Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 256
Filtrar
1.
Pharmacogenomics J ; 12(4): 319-27, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21358750

RESUMEN

Opioid addiction is a chronic disease with high genetic contribution and a large inter-individual variability in therapeutic response. The goal of this study was to identify pharmacodynamic factors that modulate methadone dose requirement. The neurotrophin family is involved in neural plasticity, learning, memory and behavior and deregulated neural plasticity may underlie the pathophysiology of drug addiction. Brain-derived neurotrophic factor (BDNF) was shown to affect the response to methadone maintenance treatment. This study explores the effects of polymorphisms in the nerve growth factor (ß polypeptide) gene, NGFB, on the methadone doses required for successful maintenance treatment for heroin addiction. Genotypes of 14 NGFB polymorphisms were analyzed for association with the stabilizing methadone dose in 72 former severe heroin addicts with no major co-medications. There was significant difference in methadone doses required by subjects with different genotypes of the NGFB intronic single-nucleotide polymorphism rs2239622 (P=0.0002). These results may have clinical importance.


Asunto(s)
Dependencia de Heroína/tratamiento farmacológico , Metadona/uso terapéutico , Factor de Crecimiento Nervioso/genética , Adolescente , Adulto , Anciano , Femenino , Humanos , Masculino , Metadona/administración & dosificación , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple
2.
J Viral Hepat ; 19(1): 47-54, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21129131

RESUMEN

Despite a high prevalence of hepatitis C virus (HCV) among drug users, HCV evaluation and treatment acceptance are extremely low among these patients when referred from drug treatment facilities for HCV management. We sought to increase HCV treatment effectiveness among patients from a methadone maintenance treatment program (MMTP) by maintaining continuity of care. We developed, instituted and retrospectively assessed the effectiveness of an integrated, co-localized care model in which an internist-addiction medicine specialist from MMTP was embedded in the hepatitis clinic. Methadone maintenance treatment program patients were referred, evaluated by the internist and hepatologist in hepatitis clinic and provided HCV treatment with integration between both sites. Of 401 evaluated patients, anti-HCV antibody was detected in 257, 86% of whom were older than 40 years. Hepatitis C virus RNA levels were measured in 222 patients, 65 of whom were aviremic. Of 157 patients with detectable HCV RNA, 125 were eligible for referral to the hepatitis clinic, 76 (61%) of whom accepted and adhered with the referral. Men engaged in MMTP <36 months were significantly less likely to be seen in hepatitis clinic than men in MMTP more than 36 months (odds ratio = 7.7; 95% confidence interval 2.6-22.9) or women. We evaluated liver histology in 63 patients, and 83% had moderate to advanced liver disease. Twenty-four patients initiated treatment with 19 completing and 13 (54%) achieving sustained response. In conclusion, integrated care between the MMTP and the hepatitis clinic improves adherence with HCV evaluation and treatment compared to standard referral practices.


Asunto(s)
Hepatitis C/tratamiento farmacológico , Hepatitis C/epidemiología , Interferón-alfa/uso terapéutico , Metadona/administración & dosificación , Polietilenglicoles/uso terapéutico , Trastornos Relacionados con Sustancias/complicaciones , Adulto , Antivirales/uso terapéutico , Conducta Adictiva , Manejo de la Enfermedad , Femenino , Genotipo , Hepacivirus/efectos de los fármacos , Hepacivirus/genética , Hepacivirus/patogenicidad , Hepatitis C/complicaciones , Humanos , Hígado/patología , Hígado/virología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Cirrosis Hepática/virología , Masculino , Metadona/uso terapéutico , Persona de Mediana Edad , Tratamiento de Sustitución de Opiáceos , ARN Viral/sangre , Proteínas Recombinantes/uso terapéutico , Resultado del Tratamiento
3.
Artículo en Inglés | MEDLINE | ID: mdl-35832748

RESUMEN

Alcohol relapse is the treatment target for medications development for alcohol dependence. Aticaprant, a selective and short-acting kappa-opioid receptor (KOR) antagonist, has recently been under development for new clinical implications (depression or anhedonia). Recent studies have also found that aticaprant reduces alcohol intake and prevents stress- triggered alcohol seeking in rodents via a KOR-mediated mechanism. Here, we further investigated whether aticaprant alone or in combination with naltrexone (mu-opioid receptor [MOR] antagonist) altered alcohol relapse-like drinking using a mouse alcohol deprivation effect (ADE) paradigm to mimic the relapse episodes in human alcoholics. A long-acting and selective KOR antagonist nor-BNI was used as a reference compound for the effects of the KOR antagonism on the ADE. After 3-week intermittent-access alcohol drinking (two-bottle choice, 24-h access every other day), male and female mice displayed excessive alcohol intake and then pronounced ADE after 1-week abstinence. Aticaprant alone decreased alcohol ADE in a dose- dependent manner (1-3 mg/kg) in both males and females. Aticaprant at a lower dose (0.3 mg/kg) than the effective one (3 mg/kg) combined with a low dose of naltrexone (1 mg/kg) reduced the ADE in both sexes, and the combination was effective after a multi-dosing regimen (5 daily injections during the abstinence) without development of tolerance, suggesting synergistic effects of the combination. In contrast, nor-BNI alone or with naltrexone had no effect on the ADE in either sex. Our present study suggests that a combination of clinically developed, short-acting KOR antagonist aticaprant with low-dose naltrexone has therapeutic potential in alcohol "relapse" treatment.

4.
Pharmacogenomics J ; 10(3): 232-42, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20010914

RESUMEN

Alterations in expression of a cannabinoid receptor (CNR1, CB1), and of fatty acid amide hydrolase (FAAH) that degrades endogenous ligands of CB1, may contribute to the development of addiction. The 385C>A in the FAAH gene and six polymorphisms of CNR1 were genotyped in former heroin addicts and control subjects (247 Caucasians, 161 Hispanics, 179 African Americans and 19 Asians). In Caucasians, long repeats (>or=14) of 18087-18131(TAA)(8-17) were associated with heroin addiction (P=0.0102). Across three ethnicities combined, a highly significant association of long repeats with heroin addiction was found (z=3.322, P=0.0009). Point-wise significant associations of allele 1359A (P=0.006) and genotype 1359AA (P=0.034) with protection from heroin addiction were found in Caucasians. Also in Caucasians, the genotype pattern, 1359G>A and -6274A>T, was significantly associated with heroin addiction experiment wise (P=0.0244). No association of FAAH 385C>A with heroin addiction was found in any group studied.


Asunto(s)
Amidohidrolasas/genética , Dependencia de Heroína/genética , Polimorfismo Genético , Receptor Cannabinoide CB1/genética , Repeticiones de Trinucleótidos , Negro o Afroamericano/genética , Conducta Adictiva/genética , Femenino , Frecuencia de los Genes , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Población Blanca/genética
5.
Neuropharmacology ; 56 Suppl 1: 32-43, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18725235

RESUMEN

The focus of this review is primarily on recent developments in bidirectional translational research on the addictions, within the Laboratory of the Biology of Addictive Diseases at The Rockefeller University. This review is subdivided into major interacting aspects, including (a) Investigation of neurobiological and molecular adaptations (e.g., in genes for the opioid receptors or endogenous neuropeptides) in response to cocaine or opiates, administered under laboratory conditions modeling chronic patterns of human self-exposure (e.g., chronic escalating "binge"). (b) The impact of such drug exposure on the hypothalamic-pituitary-adrenal (HPA) axis and interacting neuropeptidergic systems (e.g., opioid, orexin and vasopressin). (c) Molecular genetic association studies using candidate gene and whole genome approaches, to define particular systems involved in vulnerability to develop specific addictions, and response to pharmacotherapy. (d) Neuroendocrine challenge studies in normal volunteers and current addictive disease patients along with former addicts in treatment, to investigate differential pharmacodynamics and responsiveness of molecular targets, in particular those also investigated in the experimental and molecular genetic approaches as described above.


Asunto(s)
Investigación Biomédica/métodos , Trastornos Relacionados con Sustancias , Animales , Investigación Biomédica/tendencias , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Narcóticos/uso terapéutico , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/fisiopatología , Receptores Opioides , Trastornos Relacionados con Sustancias/metabolismo , Trastornos Relacionados con Sustancias/fisiopatología , Trastornos Relacionados con Sustancias/terapia
6.
Neuroscience ; 158(2): 474-83, 2009 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18938225

RESUMEN

Mu opioid receptors (MOP-r) play an important role in the rewarding and locomotor stimulatory effects of heroin. The aim of the current study was to determine whether infusion of small interfering RNAs (siRNA) targeting MOP-r into the midbrain could knock down MOP-r mRNA and affect heroin-induced locomotor activity or heroin-induced conditioned place preference. Ten-week-old male C57BL/6J mice were surgically implanted bilaterally with guide cannulae directed between the substantia nigra and ventral tegmental area. After 4 days' recovery, mice were infused bilaterally with siRNAs that target the MOP-r (2 mMx0.75 microl/side/day for 3 days) or control siRNA. Seven days after the last infusion, a procedure for conditioned place preference was begun with four heroin (3 mg/kg i.p.) administration sessions alternating with four saline sessions. While heroin induced an increase in locomotor activity in all groups, siRNAs targeting specific regions of MOP-r significantly attenuated this effect. Of particular interest, mice infused with specific siRNAs targeting the MOP-r failed to develop and express conditioned place preference to heroin, or showed a significantly attenuated preference. These alterations in reward-related behaviors are likely due to the reduction in MOP-r mRNA and protein, shown in separate studies by in situ hybridization and autoradiography using the same MOP-r- siRNA infusions. Taken together, these studies demonstrate the utility of siRNA in the neurobiological study of specific components of the reward system and should contribute to the study of other complex behaviors.


Asunto(s)
Dependencia de Heroína/fisiopatología , Locomoción/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Receptores Opioides mu/metabolismo , Recompensa , Sustancia Negra/metabolismo , Área Tegmental Ventral/metabolismo , Análisis de Varianza , Animales , Autorradiografía , Heroína/efectos adversos , Dependencia de Heroína/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Tiempo de Reacción/efectos de los fármacos , Receptores Opioides mu/genética , Sustancia Negra/efectos de los fármacos , Factores de Tiempo , Área Tegmental Ventral/efectos de los fármacos
7.
Mol Psychiatry ; 13(4): 417-28, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18195715

RESUMEN

A genome-wide association study was conducted using microarray technology to identify genes that may be associated with the vulnerability to develop heroin addiction, using DNA from 104 individual former severe heroin addicts (meeting Federal criteria for methadone maintenance) and 101 individual control subjects, all Caucasian. Using separate analyses for autosomal and X chromosomal variants, we found that the strongest associations of allele frequency with heroin addiction were with the autosomal variants rs965972, located in the Unigene cluster Hs.147755 (experiment-wise q=0.053), and rs1986513 (q=0.187). The three variants exhibiting the strongest association with heroin addiction by genotype frequency were rs1714984, located in an intron of the gene for the transcription factor myocardin (P=0.000022), rs965972 (P=0.000080) and rs1867898 (P=0.000284). One genotype pattern (AG-TT-GG) was found to be significantly associated with developing heroin addiction (odds ratio (OR)=6.25) and explained 27% of the population attributable risk for heroin addiction in this cohort. Another genotype pattern (GG-CT-GG) of these variants was found to be significantly associated with protection from developing heroin addiction (OR=0.13), and lacking this genotype pattern explained 83% of the population attributable risk for developing heroin addiction. Evidence was found for involvement of five genes in heroin addiction, the genes coding for the mu opioid receptor, the metabotropic receptors mGluR6 and mGluR8, nuclear receptor NR4A2 and cryptochrome 1 (photolyase-like). This approach has identified several new genes potentially associated with heroin addiction and has confirmed the role of OPRM1 in this disease.


Asunto(s)
Predisposición Genética a la Enfermedad , Variación Genética , Proteínas Nucleares/genética , Receptores Opioides mu/genética , Transactivadores/genética , Cromosomas Humanos X , Criptocromos , Proteínas de Unión al ADN/genética , Femenino , Flavoproteínas/genética , Frecuencia de los Genes , Pruebas Genéticas , Genotipo , Dependencia de Heroína , Humanos , Desequilibrio de Ligamiento , Masculino , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Receptores de Glutamato Metabotrópico/genética , Factores de Transcripción/genética , Población Blanca
8.
Sci Adv ; 5(10): eaax9140, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31616793

RESUMEN

Opioid use disorders (OUDs) are diseases of the brain with behavioral, psychological, neurobiological, and medical manifestations. Vulnerability to OUDs can be affected by factors such as genetic background, environment, stress, and prolonged exposure to µ-opioid agonists for analgesia. Two standard-of-care maintenance medications, methadone and buprenorphine-naloxone, have a long-term positive influence on health of persons with opioid addiction. Buprenorphine and another medication, naltrexone, have also been approved for administration as monthly depot injections. However, neither medication is used as widely as needed, due largely to stigma, insufficient medical education or training, inadequate resources, and inadequate access to treatment. Ongoing directions in the field include (i) personalized approaches leveraging genetic factors for prediction of OUD vulnerability and prognosis, or for targeted pharmacotherapy, and (ii) development of novel analgesic medicines with new neurobiological targets with reduced abuse potential, reduced toxicity, and improved effectiveness, especially for chronic pain states other than cancer pain.


Asunto(s)
Investigación Biomédica , Trastornos Relacionados con Opioides/terapia , Analgésicos Opioides/química , Encéfalo/metabolismo , Humanos , Trastornos Relacionados con Opioides/epidemiología , Salud Pública , Receptores Opioides mu/metabolismo
9.
Neuroscience ; 153(4): 1225-34, 2008 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-18436386

RESUMEN

Recent evidence suggests an important role for hypothalamic orexins/hypocretins in modulation of drug reward and addiction-like behaviors in rodents. Our recent study has shown that the aversive state of arousal during acute morphine withdrawal is associated with increased orexin gene expression in lateral hypothalamus (LH) of Fischer 344 (F344) inbred rats, with no change in the expression of preprodynorphin (ppDyn), a gene co-expressed with LH orexin. Therefore, we determined whether orexin and ppDyn mRNA levels in LH or medial hypothalamus (including perifornical and dorsomedial areas) of F344 or Sprague-Dawley (SD) outbred rats, are altered following: 1) cocaine (10 mg/kg, i.p.) conditioned place preference (CPP); 2) chronic (14 days) cocaine exposure using both "binge" pattern administration in steady-dose (45 mg/kg/day) and escalating-dose (45-90 mg/kg/day) regimens; and 3) acute (1 day) and chronic (14 days) withdrawal from cocaine with opioid receptor antagonist naloxone treatment (1 mg/kg). We found that orexin mRNA levels were decreased after cocaine place conditioning in the LH of SD rats. A decreased LH orexin mRNA level was also observed after chronic escalating-dose cocaine (but not CPP pattern regimen without conditioning, or steady-dose regimen) in both strains. In F344 rats only, acute withdrawal from chronic escalating-dose cocaine administration resulted in increases in both LH orexin and ppDyn mRNA levels, which were unaltered by naloxone or after chronic withdrawal. Our results suggest that (1) alteration of LH orexin gene expression is region-specific after cocaine place conditioning in SD rats and dose-dependent after chronic exposure in both strains; and (2) increased LH orexin and ppDyn gene expressions in F344 rats may contribute to negative affective states in cocaine withdrawal.


Asunto(s)
Cocaína/administración & dosificación , Condicionamiento Operante/efectos de los fármacos , Dinorfinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuropéptidos/metabolismo , Precursores de Proteínas/metabolismo , Síndrome de Abstinencia a Sustancias/patología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Inhibidores de Captación de Dopamina/administración & dosificación , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Dinorfinas/genética , Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Neuropéptidos/genética , Orexinas , Precursores de Proteínas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/metabolismo , Factores de Tiempo
10.
Behav Neurosci ; 122(6): 1248-56, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19045944

RESUMEN

Methadone is widely used in treatment of short-acting opiate addiction. The on-off effects of opioids have been documented to have profound differences from steady-state opioids. The authors hypothesize that opioids play important roles in either generalized arousal (GA) or aversive state of arousal during opioid withdrawal. Both male and female C57BL6 mice received steady-state methadone (SSM) through osmotic pumps at 10 or 20 mg/kg/day, and GA was measured in voluntary motor activity, sensory responsivity, and contextual fear conditioning. SSM did not have any effect on those GA behaviors in either sex. Females had higher activity and less fear conditioning than males. The effects of SSM on stress-responsive orexin gene expression in the lateral hypothalamus (LH) and medial hypothalamus (MH, including perifornical and dorsomedial areas) were measured after the behavioral tests. Females showed significantly lower basal LH (but not MH) orexin mRNA levels than males. A panel of GA stressors increased LH orexin mRNA levels in females only; these increases were blunted by SSM at 20 mg/kg. In summary, SSM had no effect on GA behaviors. In females, SSM blunted the GA stress-induced LH orexin gene expression.


Asunto(s)
Nivel de Alerta/efectos de los fármacos , Metadona/administración & dosificación , Narcóticos/administración & dosificación , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Condicionamiento Psicológico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Miedo/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Hipotálamo Medio/efectos de los fármacos , Hipotálamo Medio/metabolismo , Bombas de Infusión Implantables , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Locomoción/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Neuropéptidos/genética , Neuropéptidos/metabolismo , Orexinas , Factores Sexuales
11.
J Neurosci ; 26(10): 2645-51, 2006 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-16525043

RESUMEN

The reinforcing effect of cocaine is associated with increases in dopamine in the striatum. The phosphoprotein DARPP-32 (dopamine- and cAMP-regulated phosphoprotein) has been shown to mediate the intracellular events after activation of dopamine receptors. DARPP-32 is phosphorylated at multiple sites by different protein kinases, but little is known about the functional role of these different sites. Cocaine self-administration and striatal levels of dopamine after acute "binge" cocaine administration were measured in separate lines of mice with alanine mutations introduced into DARPP-32 at either Thr34 (protein kinase A site, Thr34A), Thr75, (cyclin-dependent kinase 5 site, Thr75A), Ser97 (kinase CK2 site, Ser97A), or Ser130 (kinase CK1 site, Ser130A). Acquisition of stable cocaine self-administration required significantly more time in Thr34A-/- mice. Both Thr34A- and Ser130A-DARPP-32 mutant mice self-administered more cocaine than their respective wild-type controls. Also, cocaine-induced increases of dopamine in dorsal striatum were attenuated in the Thr34A- and Ser130A-DARPP-32 phosphomutant mice compared with wild-type mice. Notably, levels of P-Thr34- and P-Ser130-DARPP-32 were reduced after self-administration of cocaine in wild-type mice. Thus, phosphorylation states of Thr34- and Ser130-DARPP-32 play important roles in modulating the reinforcing effects of cocaine.


Asunto(s)
Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Refuerzo en Psicología , Autoadministración , Serina/metabolismo , Treonina/metabolismo , Alanina/genética , Alanina/metabolismo , Análisis de Varianza , Animales , Conducta Animal , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microdiálisis/métodos , Fosforilación , Esquema de Refuerzo , Factores de Tiempo
12.
Mol Cell Biol ; 15(4): 2080-9, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7891703

RESUMEN

We isolated the guinea pig preproenkephalin gene from a genomic library by hybridization to a rat cDNA probe. The entire nucleotide sequence of the gene was determined. Genomic Southern blot hybridization demonstrated that the gene exists in a single copy within the genome. On the basis of RNase protection transcript mapping and homology comparisons with known preproenkephalin sequences from other species and assuming a poly(A) tail length of 100 residues, we predicted an mRNA transcript of approximately 1,400 nucleotides encoded by three exons. Northern (RNA) blot analysis of total RNA from several brain regions showed high levels of preproenkephalin mRNA in the caudate putamen, nucleus accumbens, and hypothalamus, with detectable levels in the amygdala, ventral tegmental area, and central gray and also in the pituitary. Unexpectedly, in several brain regions, the mRNA appeared not only in the 1,400-nucleotide length but also in a shorter length of approximately 1,130 bases. Significant amounts of the shorter mRNA were found in the caudate putamen, nucleus accumbens, and amygdala. The longer, but not the shorter, transcripts from the caudate putamen were found to be polyadenylated, but the difference in size was not due solely to the presence of poly(A) tails. Northern gel analysis of total RNA from the caudate putamen with probes from each exon, together with RNase protection mapping of the 3' end of the mRNA demonstrated that the 1,400-base preproenkephalin mRNA transcripts are cleaved in a site-specific manner in some brain regions, yielding a 1,130-base transcript and a 165-base polyadenylated fragment derived from the terminal end of the 3' untranslated region of the mRNA. This cleavage may serve as a preliminary step in RNA degradation and provide a mechanism for control of preproenkephalin mRNA abundance through selective degradation.


Asunto(s)
Química Encefálica , Encefalinas/genética , Precursores de Proteínas/genética , ARN Mensajero/metabolismo , Secuencia de Aminoácidos , Amígdala del Cerebelo/química , Animales , Secuencia de Bases , Northern Blotting , Encefalinas/biosíntesis , Biblioteca Genómica , Cobayas , Datos de Secuencia Molecular , Núcleo Accumbens/química , Precursores de Proteínas/biosíntesis , Putamen/química , Procesamiento Postranscripcional del ARN , Análisis de Secuencia de ADN , Distribución Tisular , Transcripción Genética
13.
Genes Brain Behav ; 16(4): 449-461, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27870313

RESUMEN

Opioid receptor antagonist naltrexone reduces alcohol consumption and relapse in both humans and rodents. This study investigated whether hypothalamic proopiomelanocortin (POMC) neurons (producing beta-endorphin and melanocortins) play a role in alcohol drinking behaviors. Both male and female mice with targeted deletion of two neuronal Pomc enhancers nPE1 and nPE2 (nPE-/-), resulting in hypothalamic-specific POMC deficiency, were studied in short-access (4-h/day) drinking-in-the-dark (DID, alcohol in one bottle, intermittent access (IA, 24-h cycles of alcohol access every other day, alcohol vs. water in a two-bottle choice) and alcohol deprivation effect (ADE) models. Wild-type nPE+/+ exposed to 1-week DID rapidly established stable alcohol drinking behavior with more intake in females, whereas nPE-/- mice of both sexes had less intake and less preference. Although nPE-/- showed less saccharin intake and preference than nPE+/+, there was no genotype difference in sucrose intake or preference in the DID paradigm. After 3-week IA, nPE+/+ gradually escalated to high alcohol intake and preference, with more intake in females, whereas nPE-/- showed less escalation. Pharmacological blockade of mu-opioid receptors with naltrexone reduced intake in nPE+/+ in a dose-dependent manner, but had blunted effects in nPE-/- of both sexes. When alcohol was presented again after 1-week abstinence from IA, nPE+/+ of both sexes displayed significant increases in alcohol intake (ADE or relapse-like drinking), with more pronounced ADE in females, whereas nPE-/- did not show ADE in either sex. Our results suggest that neuronal POMC is involved in modulation of alcohol 'binge' drinking, escalation and 'relapse', probably via hypothalamic-mediated mechanisms, with sex differences.


Asunto(s)
Insuficiencia Suprarrenal/metabolismo , Consumo de Bebidas Alcohólicas/metabolismo , Obesidad/metabolismo , Proopiomelanocortina/deficiencia , Insuficiencia Suprarrenal/genética , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Consumo de Bebidas Alcohólicas/genética , Animales , Etanol/farmacología , Femenino , Genotipo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Noqueados , Naltrexona/farmacología , Obesidad/genética , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo
14.
J Psychopharmacol ; 20(5): 650-5, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16533868

RESUMEN

The aim of this study was to assess the reliability of salivary cortisol as a measure of hypothalmic-pituitary-adrenal (HPA) axis function in cocaine dependent individuals. Saliva and total plasma samples were collected from 49 abusers on 3 testing days in the morning, across eight time points per day. Significant associations between saliva and plasma cortisol were observed across all time points collapsed across 3 days in both men and women. These moderately significant correlations suggest that salivary measurements represent a stable, non-invasive and broad indicator of HPA axis functioning in cocaine dependent individuals.


Asunto(s)
Trastornos Relacionados con Cocaína/metabolismo , Hidrocortisona/análisis , Saliva/química , Hormona Adrenocorticotrópica/sangre , Adulto , Recolección de Muestras de Sangre , Trastornos Relacionados con Cocaína/diagnóstico , Femenino , Humanos , Hidrocortisona/sangre , Hidrocortisona/metabolismo , Sistema Hipotálamo-Hipofisario/fisiología , Masculino , Ciclo Menstrual/metabolismo , Persona de Mediana Edad , Escalas de Valoración Psiquiátrica , Reproducibilidad de los Resultados , Caracteres Sexuales , Manejo de Especímenes
15.
Cancer Res ; 56(6): 1352-60, 1996 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-8640825

RESUMEN

We report the first series of studies comparing the anti-edematous effects of human corticotropin-releasing factor (hCRF) and dexamethasone in an experimental model of vasogenic peritumoral brain edema. Both hCRF and dexamethasone effectively decreased blood-brain barrier (BBB) permeability of intracerebral RG2 gliomas in rats as observed by contrast-enhanced T(1)-weighted magnetic resonance imaging. A decrease in the water content of tumor and peritumoral brain tissue was observed with proton-density magnetic resonance imaging and confirmed by direct wet/dry tissue measurements. The calculated ED(50) for hCRF was 59 micrograms/kg s.c. twice a day, and that for dexamethasone was 0.61 mg/kg i.m. twice a day; the hCRF:dexamethasone dose-potency ratio was 120:1 on a molar basis. The anti-edematous action of hCRF is not mediated by the release of adrenal corticosteroids. A direct action of hCRF on the tumor microvasculature results in restoration of BBB integrity and up-regulation of BBB-specific protein expression. The average survival time with chronic treatment was prolonged significantly in the hCRF-treated group (35 days) compared with the dexamethasone-treated group (28 days; P < 0.05) and the saline-treated control group (22 days; P < 0.0001). hCRF, as an alternative to corticosteroid therapy, may provide substantial benefits with respect to reducing the major side effects encountered with long-term, high-dose corticosteroid treatment.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/tratamiento farmacológico , Neoplasias Encefálicas/complicaciones , Hormona Liberadora de Corticotropina/uso terapéutico , Glioma/complicaciones , Adrenalectomía , Animales , Antiinflamatorios/farmacología , Antígenos/análisis , Agua Corporal/efectos de los fármacos , Edema Encefálico/sangre , Edema Encefálico/etiología , Edema Encefálico/mortalidad , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/mortalidad , Permeabilidad Capilar/efectos de los fármacos , Corticosterona/sangre , Hormona Liberadora de Corticotropina/farmacología , Dexametasona/farmacología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/inmunología , Glioma/sangre , Glioma/tratamiento farmacológico , Glioma/mortalidad , Imagen por Resonancia Magnética , Ratas , Ratas Endogámicas F344
16.
Neuroscience ; 134(4): 1391-7, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16039786

RESUMEN

In humans, stress is recognized as a major factor contributing to relapse to drug abuse in abstinent individuals; drugs of abuse themselves or withdrawal from such drugs act as stressors. In the animals, evidence suggests that centrally released arginine vasopressin in both amygdala and hypothalamus plays an important role in stress-related anxiogenic behaviors. The stress responsive hypothalamic-pituitary-adrenal axis is under tonic inhibition via endogenous opioids, and cocaine withdrawal stimulates hypothalamic-pituitary-adrenal activity. The present studies were undertaken to determine whether: (1) 14-day (chronic) "binge" pattern cocaine administration (45 mg/kg/day) or its withdrawal for 3 h (acute), 1 day (subacute) or 10 days (chronic) alters arginine vasopressin mRNA levels in amygdala or hypothalamus; (2) the opioid receptor antagonist naloxone (1mg/kg) alters arginine vasopressin mRNA or hypothalamic-pituitary-adrenal hormonal responses in acute cocaine withdrawal; and (3) there are associated changes of mu opioid receptor or proopiomelanocortin mRNA levels. In amygdala, arginine vasopressin mRNA levels were unchanged after chronic "binge" cocaine, but were increased during acute cocaine withdrawal. Naloxone completely blocked this increase. Neither chronic cocaine nor its acute withdrawal altered amygdalar mu opioid receptor mRNA levels. The increase in amygdalar arginine vasopressin mRNA levels was still observed after subacute withdrawal, but not after chronic withdrawal. Although hypothalamic-pituitary-adrenal tolerance developed with chronic "binge" cocaine, there were modestly elevated plasma adrenocorticotropin hormone levels during acute withdrawal. While naloxone produced modest adrenocorticotropin hormone elevations in cocaine-naïve rats, naloxone failed to elicit an adrenocorticotropin hormone response in cocaine-withdrawn rats. In hypothalamus, neither chronic cocaine nor acute withdrawal altered arginine vasopressin, proopiomelanocortin or mu opioid receptor mRNA levels. These results show that: (1) opioid receptors mediate increased amygdalar arginine vasopressin gene expression during acute cocaine withdrawal, and (2) cocaine withdrawal renders the hypothalamic-pituitary-adrenal axis insensitive to naloxone. Our findings suggest a potential role for amygdalar arginine vasopressin in the aversive consequences of early cocaine withdrawal.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Arginina Vasopresina/efectos de los fármacos , Arginina Vasopresina/metabolismo , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Síndrome de Abstinencia a Sustancias/fisiopatología , Animales , Arginina Vasopresina/genética , Expresión Génica , Sistema Hipotálamo-Hipofisario/fisiología , Hipotálamo/metabolismo , Masculino , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Sistema Hipófiso-Suprarrenal/fisiología , Proopiomelanocortina/efectos de los fármacos , Proopiomelanocortina/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Endogámicas F344 , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/metabolismo
17.
Neuroscience ; 289: 63-70, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25595971

RESUMEN

Recent research suggests an involvement of pro-opiomelanocortin (POMC) gene products (e.g., beta-endorphin) in modulating cocaine-induced reward and addiction-like behaviors in rodents. In this study, we investigated whether chronic "binge" cocaine and its withdrawal altered POMC gene expression in the brain of rats. Male Fischer rats were treated with two different chronic (14-day) "binge" pattern cocaine administration regimens (three injections at 1-h intervals, i.p.): steady-dose (45mg/kg/day) and escalating-dose (90mg/kg on the last day). Although there was no POMC mRNA alteration after chronic steady-dose cocaine, a significant decrease in POMC mRNA levels in the hypothalamus was found after chronic escalating-dose cocaine. In contrast, after acute (1-day) withdrawal from chronic "binge" escalating-dose regimen, but not steady-dose regimen, there were increased hypothalamic POMC mRNA levels that persisted into 14days of protracted withdrawal. To study the role of the endogenous opioid systems in the cocaine withdrawal effects, we administered a single naloxone injection (1mg/kg) that caused elevated POMC mRNA levels observed 24h later in cocaine naïve rats, but it did not lead to further increases in cocaine-withdrawn rats. Our results suggest that during withdrawal from chronic "binge" escalating-dose cocaine: (1) there was a persistent increase in hypothalamic POMC gene expression; and (2) hyposensitivity of the POMC gene expression to naloxone indicates altered opioidergic tone at or above the hypothalamic level.


Asunto(s)
Trastornos Relacionados con Cocaína/metabolismo , Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Hipotálamo/metabolismo , Proopiomelanocortina/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Corticosterona/sangre , Hipotálamo/efectos de los fármacos , Masculino , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , ARN Mensajero/metabolismo , Ratas Endogámicas F344 , Esquema de Refuerzo
18.
Neuroscience ; 285: 34-46, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25446355

RESUMEN

Abuse and addiction to prescription opioids such as oxycodone (a short-acting Mu opioid receptor (MOP-r) agonist) in adolescence is a pressing public health issue. We have previously shown differences in oxycodone self-administration behaviors between adolescent and adult C57BL/6J mice and expression of striatal neurotransmitter receptor genes, in areas involved in reward. In this study, we aimed to determine whether oxycodone self-administration differentially affects genes regulating synaptic plasticity in the hippocampus of adolescent compared to adult mice, since the hippocampus may be involved in learning aspects associated with chronic drug self administration. Hippocampus was isolated for mRNA analysis from mice that had self administered oxycodone (0.25 mg/kg/infusion) 2h/day for 14 consecutive days or from yoked saline controls. Gene expression was analyzed with real-time polymerase chain reaction (PCR) using a commercially available "synaptic plasticity" PCR array containing 84 genes. We found that adolescent and adult control mice significantly differed in the expression of several genes in the absence of oxycodone exposure, including those coding for mitogen-activated protein kinase, calcium/calmodulin-dependent protein kinase II gamma subunit, glutamate receptor, ionotropic AMPA2 and metabotropic 5. Chronic oxycodone self administration increased proviral integration site 1 (Pim1) and thymoma viral proto-oncogene 1 mRNA levels compared to controls in both age groups. Both Pim1 and cadherin 2 mRNAs showed a significant combined effect of Drug Condition and Age × Drug Condition. Furthermore, the mRNA levels of both cadherin 2 and cAMP response element modulators showed an experiment-wise significant difference between oxycodone and saline control in adult but not in adolescent mice. Overall, this study demonstrates for the first time that chronic oxycodone self-administration differentially alters synaptic plasticity gene expression in the hippocampus of adolescent and adult mice.


Asunto(s)
Hipocampo/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Narcóticos/administración & dosificación , Trastornos Relacionados con Opioides/metabolismo , Oxicodona/administración & dosificación , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Animales , Cadherinas/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Modulador del Elemento de Respuesta al AMP Cíclico/metabolismo , Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , ARN Mensajero/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores AMPA/metabolismo , Autoadministración
19.
Biol Psychiatry ; 32(6): 523-8, 1992 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-1445968

RESUMEN

Beta endorphin (BE) is a polypeptide agonist for the brain's endogenous opioid system. Levels of BE are elevated by opioid antagonists such as naloxone and depressed by short-acting agonists such as heroin and morphine; they become normalized during steady-state methadone. Buprenorphine (BUP) is a partial opioid agonist whose effects on BE levels were examined in six former heroin addicts and 14 methadone-maintained patients before and after being switched to sublingual BUP 2 mg daily for 1 month. In six former methadone-treated subjects BE levels also were measured after stopping BUP and after naloxone challenge. Levels of BE were not significantly lower in subjects started on BUP after stopping heroin (n = 6) (8.0 versus 8.1 ng/ml) or in subjects started on BUP after stopping methadone (n = 14) (11.6 vs 15.6 ng/ml). However, BE levels were lower on BUP than after naloxone challenge (n = 6) (7.0 versus 34.9 ng/ml). Levels of BE did not significantly change between the first 2 weeks ("early") and "later," although BE levels on methadone significantly correlated with BE levels on BUP in the "early" but not the "later" phase. The BE levels on BUP also did not differ from BE levels of unmedicated normals.


Asunto(s)
Buprenorfina/uso terapéutico , Heroína/sangre , Metadona/sangre , Naloxona/sangre , Trastornos Relacionados con Sustancias/metabolismo , betaendorfina/análisis , Adulto , Buprenorfina/sangre , Femenino , Humanos , Masculino , Metadona/uso terapéutico , Persona de Mediana Edad , Radioinmunoensayo , Síndrome de Abstinencia a Sustancias , Trastornos Relacionados con Sustancias/rehabilitación , betaendorfina/sangre
20.
Biol Psychiatry ; 18(9): 1007-21, 1983 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-6685534

RESUMEN

Narcotic withdrawal is often accompanied by an atypical depression with responds to resumption of narcotics. We hypothesized that methadone might exert its antidepressant effects through monoamine oxidase (MAO) inhibition. The current study examined 3H-methadone distribution in rat brain and effects on regional MAO activity with acute doses (2.5 mg/kg) which approximate those found during chronic methadone maintenance in man. Limbic areas (amygdala, basomedial hypothalamus, caudate-putamen, hippocampus, preoptic nucleus), as well as pituitary and liver were assayed for MAO activity and methadone concentration. MAO activities did not differ significantly in acute methadone or saline-treated cage-mates at 1 or 24 hr. The concentrations of methadone at 1 hr ranged between 17 and 223 ng/100 mg wet wt tissue in the preoptic nucleus and pituitary, respectively. No significant correlation was found between change in MAO activity (MAO methadone/MAO saline) and methadone concentration in any region at 1 or 24 hr. This study does not support the hypothesis that methadone acts as an antidepressant through MAO inhibition, at least not following acute administration of this exogenous opioid.


Asunto(s)
Metadona/farmacología , Monoaminooxidasa/metabolismo , Animales , Depresión/inducido químicamente , Humanos , Hipotálamo/enzimología , Sistema Límbico/enzimología , Hígado/enzimología , Masculino , Metadona/efectos adversos , Inhibidores de la Monoaminooxidasa/farmacología , Hipófisis/enzimología , Ratas , Ratas Endogámicas , Síndrome de Abstinencia a Sustancias/fisiopatología , Tritio
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda