Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Clin Psychopharmacol ; 30(6): 897-906, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35025587

RESUMO

Substance use is characterized by reward processing dysregulation and cognitive control deficits. One area of research that remains relatively unexplored is the relationship between substance use and exploration-exploitation trade-offs, which involve a continuum from switching (exploration) to perseverative (exploitation). In dynamic, volatile environments, exploitation of well-known options can lead to habit-driven perseveration, and exploration of new opportunities can produce new information that may enhance one's future state. The primary aim was to investigate the relationship between regular substance use and spontaneous eyeblink rate (EBR) on exploration-exploitation behavior. Young adults (N = 83) aged 18-23 completed a single laboratory session. A dynamic foraging task was used to characterize exploration/exploitation behavior. Substance use was defined using the Externalizing Spectrum Inventory-Brief Form. Dopamine levels were operationalized using spontaneous EBR. The primary outcome was proportion of switch choices on the foraging task, which reflects a continuum of exploitation (low values) to exploration (high values) behavior. A linear mixed-effects regression was conducted to examine the effect of substance use and EBR on the proportion of switch trials. Results demonstrated a significant negative interaction between substance use and EBR on proportion of switch trials (p < .001). Participants with regular substance use and low EBR showed decreased switch choices, indicative of increased exploitation, compared to those with higher EBR. EBR was positively associated with proportion of switch trials (p = .032) and thus greater exploration. The relationship between substance use and increased exploitation in young adults appears specific to those with low EBR. (PsycInfo Database Record (c) 2022 APA, all rights reserved).


Assuntos
Comportamento Exploratório , Transtornos Relacionados ao Uso de Substâncias , Humanos , Adulto Jovem , Comportamento Exploratório/fisiologia , Recompensa , Hábitos , Transtornos Relacionados ao Uso de Substâncias/psicologia
2.
Annu Rev Neurosci ; 43: 355-374, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32109184

RESUMO

Opioid addiction and overdose are at record levels in the United States. This is driven, in part, by their widespread prescription for the treatment of pain, which also increased opportunity for diversion by sensation-seeking users. Despite considerable research on the neurobiology of addiction, treatment options for opioid abuse remain limited. Mood disorders, particularly depression, are often comorbid with both pain disorders and opioid abuse. The endogenous opioid system, a complex neuromodulatory system, sits at the neurobiological convergence point of these three comorbid disease states. We review evidence for dysregulation of the endogenous opioid system as a mechanism for the development of opioid addiction and/or mood disorder. Specifically, individual differences in opioid system function may underlie differences in vulnerability to opioid addiction and mood disorders. We also review novel research, which promises to provide more detailed understanding of individual differences in endogenous opioid neurobiology and its contribution to opioid addiction susceptibility.


Assuntos
Analgésicos Opioides/uso terapêutico , Dor Crônica/tratamento farmacológico , Depressão/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Animais , Overdose de Drogas/tratamento farmacológico , Humanos , Medicina de Precisão/métodos
3.
Burns ; 46(3): 503-513, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31859093

RESUMO

Burn injury pain is a significant public health problem. Burn injury treatment has improved tremendously in recent decades. However, an unintended consequence is that a larger number of patients now survive more severe injuries, and face intense pain that is very hard to treat. Although many efforts have been made to find alternative treatments, opioids remain the most effective medication available. Burn patients are frequently prescribed opioids in doses and durations that are significantly higher and longer than standard analgesic dosing guidelines. Despite this, many continue to experience unrelieved pain. They are also placed at a higher risk for developing dependence and opioid use disorder. Burn injury profoundly alters the functional state of the immune system. It also alters the expression levels of receptor, effector, and signaling molecules within the spinal cord's dorsal horn. These alterations could explain the reduced potency of opioids. However, recent studies demonstrate that different opioids signal preferentially via differential signaling pathways. This ligand-specific signaling by different opioids implies that burn injury may reduce the antinociceptive potency of opioids to different degrees, in a drug-specific manner. Indeed, recent findings hint at drug-specific differences in the ability of opioids to manage burn pain early after injury, as well as differences in their ability to prevent or treat the development of chronic and neuropathic pain. Here we review the current state of opioid treatment, as well as new findings that could potentially lead to opioid-based pain management strategies that may be significantly more effective than the current solutions.


Assuntos
Analgésicos Opioides/uso terapêutico , Dor Irruptiva/tratamento farmacológico , Queimaduras/terapia , Dor Processual/tratamento farmacológico , Receptores Opioides mu/metabolismo , Queimaduras/metabolismo , Regulação para Baixo , Expressão Gênica , Humanos , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Inflamação/metabolismo , Dor/tratamento farmacológico , Manejo da Dor/métodos , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Transdução de Sinais
4.
Artigo em Inglês | MEDLINE | ID: mdl-30790677

RESUMO

Pain management is the specialized medical practice of modulating pain perception and thus easing the suffering and improving the life quality of individuals suffering from painful conditions. Since this requires the modulation of the activity of endogenous systems involved in pain perception, and given the large role that the opioidergic system plays in pain perception, opioids are currently the most effective pain treatment available and are likely to remain relevant for the foreseeable future. This contributes to the rise in opioid use, misuse, and overdose death, which is currently characterized by public health officials in the United States as an epidemic. Historically, the majority of preclinical rodent studies were focused on morphine. This has resulted in our understanding of opioids in general being highly biased by our knowledge of morphine specifically. However, recent in vitro studies suggest that direct extrapolation of research findings from morphine to other opioids is likely to be flawed. Notably, these studies suggest that different opioid analgesics (opioid agonists) engage different downstream signaling effects within the cell, despite binding to and activating the same receptors. This recognition implies that, in contrast to the historical status quo, different opioids cannot be made equivalent by merely dose adjustment. Notably, even at equianalgesic doses, different opioids could result in different beneficial and risk outcomes. In order to foster further translational research regarding drug-specific differences among opioids, here we review basic research elucidating differences among opioids in pharmacokinetics, pharmacodynamics, their capacity for second messenger pathway activation, and their interactions with the immune system and the dopamine D2 receptors.


Assuntos
Analgésicos Opioides/farmacologia , Analgésicos Opioides/farmacocinética , Sistema Imunitário/efeitos dos fármacos , Receptores de Dopamina D2/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Analgésicos Opioides/classificação , Animais , Humanos , Receptores de Dopamina D2/metabolismo
5.
Drug Alcohol Depend ; 188: 113-118, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29772497

RESUMO

BACKGROUND: The association with opioid-abusing individuals or even the perception of opioid abuse by peers are risk factors for the initiation and escalation of abuse. Similarly, we demonstrated that morphine-treated animals housed with only morphine-treated animals (referred to as morphine only) acquire morphine conditioned place-preference (CPP) more readily than morphine-treated animals housed with drug-naïve animals (referred to as morphine cage-mates). However, the molecular mechanisms underlying these effects are still elusive. METHODS: Mice received repeated morphine or saline while housed as saline only, morphine only, or cage-mates. Then, they were examined for the expression levels of D1 dopamine receptor (D1DR), D2 dopamine receptor (D2DR), dopamine transporter (DAT), oxytocin, and Arginine-vasopressin (AVP) in the striatum using qPCR. Additionally, we examined the effects of the AVP-V1b receptor antagonist, SSR149415, on the acquisition of morphine conditioned place-preference (CPP). RESULTS: Increased striatal expression of D1DR and AVP was observed in morphine only animals, but not morphine cage-mates. No significant effects were observed on the striatal expression of D2DR, DAT, or oxytocin. Antagonizing the AVP-V1b receptors decreased the acquisition of morphine CPP in the morphine only mice, but did not alter the acquisition of morphine CPP in the morphine cage-mate mice. CONCLUSIONS: Housing with drug-naïve animals protects against the increase in striatal expression of D1DR and AVP elicited by morphine exposure. Moreover, our studies suggest that the protective effect of housing with drug-naïve animals on the acquisition of morphine reward might be, at least partially, mediated by AVP.


Assuntos
Arginina Vasopressina/biossíntese , Abrigo para Animais , Morfina/administração & dosagem , Receptores de Dopamina D1/biossíntese , Recompensa , Comportamento Social , Animais , Arginina Vasopressina/antagonistas & inibidores , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Dopamina D1/antagonistas & inibidores
6.
Behav Pharmacol ; 28(8): 642-647, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29049082

RESUMO

Our previous studies showed that altering solely the drug experience of the cage mates with which rodents are housed affects the development of morphine dependence. In this study, we used designer receptors exclusively activated by designer drugs to artificially increase or decrease the activity of peripheral dorsal root ganglia sensory neurons expressing the G-protein-coupled receptor MRGPRB4. This is because sensory MRGPRB4-expressing neurons were shown to specifically detect the sensation of massage-like stroking resulting from social grooming, which is an important affiliative social behavior in the rodent. Blocking the sensation of social grooming in morphine-treated mice housed with drug-naive mice (i.e. morphine cage mates) significantly increased the display of jumping behavior in morphine-withdrawn animals. Activating the sensation of social grooming in morphine-treated animals housed solely with other morphine-treated animals (i.e. morphine only) did not significantly alter the display of jumping behavior in morphine-withdrawn animals. Repetitive jumping behaviors have been shown to correlate with morphine dependence. Thus, this study showed a role of social grooming in the protective effect of being housed with drug-naive mice on the development of morphine dependence. It further confirms a role of social support in the development of substance use problems.


Assuntos
Asseio Animal , Dependência de Morfina/psicologia , Comportamento Social , Percepção do Tato , Animais , Drogas Desenhadas/farmacologia , Modelos Animais de Doenças , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Asseio Animal/efeitos dos fármacos , Asseio Animal/fisiologia , Camundongos Transgênicos , Morfina/administração & dosagem , Dependência de Morfina/fisiopatologia , Atividade Motora/efeitos dos fármacos , Entorpecentes/administração & dosagem , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Meio Social , Síndrome de Abstinência a Substâncias , Tato/efeitos dos fármacos , Tato/fisiologia , Percepção do Tato/efeitos dos fármacos , Percepção do Tato/fisiologia
7.
Neurosci Biobehav Rev ; 83: 697-712, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28552458

RESUMO

Opioid addiction is a chronic and relapsing mental health disorder. However, only some individuals exposed to opioids, either recreationally or during the course of pain management, will develop addiction. The reasons why some individuals develop addiction and some are spared are not fully understood. Studies indicate that it is likely a combination of genetic predispositions and environmental conditions. Given the role of environmental factors in human addiction, this review examines the role of social environments and social interactions in the development of opioid addictive-like behaviors in rodent studies. To date, three major behavioral approaches have been used in these studies, namely social isolation, environmental enrichment, and social housing with a variety of cage-mates that differ in their drug administration conditions. This review highlights the importance of an individual's social network in influencing the outcomes of drug abuse and the need to further elucidate the molecular mechanisms underlying these effects. Better understanding is likely to contribute to the development of novel and more effective treatments for addiction disorders.


Assuntos
Comportamento Aditivo/psicologia , Amigos , Transtornos Relacionados ao Uso de Opioides/psicologia , Comportamento Social , Animais , Humanos
8.
Pain Med ; 18(11): 2170-2180, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28340258

RESUMO

BACKGROUND: Pain is the most frequent complaint of burn-injured patients. Opioids are commonly used in the course of treatment. However, there is a lack of rodent studies that examine the differential effects of various opioids on burn pain. OBJECTIVE: This study compared the ability of morphine, oxycodone, and hydrocodone to suppress the development of burn-induced mechanical allodynia and reduce pain sensitivity. METHODS: Mice were examined for their baseline pain sensitivity thresholds using the von Frey Filaments test. Then, they were subjected to burn or sham injury and treated orally with morphine, oxycodone, hydrocodone (20 or 40 mg/kg), or saline twice daily throughout the study. They were retested on days 4, 7, 11, 14, 21, and 28 postburn. RESULTS: In the sham animals, morphine produced significant opioid-induced hyperalgesia (OIH). Development of OIH was minimal for hydrocodone and was not observed for oxycodone. Secondary mechanical allodynia was observed beginning four days after the burn injury and intensified with time. All opioids produced comparable antinociceptive effects. Hydrocodone was effective in suppressing the development of burn-induced mechanical allodynia and fully treated the burn-induced increase in pain sensitivity. In contrast, morphine and oxycodone had only minimal effects on the development of burn-induced mechanical allodynia and only partially treated the burn-induced increase in pain sensitivity. CONCLUSIONS: This study demonstrated that hydrocodone is effective in suppressing the development of burn-induced mechanical allodynia, while both morphine and oxycodone had minimal effects. These findings underscore the need for additional studies on the differences among various opioids using clinically relevant pain models.


Assuntos
Queimaduras/tratamento farmacológico , Hidrocodona/uso terapêutico , Morfina/uso terapêutico , Oxicodona/uso terapêutico , Analgésicos Opioides/uso terapêutico , Animais , Queimaduras/complicações , Relação Dose-Resposta a Droga , Hidrocodona/administração & dosagem , Hiperalgesia/tratamento farmacológico , Masculino , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Oxicodona/administração & dosagem , Dor/tratamento farmacológico , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos
9.
J Burn Care Res ; 38(5): 319-326, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28319527

RESUMO

Opioids are commonly used to treat severe, burn-induced pain. However, there is a lack of rodent studies that examine the differential effects of various opioids on burn pain. We recently demonstrated that hydrocodone was superior to other opioids in suppressing the development of burn-induced mechanical allodynia in the burned limb. This study monitored the development of mechanical allodynia and compared the abilities of morphine, oxycodone, and hydrocodone to reduce burn-induced mechanical allodynia in the limb contralateral to the burn. Mice were examined for their baseline pain sensitivity thresholds using the von Frey filaments test. Then, they were subjected to burn or sham injury and treated orally with morphine, oxycodone, hydrocodone (20 or 40 mg/kg), or saline twice daily throughout the study. They were retested on days 4, 7, 11, 14, 21, and 28 postburn. Hyperalgesia was developed in the contralateral, uninjured foot beginning 21 days after the burn injury. Hydrocodone was effective in suppressing the development of burn-induced mechanical allodynia. In contrast, morphine and oxycodone had only minimal effects on the development of burn-induced mechanical allodynia. The abnormal pain sensitivities that develop as a result of burn injuries are very difficult to treat and remain a significant public health problem. More rodent studies are required to improve our understanding of the differences among the currently available opioid analgesics in order to optimize the care provided to burn victims as well as those suffering from other pain modalities.


Assuntos
Analgésicos Opioides/farmacologia , Queimaduras/tratamento farmacológico , Temperatura Alta , Hiperalgesia/tratamento farmacológico , Dor/tratamento farmacológico , Animais , Queimaduras/complicações , Queimaduras/fisiopatologia , Relação Dose-Resposta a Droga , Hidrocodona , Hiperalgesia/etiologia , Camundongos , Morfina/farmacologia , Oxicodona/farmacologia , Dor/etiologia , Limiar da Dor
10.
Behav Pharmacol ; 28(4): 285-293, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28134661

RESUMO

Burn victim patients are frequently prescribed opioids at doses that are significantly higher than standard analgesic dosing guidelines, and, even despite an escalation in opioid dosing, many continue to experience pain. Thus, the aim of this study was to determine the effect of burn injury on opioid antinociception. Mice were examined for their baseline pain sensitivity thresholds using the von Frey filaments test. Then, they were subjected to burn or sham injury to the dorsal surface of the hindpaw and treated orally with morphine, oxycodone, hydrocodone (20 or 40 mg/kg), or saline twice daily throughout the study. They were retested on days 4, 7, 11, 14, 21, and 28 following the burn injury. The antinociceptive effects of the various drugs were analyzed by computing the daily difference between pain sensitivity threshold scores (in g) before and after treatment. This study showed that burn injury decreases opioid antinociception potency. A marked reduction was observed in the antinociceptive effectiveness of all opioids, and for both doses, in the burn-injured versus the sham animals. These results suggest that burn trauma limits the ability of opioids to be effective in reducing pain.


Assuntos
Analgésicos Opioides/farmacologia , Queimaduras/complicações , Limiar da Dor/efeitos dos fármacos , Dor/tratamento farmacológico , Administração Oral , Analgésicos Opioides/administração & dosagem , Animais , Queimaduras/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hidrocodona/administração & dosagem , Hidrocodona/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Morfina/farmacologia , Oxicodona/administração & dosagem , Oxicodona/farmacologia , Fatores de Tempo
11.
Pain Med ; 17(5): 908-914, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26349634

RESUMO

BACKGROUND: Opioids alter the responses of D2-like dopamine receptors (D2DRs), known to be involved in the pathology of addiction and other mental illnesses. Importantly, our recent results demonstrated that various opioids differentially modulate the behavioral responses of D2DRs. OBJECTIVE: To examine the effect of various opioids on striatal activation levels of Akt and ERK1/2, as well as the signaling responses of D2DRs following opioid exposure. METHODS: Mice were pre-treated with 20 mg/kg morphine, hydrocodone, oxycodone, or saline for 6 days. Twenty-four hours later, mice were injected with vehicle or a D2/D3 receptor agonist, quinpirole. Thirty minutes later, dorsal striatum was collected and analyzed using Western blot. RESULTS: In morphine-pretreated animals, baseline Akt activation level was unchanged, but was reduced in response to quinpirole. In contrast, baseline Akt activation levels were reduced in mice pretreated with hydrocodone and oxycodone, but were unchanged in response to quinpirole. In mice pretreated with all opioids, baseline ERK2 activation levels were unchanged and increased in response to quinpirole. However, quinpirole-induced ERK2 activation was significantly higher than drug naïve animals only in the morphine-pretreated mice. CONCLUSIONS: Various opioids differentially modulate the baseline activation levels of signaling molecules, which in turn results in ligand-selective effects on the responses to a D2/D3 dopamine receptor agonist. This demonstrates a complex interplay between opioid receptors and D2DRs, and supports the notion that various opioids carry differential risks to the dopamine reward system. This information should be considered when prescribing opioid pain medication, to balance effectiveness with minimal risk.

12.
Behav Brain Res ; 284: 37-41, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25617530

RESUMO

Oxycodone and hydrocodone are opioids which are widely used for pain management and are also commonly misused and abused. The exposure to opioid analgesics has been associated with altered responses of D2-like dopamine receptors (D2DRs). Our recent results suggest that various opioids will differentially modulate the responses of D2DRs. The D2DRs are known to be involved in the pathology of addiction and other mental illnesses, indicating the need to improve our understanding of the effects of opioid analgesics on the responses of the D2DRs. Thus, in this study, we first established equianalgesic oral doses of oxycodone, hydrocodone, and morphine using the tail withdrawal assay. Then, mice were orally administered (gavage) with the various opioids or saline once daily for 6 days. Twenty-four hours later, the mice were tested for their locomotor response to quinpirole, a D2/D3 dopamine receptor agonist. Mice pretreated with oxycodone showed significantly greater locomotor supersensitivity to quinpirole than did morphine-pretreated mice, while hydrocodone-pretreated mice showed sensitivity in between that of mice treated with morphine and oxycodone. This finding suggests that various opioids differentially modulate the responses of D2DRs. It provides further evidence supporting of the notion that various opioids carry differential risks to the dopamine reward system.


Assuntos
Analgésicos Opioides/farmacologia , Hidrocodona/farmacologia , Morfina/farmacologia , Oxicodona/farmacologia , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/metabolismo , Acatisia Induzida por Medicamentos/metabolismo , Animais , Agonistas de Dopamina/farmacologia , Temperatura Alta , Masculino , Camundongos Endogâmicos C57BL , Dor/tratamento farmacológico , Dor/metabolismo , Medição da Dor , Quimpirol/farmacologia , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D3/agonistas
13.
Drug Alcohol Depend ; 142: 283-9, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25048393

RESUMO

BACKGROUND: Adolescent opioid abuse is on the rise, and current treatments are not effective in reducing rates of relapse. Our previous studies demonstrated that social housing conditions alter the acquisition rate of morphine conditioned place preference (CPP) in adolescent mice. Specifically, the acquisition rate of morphine CPP is slower in morphine-treated animals housed with drug-naïve animals. Thus, here we tested the effect of social housing conditions on the development of morphine dependence and the extinction rate of an acquired morphine CPP. METHODS: Adolescent male mice were group-housed in one of two housing conditions. They were injected for 6 days (PND 28-33) with 20 mg/kg morphine. Morphine only mice are animals where all four mice in the cage received morphine. Morphine cage-mate mice are morphine-injected animals housed with drug-naïve animals. Mice were individually tested for spontaneous withdrawal signs by quantifying jumping behavior 4, 8, 24, and 48 h after the final morphine injection. Then, mice were conditioned to acquire morphine CPP and were tested for the rate of extinction. RESULTS: Morphine cage-mates express less jumping behavior during morphine withdrawal as compared to morphine only mice. As expected, morphine cage-mate animals acquired morphine CPP more slowly than the morphine only animals. Additionally, morphine cage-mates extinguished morphine CPP more readily than morphine only mice. CONCLUSIONS: Social housing conditions modulate morphine dependence and the extinction rate of morphine CPP. Extinction testing is relevant to human addiction because rehabilitations like extinction therapy may be used to aid human addicts in maintaining abstinence from drug use.


Assuntos
Condicionamento Operante/fisiologia , Extinção Psicológica/fisiologia , Abrigo para Animais , Dependência de Morfina/psicologia , Morfina/administração & dosagem , Entorpecentes/administração & dosagem , Meio Social , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Extinção Psicológica/efeitos dos fármacos , Masculino , Camundongos
14.
Drug Alcohol Depend ; 132(3): 420-6, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23932842

RESUMO

BACKGROUND: There is a lack of studies that examine the effects of opioid maintenance drugs on the developing adolescent brain, limiting the ability of physicians to conduct a science-based risk assessment on the appropriateness of these treatments for that age group. Our recent observations indicate higher potential risks in repeated exposure to morphine during adolescence, specifically to the D2/D3 dopamine receptors' signaling. Disturbances in dopaminergic signaling could have broader implications for long-term mental health. Thus, this study examined whether buprenorphine and methadone differentially alter the responses of the D2/D3 dopamine receptors in adolescents. METHODS: Adolescent mice were orally administered buprenorphine (0.1-0.4 mg/kg), methadone (25-100 mg/kg), or saline once daily for 6 days. Two hours or three days later, the mice were tested for their locomotor response to 10 mg/kg quinpirole, a D2/D3 dopamine receptor agonist. RESULTS: Buprenorphine-treated adolescent mice did not significantly differ from control drug-naïve animals in their response to quinpirole. However, an enhanced response was observed in methadone-treated adolescent animals. This enhanced locomotion was significantly higher two hours following the final dose of methadone, as compared to three days afterwards. CONCLUSIONS: This study suggests that exposure to various opioids carries differential probabilities of altering the highly sensitive neurochemistry of adolescent brains. Methadone exposure disturbs the D2-like receptor's response, indicating a potential risk in administering methadone to adolescents (either for the treatment of opioid dependency/abuse or for pain management). In contrast, buprenorphine appears to have a significantly lower effect on the behavioral sensitivity of D2/D3 dopamine receptors in adolescents.


Assuntos
Analgésicos Opioides/farmacologia , Buprenorfina/farmacologia , Metadona/farmacologia , Atividade Motora/fisiologia , Receptores de Dopamina D2/fisiologia , Receptores de Dopamina D3/fisiologia , Fatores Etários , Animais , Agonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D3/agonistas
15.
Restor Neurol Neurosci ; 30(4): 291-302, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22572023

RESUMO

PURPOSE: Traumatic brain injury (TBI) is a major health concern today and effective treatments must be developed in order to combat the numerous TBIs that occur each year. Multiple b-vitamins have been shown to have neuroprotective effects, however, folic acid (B9) has not been widely studied. The current study examined two different doses in a rodent model of controlled cortical impact (CCI) TBI. METHODS: Sham procedures or a unilateral parietal controlled cortical impact injury was induced. Rats were administered either vehicle or folic acid in an 80 µg/kg or 800 µg/kg dose. Rats were tested on the bilateral tactile adhesive removal task, rotarod task and the Morris water maze. Brains were examined to determine lesion size and neuronal loss. RESULTS: Neither of the folic acid-treated groups showed improvement on any behavioral task or anatomical measure post-CCI and the high dose group had increased neuronal loss compared to the vehicle. Administration of the high dose in sham animals resulted in some behavioral dysfunction and significant neuronal loss. CONCLUSIONS: The results from this study suggest that folic acid may not represent an effective avenue for treatment and that higher doses may actually be detrimental following TBI.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Ácido Fólico/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Lesões Encefálicas/patologia , Ácido Fólico/administração & dosagem , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos
16.
J Alzheimers Dis ; 29(3): 503-14, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22330832

RESUMO

This paper propounds the Amyloids as Sensors and Protectors (ASAP) hypothesis. In this novel hypothesis, we provide evidence that amyloids are capable of sensing dysfunction, and after misfolding, initiate protective cellular responses. Amyloid proteins are initially protective, but chronic stress and overstimulation of the amyloid sensor leads to pathology. This proposed ASAP hypothesis has two sequential stages: (i) sensing, and then (ii) protection. Sensing involves a conformational change of amyloids in response to the cellular environment. The protection aspect translates conformational change into a cellular response via several mechanisms. The most obvious mechanism is that protein misfolding triggers the protective unfolded protein response, and thus downregulates protein translation and increases chaperone proteins. Other documented responses include metabolic pathways and microRNAs. This ASAP hypothesis has precedence, as amyloid sensors exist (evidenced by CPEB and Sup35), and both prion and amyloid-ß sensing redox stress and metals. Our hypothesis expands on previous observations to link sensing with inciting protective cellular response. Furthermore, we substantiate the ASAP hypothesis with previously published evidence from several amyloid diseases. This novel hypothesis links disparate findings in amyloid diseases: metabolic dysfunction, unfolding protein response/chaperones, modification of amyloids, and nutrient or caloric sensing. While this hypothesis can be applied to Alzheimer's disease, it goes beyond the Alzheimer's context. Thus all amyloid proteins can potentially act as sensors of misfolding-causing stress. Finally, this hypothesis will allow for the sensor mechanism and metabolic dysfunction to serve as biomarkers of the diseases as well as therapeutic targets early in disease pathology.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Amiloide/metabolismo , Amiloide/uso terapêutico , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...