Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Neurotrauma ; 37(1): 27-42, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31347447

RESUMO

Although clinical studies identify traumatic brain injury (TBI) as a risk factor for the development of substance use disorder, much remains unknown about the possible underlying pathogenesis and age-specific effects. Thus, the aim of this study is to test the hypothesis that at an age of ongoing maturation, adolescent TBI alters elements of the reward pathway, resulting in increased sensitivity to the rewarding effects of a subthreshold dose of cocaine that does not induce significant behavioral changes in naïve, non-injured mice. Specifically, these results were derived from the combination of the controlled cortical impact model of TBI, performed on either adolescent (6 weeks) or young adult (8 weeks) mice, followed by the cocaine-induced conditioned place preference assay 2 weeks later. Using three-dimensional isosurface rendering and volumetric image analysis, TBI was found to induce neuromorphological changes such as decreased dendritic complexity and reduced spine density in brain regions essential for reward perception and processing of drug-induced euphoria. Further, we demonstrated that these neuronal changes may affect the differential expression of dopamine-associated genes. Our analysis also provided evidence for age-related differences in immune response and the distinct involvement of augmented microglial phagocytic activity in the remodeling of neuronal structures in the adolescent TBI brain. Our studies suggest that TBI during adolescence, a period associated with ongoing maturation of dopaminergic systems, may subsequently enhance the abuse liability of cocaine in adulthood.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Dopamina/metabolismo , Neuroimunomodulação/fisiologia , Recompensa , Fatores Etários , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Behav Brain Res ; 379: 112333, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31682867

RESUMO

The pathology of traumatic brain injury (TBI) adversely affects many brain regions, often resulting in the development of comorbid psychiatric disorders including substance use disorders (SUD). Although traditionally thought to be an epidemic that predominantly affects males, recent clinical studies report females have higher rates of concussions and longer recovery times than males. Yet, how neurotrauma, particularly deep within the brain, between the sexes is differentially manifested remains largely unknown. The risk of TBI peaks during adolescence when neuronal networks that regulate reward behaviors are not fully developed. Previously, using the conditioned place preference (CPP) assay, we found that adolescent TBI increased susceptibility to the rewarding effects of cocaine in male mice. Further, we observed augmented inflammatory profiles, increased microglial phagocytosis of neuronal proteins, and decreased neuronal spine density in the NAc. Notably, the extent of sex differences in SUD susceptibility following TBI has not be investigated. Thus, here we ask the central question of whether the adolescent TBI-induced neuroinflammatory profile at reward centers is divergent in a sex-dependent manner. Using the CPP assay, we found that female mice with high levels of female sex hormones at the time of adolescent TBI demonstrated neuroprotection against increased sensitivity to the rewarding effects of cocaine. These studies also provide evidence of significantly reduced microglial activation and phagocytosis of neuronal proteins within the NAc of females. Overall, our results offer crucial insight into how adolescent TBI impacts the reward pathway in a sex depending manner that could explain a vulnerability to addiction-like behavior.


Assuntos
Comportamento Aditivo , Comportamento Animal , Lesões Encefálicas Traumáticas , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Estradiol/metabolismo , Inflamação , Neuroproteção , Progesterona/metabolismo , Recompensa , Caracteres Sexuais , Animais , Comportamento Aditivo/imunologia , Comportamento Aditivo/metabolismo , Comportamento Aditivo/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Feminino , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroproteção/imunologia , Neuroproteção/fisiologia
3.
Exp Neurol ; 317: 191-201, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30862466

RESUMO

Recent reports provide evidence for increased risk of substance use disorders (SUD) among patients with a history of early-life traumatic brain injury (TBI). Preclinical research utilizing animal models of TBI have identified injury-induced inflammation, blood-brain barrier permeability, and changes to synapses and neuronal networks within regions of the brain associated with the perception of reward. Importantly, these reward pathway networks are underdeveloped during childhood and adolescence, and early-life TBI pathology may interrupt ongoing maturation. As such, maladaptive changes induced by juvenile brain injury may underlie increased susceptibility to SUD. In this review, we describe the available clinical and preclinical evidence that identifies SUD as a persistent psychiatric consequence of pediatric neurotrauma by discussing (1) the incidence of early-life TBI, (2) how preclinical studies model TBI and SUD, (3) TBI-induced neuropathology and neuroinflammation in the corticostriatal regions of the brain, and (4) the link between childhood or adolescent TBI and addiction in adulthood. In summary, preclinical research utilizes an innovative combination of models of early-life TBI and SUD to recapitulate clinical features and to determine how TBI promotes a risk for the development of SUD. However, causal processes that link TBI and SUD remain unclear. Additional research to identify and therapeutically target underlying mechanisms of aberrant reward pathway development will provide a launching point for TBI and SUD treatment strategies.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Transtornos Relacionados ao Uso de Substâncias/etiologia , Adolescente , Animais , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/fisiopatologia , Criança , Pré-Escolar , História Antiga , Humanos , Recém-Nascido , Fatores de Risco , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia
4.
Cell Transplant ; 26(7): 1178-1192, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28933216

RESUMO

Clinical studies have identified traumatic brain injury (TBI) as a risk factor for the development of cocaine dependence. This claim is supported by our recent preclinical studies showing enhancement of the rewarding effects of cocaine in mice sustaining moderate controlled cortical impact (CCI) injury during adolescence. Here we test the efficacy of dexamethasone, an anti-inflammatory corticosteroid, to attenuate augmentation of the behavioral response to cocaine observed in CCI-TBI animals using the conditioned place preference (CPP) assay. These studies were performed in order to determine whether proinflammatory activity in the nucleus accumbens (NAc), a key brain nucleus in the reward pathway, mediates enhanced cocaine-induced CPP in adolescent animals sustaining moderate CCI-TBI. Our data reveal robust glial activation in the NAc following CCI-TBI and a significant increase in the cocaine-induced CPP of untreated CCI-TBI mice. Furthermore, our results show that dexamethasone treatment following CCI-TBI can attenuate the cocaine place preference of injured animals without producing aversion in the CPP assay. Our studies also found that dexamethasone treatment significantly reduced the expression of select immune response genes including Monocyte chemoattractant protein-1 (MCP-1/CCL2) and intercellular adhesion molecule-1 ( ICAM-1), returning their expression to control levels, which prompted an investigation of peripheral blood monocytes in dexamethasone-treated animals. Experimental findings showed that no craniectomy/dexamethasone mice had a significant increase, while CCI-TBI/dexamethasone animals had a significant decrease in the percentage of circulating nonclassical patrolling monocytes. These results suggest that a portion of these monocytes may migrate to the brain in response to CCI-TBI, potentially sparing the development of chronic neuroinflammation in regions associated with the reward circuitry such as the NAc. Overall, our findings indicate that anti-inflammatory agents, such as dexamethasone, may be effective in normalizing the rewarding effects of cocaine following CCI-TBI.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Cocaína/efeitos adversos , Dexametasona/uso terapêutico , Recompensa , Envelhecimento/patologia , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Lesões Encefálicas Traumáticas/diagnóstico , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/patologia , Dexametasona/farmacologia , Diagnóstico por Imagem , Regulação da Expressão Gênica/efeitos dos fármacos , Imunidade/genética , Masculino , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Monócitos/patologia , Neuroglia/patologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Núcleo Accumbens/patologia , Fenótipo
5.
Sci Rep ; 7(1): 3846, 2017 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-28630485

RESUMO

Traumatic brain injury (TBI) contributes to one third of injury related deaths in the US. Treatment strategies for TBI are supportive, and the pathophysiology is not fully understood. Secondary mechanisms of injury in TBI, such as oxidative stress and inflammation, are points at which intervention may reduce neuropathology. Evidence suggests that reactive oxygen species (ROS) propagate blood-brain barrier (BBB) hyperpermeability and inflammation following TBI. We hypothesized that targeted detoxification of ROS may improve the pathological outcomes of TBI. Following TBI, endothelial activation results in a time dependent increase in vascular expression of ICAM-1. We conjugated catalase to anti-ICAM-1 antibodies and administered the conjugate to 8 wk old C57BL/6J mice 30 min after moderate controlled cortical impact injury. Results indicate that catalase targeted to ICAM-1 reduces markers of oxidative stress, preserves BBB permeability, and attenuates neuropathological indices more effectively than non-targeted catalase and anti-ICAM-1 antibody alone. Furthermore, the study of microglia by two-photon microscopy revealed that anti-ICAM-1/catalase prevents the transition of microglia to an activated phenotype. These findings demonstrate the use of a targeted antioxidant enzyme to interfere with oxidative stress mechanisms in TBI and provide a proof-of-concept approach to improve acute TBI management that may also be applicable to other neuroinflammatory conditions.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Catalase/administração & dosagem , Molécula 1 de Adesão Intercelular/metabolismo , Substâncias Protetoras/administração & dosagem , Animais , Barreira Hematoencefálica/metabolismo , Lesões Encefálicas Traumáticas/tratamento farmacológico , Células Endoteliais/metabolismo , Peróxido de Hidrogênio/metabolismo , Camundongos , Microglia/metabolismo , Neuroglia/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
6.
Neurosci Biobehav Rev ; 77: 209-218, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28359860

RESUMO

Recent studies have helped identify multiple factors affecting increased risk for substance use disorders (SUDs) following traumatic brain injury (TBI). These factors include age at the time of injury, repetitive injury and TBI severity, neurocircuits, neurotransmitter systems, neuroinflammation, and sex differences. This review will address each of these factors by discussing 1) the clinical and preclinical data identifying patient populations at greatest risk for SUDs post-TBI, 2) TBI-related neuropathology in discrete brain regions heavily implicated in SUDs, and 3) the effects of TBI on molecular mechanisms that may drive substance abuse behavior, like dopaminergic and glutamatergic transmission or neuroimmune signaling in mesolimbic regions of the brain. Although these studies have laid the groundwork for identifying factors that affect risk of SUDs post-TBI, additional studies are required. Notably, preclinical models have been shown to recapitulate many of the behavioral, cellular, and neurochemical features of SUDs and TBI. Therefore, these models are well suited for answering important questions that remain in future investigations.


Assuntos
Lesões Encefálicas Traumáticas , Transtornos Relacionados ao Uso de Substâncias , Animais , Humanos , Modelos Animais , Risco
7.
Psychopharmacology (Berl) ; 234(11): 1671-1681, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28251297

RESUMO

RATIONALE: Metabotropic glutamate 2 and 3 (mGluR2/3) receptors are implicated in drug addiction as they limit excessive glutamate release during relapse. N-acetylaspartylglutamate (NAAG) is an endogenous mGluR2/3 agonist that is inactivated by the glutamate carboxypeptidase II (GCPII) enzyme. GCPII inhibitors, and NAAG itself, attenuate cocaine-seeking behaviors. However, their effects on the synthetic cathinone 3,4-methylenedioxypyrovalerone (MDPV) have not been examined. OBJECTIVES: We determined whether withdrawal following repeated MDPV administration alters GCPII expression in corticolimbic regions. We also examined whether a GCPII inhibitor (2-(phosphonomethyl)-pentanedioic acid (2-PMPA)), and NAAG, reduce the rewarding and locomotor-stimulant effects of MDPV in rats. METHODS: GCPII was assessed following repeated MDPV exposure (7 days). The effects of 2-PMPA and NAAG on acute MDPV-induced hyperactivity were determined using a locomotor test. We also examined the inhibitory effects of 2-PMPA and NAAG on MDPV-induced place preference, and whether the mGluR2/3 antagonist LY341495 could prevent these effects. RESULTS: MDPV withdrawal reduced GCPII expression in the prefrontal cortex. Systemic injection of 2-PMPA (100 mg/kg) did not affect the hyperactivity produced by MDPV (0.5-3 mg/kg). However, nasal administration of NAAG did reduce MDPV-induced ambulation, but only at the highest dose (500 µg/10 µl). We also showed that 2-PMPA (10-30 mg/kg) and NAAG (10-500 µg/10 µl) dose-dependently attenuated MDPV place preference, and that the effect of NAAG was blocked by LY341495 (3 mg/kg). CONCLUSIONS: These findings demonstrate that MDPV withdrawal produces dysregulation in the endogenous NAAG-GCPII signaling pathway in corticolimbic circuitry. Systemic administration of the GCPII inhibitor 2-PMPA, or NAAG, attenuates MDPV reward.


Assuntos
Alcaloides/administração & dosagem , Benzodioxóis/administração & dosagem , Dipeptídeos/farmacologia , Glutamato Carboxipeptidase II/biossíntese , Compostos Organofosforados/farmacologia , Pirrolidinas/administração & dosagem , Recompensa , Aminoácidos/farmacologia , Animais , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Relação Dose-Resposta a Droga , Antagonistas de Aminoácidos Excitatórios/farmacologia , Glutamato Carboxipeptidase II/antagonistas & inibidores , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de Glutamato Metabotrópico/fisiologia , Xantenos/farmacologia , Catinona Sintética
8.
J Neurotrauma ; 34(1): 165-181, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27026056

RESUMO

Clinical psychiatric disorders of depression, anxiety, and substance abuse are most prevalent after traumatic brain injury (TBI). Pre-clinical research has focused on depression and anxiety post-injury; however, virtually no data exist examining whether the preference for illicit drugs is affected by traumatic injury in the developing adolescent brain. Using the controlled cortical impact (CCI) model of TBI and the conditioned place preference (CPP) assay, we tested the underlying hypothesis that brain injury during adolescence exacerbates the rewarding properties of cocaine in adulthood possibly through an active inflammatory status in the mesolimbic pathway. Six-week old, C57BL/6 mice sustained a single CCI-TBI to the right somatosensory cortex. CPP experiments with cocaine began 2 weeks post-TBI. Animals receiving cocaine displayed significant place preference shifts compared to saline controls. Further, within the cocaine-experienced cohort, moderate CCI-TBI during adolescence significantly increased the preference shift in adulthood when compared to naïve controls. Additionally, persistent neuroinflammatory responses were observed in the cortex, nucleus accumbens (NAc), and ventral tegmental area post-CCI-TBI. Significant increases in both astrocytic, glial fibrillary acidic protein, and microglial, ionization basic acid 1, markers were observed in the NAc at the end of CPP testing. Moreover, analysis using focused array gene expression panels identified the upregulation of numerous inflammatory genes in moderate CCI-TBI animals, compared to naïve controls, both in the cortex and NAc at 2 weeks post-TBI, before onset of cocaine administration. These results suggest that sustaining moderate TBI during adolescence may augment the rewarding effects of psychostimulants in adulthood, possibly by induction of chronic mesolimbic neuroinflammation.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Córtex Cerebral/metabolismo , Cocaína/administração & dosagem , Mediadores da Inflamação/metabolismo , Núcleo Accumbens/metabolismo , Recompensa , Fatores Etários , Animais , Lesões Encefálicas Traumáticas/patologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Inflamação/metabolismo , Inflamação/patologia , Sistema Límbico/efeitos dos fármacos , Sistema Límbico/metabolismo , Sistema Límbico/patologia , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/patologia , Autoadministração
9.
Neurobiol Learn Mem ; 134 Pt B: 275-86, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27481220

RESUMO

Human studies of sleep and cognition have established thatdifferent sleep stages contribute to distinct aspects of cognitive and emotional processing. However, since the majority of these findings are based on single-night studies, it is difficult to determine whether such effects arise due to individual, between-subject differences in sleep patterns, or from within-subject variations in sleep over time. In the current study, weinvestigated the longitudinal relationship between sleep patterns and cognitive performance by monitoring both in parallel, daily, for a week. Using two cognitive tasks - one assessing emotional reactivity to facial expressions and the other evaluating learning abilities in a probabilistic categorization task - we found that between-subjectdifferences in the average time spent in particular sleep stages predicted performance in these tasks far more than within-subject daily variations. Specifically, the typical time individualsspent in Rapid-Eye Movement (REM) sleep and Slow-Wave Sleep (SWS) was correlated to their characteristic measures of emotional reactivity, whereas the typical time spent in SWS and non-REM stages 1 and 2 was correlated to their success in category learning. These effects were maintained even when sleep properties werebased onbaseline measures taken prior to the experimental week. In contrast, within-subject daily variations in sleep patterns only contributed to overnight difference in one particular measure of emotional reactivity. Thus, we conclude that the effects of natural sleep onemotional cognition and categorylearning are more trait-dependent than state-dependent, and suggest ways to reconcile these results with previous findings in the literature.


Assuntos
Reconhecimento Facial/fisiologia , Aprendizagem por Probabilidade , Fases do Sono/fisiologia , Adulto , Emoções/fisiologia , Expressão Facial , Feminino , Humanos , Masculino , Sono REM/fisiologia , Adulto Jovem
10.
J Hematol Oncol ; 8: 33, 2015 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-25888494

RESUMO

As the population ages and lifestyles change in concordance, the number of patients suffering from ischemic stroke and its associated disabilities is increasing. Studies on determining the relationship between endothelial progenitor cells (EPCs) and ischemic stroke have become a new hot spot and have reported that EPCs may protect the brain against ischemic injury, promote neurovascular repair, and improve long-term neurobehavioral outcomes. More importantly, they introduce a new perspective for prognosis assessment and therapy of ischemic stroke. However, EPCs' origin, function, influence factors, injury repair mechanisms, and cell-based therapy strategies remain controversial. Particularly, research conducted to date has less clinical studies than pre-clinical experiments on animals. In this review, we summarized and analyzed the current understanding of basic characteristics, influence factors, functions, therapeutic strategies, and disadvantages of EPCs as well as the regulation of inflammatory factors involved in the function and survival of EPCs after ischemic stroke. Identifying potential therapeutic effects of EPCs in ischemic stroke will be a challenging but an incredibly important breakthrough in neurology, which may bring promise for patients with ischemic stroke.


Assuntos
Células Progenitoras Endoteliais/metabolismo , Acidente Vascular Cerebral/sangue , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA