.e. microglia) to pro-inflammatory stimuli. In the present set of studies, male Sprague-Dawley rats underwent sham surgery or were adrenalectomized and then treated with varying concentrations of corticosterone (CORT; 0, 25, 50, and 75 µg/ml) administered in their drinking water. After 10 days of CORT exposure, whole hippocampus was collected and expression of glial activation markers measured or hippocampal microglia were isolated and challenged with LPS to probe for CORT-induced sensitization of pro-inflammatory responses. Chronic CORT exposure increased the gene expression of NLRP3, Iba-1, MHCII, and NF-κBIα in a concentration dependent manner. Chronic CORT (75 µg/ml) exposure potentiated the microglial proinflammatory response (TNFα, IL-1ß, IL-6 and NLRP3) to LPS compared to the microglial response of sham surgery animals treated with vehicle. The present set of results demonstrate that chronic exposure to GCs primes microglia to pro-inflammatory stimuli and add to a growing body of evidence suggesting that a permissive function of GCs is that of an endogenous danger signal or alarmin.


Assuntos
Glucocorticoides/farmacologia , Hipocampo/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Microglia/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/genética , Animais , Proteínas de Transporte , Citocinas/metabolismo , Sinergismo Farmacológico , Hipocampo/metabolismo , Masculino , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/metabolismo , Estresse Psicológico/induzido quimicamente , Estresse Psicológico/genética , Estresse Psicológico/metabolismo , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
10.
J Neurosci ; 32(42): 14641-8, 2012 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-23077050

RESUMO

To investigate the role of the pro-inflammatory cytokine interleukin-1ß (IL-1ß) in postoperative cognitive dysfunction (POCD) in aged rats, we used laparotomy to mimic human abdominal surgery in adult (3 months) and aged (24 months) F344/BN rats. We demonstrated that memory consolidation of the hippocampal-dependent contextual fear-conditioning task is significantly impaired in aged but not young rats 4 d after surgery. Hippocampal-independent auditory-cued fear memory was not disrupted by laparotomy in either age group. The hippocampal-dependent memory impairment was paralleled by elevations of IL-1ß in the hippocampus of aged animals 1 and 4 d after surgery. These findings support our substantial line of previous research showing that aged animals are more vulnerable to cognitive decline after a peripheral immune challenge. In addition, we demonstrated that a single intracisternal administration of interleukin-1 receptor antagonist (IL-1RA; 112 µg) at the time of surgery was sufficient to block both the behavioral deficit and the neuroinflammatory response. Injecting the same dose of IL-1RA peripherally failed to have a protective effect. These data provide strong support for the specific role of central, not peripheral, IL-1ß in POCD. Furthermore, the long-lasting presence of IL-1RA in the brain (4 d) compared with in the blood (<24 h) underscores the value of intracisternal administration of IL-1RA for therapeutic purposes.


Assuntos
Envelhecimento/efeitos dos fármacos , Cisterna Magna , Transtornos Cognitivos/prevenção & controle , Mediadores da Inflamação/administração & dosagem , Proteína Antagonista do Receptor de Interleucina 1/administração & dosagem , Interleucina-1beta/antagonistas & inibidores , Complicações Pós-Operatórias/prevenção & controle , Receptores de Interleucina-1/antagonistas & inibidores , Envelhecimento/patologia , Animais , Cisterna Magna/efeitos dos fármacos , Cisterna Magna/metabolismo , Cisterna Magna/patologia , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Injeções Intraventriculares , Interleucina-1beta/metabolismo , Masculino , Complicações Pós-Operatórias/metabolismo , Complicações Pós-Operatórias/patologia , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344 , Receptores de Interleucina-1/metabolismo
11.
Horm Behav ; 62(3): 219-27, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22370243

RESUMO

Healthy aged individuals are more likely to suffer profound memory impairments following a challenging life event such as a severe bacterial infection, surgery, or an intense psychological stressor, than are younger adults. Importantly, these peripheral challenges are capable of producing a neuroinflammatory response, (e.g., increased pro-inflammatory cytokines). In this review we will present the literature demonstrating that in the healthy aged brain this response is exaggerated and prolonged. Normal aging primes or sensitizes microglia and this appears to be the source of this amplified response. We will review the growing literature suggesting that a dysregulated neuroendocrine response in the aged organism is skewed toward higher brain CORT levels, and that this may play a critical role in priming microglia. Among the outcomes of an exaggerated neuroinflammatory response are impairments in synaptic plasticity, and reductions in key downstream mediators such as Arc and BDNF. We will show that each of these mechanisms is important for long-term memory formation, and is compromised by elevated pro-inflammatory cytokines.


Assuntos
Envelhecimento/imunologia , Cognição/fisiologia , Hipocampo/imunologia , Sistema Hipotálamo-Hipofisário/imunologia , Sistema Imunitário/imunologia , Humanos , Microglia/fisiologia
12.
Brain Behav Immun ; 25(7): 1408-15, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21536123

RESUMO

While stress and stress-induced glucocorticoids are classically considered immunosuppressive, they can also enhance proinflammatory responses to subsequent challenges. Corticosterone (CORT) primes rat immune cells, exacerbating pro-inflammatory responses to subsequent immune challenges. Stress can also sensitize pain. One possibility is that stress primes spinal immune cells, predominantly glia, which are key mediators in pain enhancement through their release of proinflammatory cytokines. Therefore, we aimed to identify whether prior CORT sensitizes spinal cord glia such that a potentiated pro-inflammatory response occurs to later intrathecal (IT) lipopolysaccharide (LPS), thereby enhancing pain. Rats received subcutaneous CORT/vehicle 24 h before IT LPS/vehicle. Hind paw pain thresholds were measured before CORT/vehicle, before and up to 48 h after IT LPS/vehicle. In separate rats treated as above, lumbar spinal cord tissue was collected and processed for proinflammatory mediators. CORT alone had no effect on pain responses, nor on any pro-inflammatory cytokines measured. LPS induced allodynia (decreased pain threshold) lasting <4 h and elevated spinal IL-1ß and IL-6 protein. Prior CORT potentiated allodynia, lasting >24 h following LPS and potentiated spinal IL-1 and IL-6 protein. Coadministration of IL-1 receptor antagonist with LPS IT completely blocked the allodynia irrespective of whether the system was primed by CORT or not. At 24 h, TLR2, TLR4, MD2, and CD14 mRNAs were significantly elevated within the spinal cord in the CORT+LPS group compared to all other groups. Prior CORT before a direct spinal immune challenge is able to potentiate pain responses and pro-inflammatory cytokine production.


Assuntos
Corticosterona/farmacologia , Glucocorticoides/farmacologia , Hiperalgesia/fisiopatologia , Inflamação/fisiopatologia , Lipopolissacarídeos/farmacologia , Medula Espinal/fisiopatologia , Animais , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Medição da Dor , Estimulação Física , Ratos , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
13.
Aging Dis ; 1(3): 212-231, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21132050

RESUMO

Healthy aged individuals are more likely to suffer profound memory impairments following a challenging life event such as a severe bacterial infection, surgery, or an intense psychological stressor, than are younger adults. These peripheral challenges are capable of producing a neuroinflammatory response, (e.g., increased pro-inflammatory cytokines), and in the healthy aged brain this response is exaggerated and prolonged. Normal aging primes or sensitizes microglia and this appears to be the source of this amplified response. Among the outcomes of this exaggerated neuroinflammatory response is an impairment in synaptic plasticity, and a reduction in key downstream mediators such as Arc and BDNF. Each of these mechanisms is important for long-term memory formation, and is compromised by elevated pro-inflammatory cytokines. Pharmacological, dietary and physical interventions are discussed as potential therapies to abrogate the challenge-induced neuroinflammatory response, thereby preventing or reducing memory deficits in aged subjects.

14.
J Pain ; 11(10): 1004-14, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20434956

RESUMO

UNLABELLED: Activation of spinal microglia and consequent release of proinflammatory mediators facilitate pain. Under certain conditions, responses of activated microglia can become enhanced. Enhanced microglial production of proinflammatory products may result from priming (sensitization), similar to macrophage priming. We hypothesized that if spinal microglia were primed by an initial inflammatory challenge, subsequent challenges may create enhanced pain. Here, we used a "two-hit" paradigm using 2 successive challenges, which affect overlapping populations of spinal microglia, presented 2 weeks apart. Mechanical allodynia and/or activation of spinal glia were assessed. Initially, laparotomy preceded systemic lipopolysaccharide (LPS). Prior laparotomy caused prolonged microglial (not astrocyte) activation plus enhanced LPS-induced allodynia. In this "two-hit" paradigm, minocycline, a microglial activation inhibitor, significantly reduced later exaggerated pain induced by prior surgery when minocycline was administered intrathecally for 5 days starting either at the time of surgery or 5 days before LPS administration. To test generality of the priming effect, subcutaneous formalin preceded intrathecal HIV-1 gp120, which activates spinal microglia and causes robust allodynia. Prior formalin enhanced intrathecal gp120-induced allodynia, suggesting that microglial priming is not limited to laparotomy and again supporting a spinal site of action. Therefore, spinal microglial priming may increase vulnerability to pain enhancement. PERSPECTIVE: Spinal microglia may become "primed" (sensitized) following their activation by disparate forms of peripheral trauma/inflammation. As a result, such primed microglia may overrespond to subsequent challenges, thereby enhancing pain intensity and duration.


Assuntos
Microglia/patologia , Dor/metabolismo , Dor/patologia , Animais , Antígeno CD11b/metabolismo , Modelos Animais de Doenças , Medicina Baseada em Evidências , Proteína Glial Fibrilar Ácida/metabolismo , Proteína gp120 do Envelope de HIV/administração & dosagem , Hiperalgesia/diagnóstico , Hiperalgesia/patologia , Injeções Espinhais , Laparotomia/efeitos adversos , Masculino , Microglia/metabolismo , Microglia/virologia , Dor/virologia , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/virologia , Fatores de Tempo
15.
Pharm Res ; 27(5): 841-54, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20224990

RESUMO

PURPOSE: Interleukin-10 (IL-10) is an anti-inflammatory molecule that has achieved interest as a therapeutic for neuropathic pain. In this work, the potential of plasmid DNA-encoding IL-10 (pDNA-IL-10) slowly released from biodegradable microparticles to provide long-term pain relief in an animal model of neuropathic pain was investigated. METHODS: PLGA microparticles encapsulating pDNA-IL-10 were developed and assessed both in vitro and in vivo. RESULTS: In vitro, pDNA containing microparticles activated macrophages, enhanced the production of nitric oxide, and increased the production of IL-10 protein relative to levels achieved with unencapsulated pDNA-IL-10. In vivo, intrathecally administered microparticles embedded in meningeal tissue, induced phagocytic cell recruitment to the cerebrospinal fluid, and relieved neuropathic pain for greater than 74 days following a single intrathecal administration, a feat not achieved with unencapsulated pDNA. Therapeutic effects of microparticle-delivered pDNA-IL-10 were blocked in the presence of IL-10-neutralizing antibody, and elevated levels of plasmid-derived IL-10 were detected in tissues for a prolonged time period post-injection (>28 days), demonstrating that therapeutic effects are dependent on IL-10 protein production. CONCLUSIONS: These studies demonstrate that microparticle encapsulation significantly enhances the potency of intrathecally administered pDNA, which may be extended to treat other disorders that require intrathecal gene therapy.


Assuntos
DNA/administração & dosagem , DNA/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Interleucina-10/genética , Doenças do Sistema Nervoso Periférico/terapia , Plasmídeos/genética , Animais , Comportamento Animal/fisiologia , Células Cultivadas , Imuno-Histoquímica , Injeções Espinhais , Interleucina-10/biossíntese , Ácido Láctico , Macrófagos/metabolismo , Masculino , Nanopartículas , Óxido Nítrico/metabolismo , Tamanho da Partícula , Doenças do Sistema Nervoso Periférico/líquido cefalorraquidiano , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Brain Behav Immun ; 24(2): 254-62, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19822205

RESUMO

In normal aging, a peripheral immune challenge induces a sensitized and protracted neuroinflammatory response in parallel with long-term memory (LTM) impairments. Pro-inflammatory mediators of neuroinflammation impair LTM, synaptic plasticity and LTP. The immediate early gene Arc is considered a critical protein regulating LTM and synaptic plasticity. The present investigation examined whether (1) a peripheral Escherichia coli infection suppresses hippocampal Arc expression, and (2) central pro-inflammatory cytokines (IL-1beta and IL-6) mediate the effects of peripheral E. coli infection on Arc and LTM. In 24 months F344xBN F1 rats, E. coli infection suppressed basal Arc gene expression as well as contextual fear conditioning-induced Arc expression. E. coli treatment failed to alter either basal or conditioning-induced c-Fos expression. At 24h post-infection, intra-cisterna magna (ICM) treatment with the anti-inflammatory cytokine IL-1RA blocked the E. coli-induced suppression of hippocampal Arc and increases in IL-6 protein. At 4-day post-infection, IL-1RA blocked the E. coli-induced LTM impairments and increases in IL-6 protein. The present results suggest that central pro-inflammatory cytokines play a salient role in the suppression of Arc and impairments of LTM by a peripheral immune challenge in older animals.


Assuntos
Envelhecimento/psicologia , Proteínas do Citoesqueleto/biossíntese , Infecções por Escherichia coli/psicologia , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Transtornos da Memória/psicologia , Memória/efeitos dos fármacos , Proteínas do Tecido Nervoso/biossíntese , Receptores de Interleucina-1/antagonistas & inibidores , Animais , Cisterna Magna , Condicionamento Psicológico/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Infecções por Escherichia coli/complicações , Infecções por Escherichia coli/genética , Medo/efeitos dos fármacos , Medo/psicologia , Genes fos/genética , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Injeções , Interleucina-6/biossíntese , Masculino , Transtornos da Memória/etiologia , Transtornos da Memória/prevenção & controle , Plasticidade Neuronal/efeitos dos fármacos , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Brain Behav Immun ; 21(5): 660-7, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17204394

RESUMO

Spinal cord glia (microglia and astrocytes) contribute to enhanced pain states. One model that has been used to study this phenomenon is intrathecal (i.t.) administration of gp120, an envelope glycoprotein of HIV-1 known to activate spinal cord glia and thereby induce low-threshold mechanical allodynia, a pain symptom where normally innocuous (non-painful) stimuli are perceived as painful. Previous studies have shown that i.t. gp120-induced allodynia is mediated via the release of the glial pro-inflammatory cytokines, tumor necrosis factor-alpha (TNF), and interleukin-1beta (IL-1). As we have recently reported that i.t. gp120 induces the release of interleukin-6 (IL-6), in addition to IL-1 and TNF, the present study tested whether this IL-6 release in spinal cord contributes to gp120-induced mechanical allodynia and/or to gp120-induced increases in TNF and IL-1. An i.t. anti-rat IL-6 neutralizing antibody was used to block IL-6 actions upon its release by i.t. gp120. This IL-6 blockade abolished gp120-induced mechanical allodynia. While the literature predominantly documents the cascade of pro-inflammatory cytokines as beginning with TNF, followed by the stimulation of IL-1, and finally TNF plus IL-1 stimulating the release of IL-6, the present findings indicate that a blockade of IL-6 inhibits the gp120-induced elevations of TNF, IL-1, and IL-6 mRNA in dorsal spinal cord, elevation of IL-1 protein in lumbar dorsal spinal cord, and TNF and IL-1 protein release into the surrounding lumbosacral cerebrospinal fluid. These results would suggest that IL-6 induces pain facilitation, and may do so in part by stimulating the production and release of other pro-inflammatory cytokines.


Assuntos
Proteína gp120 do Envelope de HIV/imunologia , Interleucina-1/imunologia , Interleucina-6/metabolismo , Limiar da Dor/fisiologia , Medula Espinal/imunologia , Fator de Necrose Tumoral alfa/imunologia , Análise de Variância , Animais , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Proteína gp120 do Envelope de HIV/administração & dosagem , Mediadores da Inflamação/metabolismo , Injeções Espinhais , Interleucina-1/genética , Interleucina-1/metabolismo , Interleucina-6/genética , Interleucina-6/imunologia , Masculino , Neuroglia/citologia , Neuroglia/imunologia , Neuroglia/metabolismo , Dor/imunologia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Medula Espinal/citologia , Medula Espinal/metabolismo , Estresse Mecânico , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
18.
Brain Behav Immun ; 21(1): 47-59, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16647243

RESUMO

Prior exposure to a stressor can potentiate CNS pro-inflammatory immune responses to a peripheral immune challenge. However, the neuroimmune substrate(s) mediating this effect has not been determined. The present investigation examined whether microglia serve as this neuroimmune substrate given that microglia are the primary immune effector cell in the CNS. The effect of inescapable shock (IS) on glial activation (MHC II, CD11b, Iba-1, and GFAP) and regulatory markers (CD200) in vivo, and microglia pro-inflammatory responses (interleukin-1beta; IL-1beta) to lipopolysaccharide (LPS) ex vivo, were assessed in rat hippocampus. IS upregulated the microglia activation marker MHC II 24h post-IS, while the astroglia marker GFAP was unaffected. IS also downregulated the neuronal glycoprotein CD200, which functions to hold microglia in a quiescent state. Moreover, IS potentiated the pro-inflammatory response to LPS ex vivo 24h post-IS in isolated hippocampal microglia. Finally, the behavioral controllability of shock was manipulated and the effect of escapable (controllable) shock was comparable to the effect of IS on hippocampal microglia responses to LPS ex vivo. The present results suggest that stress can activate microglia, thereby sensitizing the pro-inflammatory reactivity of microglia to immunogenic stimuli.


Assuntos
Hipocampo/imunologia , Antígenos de Histocompatibilidade/metabolismo , Interleucinas/metabolismo , Microglia/imunologia , Estresse Psicológico/imunologia , Animais , Perfilação da Expressão Gênica , Hipocampo/citologia , Antígenos de Histocompatibilidade/genética , Antígenos de Histocompatibilidade/imunologia , Imuno-Histoquímica , Interleucinas/genética , Interleucinas/imunologia , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Microglia/citologia , Microglia/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley
19.
Pain ; 115(1-2): 71-83, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15836971

RESUMO

Activated glial cells (microglia and astroglia) in the spinal cord play a major role in mediating enhanced pain states by releasing proinflammatory cytokines and other substances thought to facilitate pain transmission. In the present study, we report that intrathecal administration of minocycline, a selective inhibitor of microglial cell activation, inhibits low threshold mechanical allodynia, as measured by the von Frey test, in two models of pain facilitation. In a rat model of neuropathic pain induced by sciatic nerve inflammation (sciatic inflammatory neuropathy, SIN), minocycline delayed the induction of allodynia in both acute and persistent paradigms. Moreover, minocycline was able to attenuate established SIN-induced allodynia 1 day, but not 1 week later, suggesting a limited role of microglial activation in more perseverative pain states. Our data are consistent with a crucial role for microglial cells in initiating, rather than maintaining, enhanced pain responses. In a model of spinal immune activation by intrathecal HIV-1 gp120, we show that the anti-allodynic effects of minocycline are associated with decreased microglial activation, attenuated mRNA expression of interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNF-alpha), IL-1beta-converting enzyme, TNF-alpha-converting enzyme, IL-1 receptor antagonist and IL-10 in lumbar dorsal spinal cord, and reduced IL-1beta and TNF-alpha levels in the CSF. In contrast, no significant effects of minocycline were observed on gp120-induced IL-6 and cyclooxygenase-2 expression in spinal cord or CSF IL-6 levels. Taken together these data highlight the importance of microglial activation in the development of exaggerated pain states.


Assuntos
Citocinas/metabolismo , Hiperestesia/tratamento farmacológico , Hiperestesia/metabolismo , Microglia/metabolismo , Minociclina/administração & dosagem , Doenças do Sistema Nervoso Periférico/metabolismo , Medula Espinal/metabolismo , Animais , Relação Dose-Resposta a Droga , Hiperestesia/imunologia , Injeções Espinhais , Masculino , Microglia/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/imunologia , Ratos , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos
20.
J Neurosci ; 24(33): 7353-65, 2004 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-15317861

RESUMO

The present experiments examined the role of spinal proinflammatory cytokines [interleukin-1beta (IL-1)] and chemokines (fractalkine) in acute analgesia and in the development of analgesic tolerance, thermal hyperalgesia, and tactile allodynia in response to chronic intrathecal morphine. Chronic (5 d), but not acute (1 d), intrathecal morphine was associated with a rapid increase in proinflammatory cytokine protein and/or mRNA in dorsal spinal cord and lumbosacral CSF. To determine whether IL-1 release modulates the effects of morphine, intrathecal morphine was coadministered with intrathecal IL-1 receptor antagonist (IL-1ra). This regimen potentiated acute morphine analgesia and inhibited the development of hyperalgesia, allodynia, and analgesic tolerance. Similarly, intrathecal IL-1ra administered after the establishment of morphine tolerance reversed hyperalgesia and prevented the additional development of tolerance and allodynia. Fractalkine also appears to modulate the effects of intrathecal morphine because coadministration of morphine with intrathecal neutralizing antibody against the fractalkine receptor (CX3CR1) potentiated acute morphine analgesia and attenuated the development of tolerance, hyperalgesia, and allodynia. Fractalkine may be exerting these effects via IL-1 because fractalkine (CX3CL1) induced the release of IL-1 from acutely isolated dorsal spinal cord in vitro. Finally, gene therapy with an adenoviral vector encoding for the release of the anti-inflammatory cytokine IL-10 also potentiated acute morphine analgesia and attenuated the development of tolerance, hyperalgesia, and allodynia. Taken together, these results suggest that IL-1 and fractalkine are endogenous regulators of morphine analgesia and are involved in the increases in pain sensitivity that occur after chronic opiates.


Assuntos
Analgésicos Opioides/farmacologia , Quimiocinas CX3C/fisiologia , Hiperalgesia/imunologia , Interleucina-1/fisiologia , Proteínas de Membrana/fisiologia , Morfina/farmacologia , Analgésicos Opioides/administração & dosagem , Animais , Receptor 1 de Quimiocina CX3C , Quimiocina CX3CL1 , Quimiocinas CX3C/farmacologia , Tolerância a Medicamentos , Terapia Genética , Temperatura Alta , Hiperalgesia/terapia , Inflamação/imunologia , Injeções Espinhais , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/biossíntese , Interleucina-1/líquido cefalorraquidiano , Interleucina-10/genética , Masculino , Proteínas de Membrana/farmacologia , Morfina/administração & dosagem , Dor/imunologia , Manejo da Dor , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Citocinas/antagonistas & inibidores , Receptores de HIV/antagonistas & inibidores , Sialoglicoproteínas/administração & dosagem , Sialoglicoproteínas/farmacologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA