Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cereb Cortex ; 25(1): 97-108, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23960211

RESUMEN

Lateralization of the processing of positive and negative emotions and pain suggests an asymmetric distribution of the neurotransmitter systems regulating these functions between the left and right brain hemispheres. By virtue of their ability to selectively mediate euphoria, dysphoria, and pain, the µ-, δ-, and κ-opioid receptors and their endogenous ligands may subserve these lateralized functions. We addressed this hypothesis by comparing the levels of the opioid receptors and peptides in the left and right anterior cingulate cortex (ACC), a key area for emotion and pain processing. Opioid mRNAs and peptides and 5 "classical" neurotransmitters were analyzed in postmortem tissues from 20 human subjects. Leu-enkephalin-Arg (LER) and Met-enkephalin-Arg-Phe, preferential δ-/µ- and κ-/µ-opioid agonists, demonstrated marked lateralization to the left and right ACC, respectively. Dynorphin B (Dyn B) strongly correlated with LER in the left, but not in the right ACC suggesting different mechanisms of the conversion of this κ-opioid agonist to δ-/µ-opioid ligand in the 2 hemispheres; in the right ACC, Dyn B may be cleaved by PACE4, a proprotein convertase regulating left-right asymmetry formation. These findings suggest that region-specific lateralization of neuronal networks expressing opioid peptides underlies in part lateralization of higher functions, including positive and negative emotions and pain in the human brain.


Asunto(s)
Emociones/fisiología , Lateralidad Funcional/fisiología , Giro del Cíngulo/metabolismo , Péptidos Opioides/metabolismo , Dolor/metabolismo , Adulto , Anciano , Encéfalo/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/metabolismo , Adulto Joven
2.
Addict Biol ; 18(1): 161-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21955155

RESUMEN

The endogenous opioid system (EOS) plays a critical role in addictive processes. Molecular dysregulations in this system may be specific for different stages of addiction cycle and neurocircuitries involved and therefore may differentially contribute to the initiation and maintenance of addiction. Here we evaluated whether the EOS is altered in brain areas involved in cognitive control of addiction including the dorsolateral prefrontal cortex (dl-PFC), orbitofrontal cortex (OFC) and hippocampus in human alcohol-dependent subjects. Levels of EOS mRNAs were measured by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and levels of dynorphins by radioimmunoassay (RIA) in post-mortem specimens obtained from 14 alcoholics and 14 controls. Prodynorphin mRNA and dynorphins in dl-PFC, κ-opioid receptor mRNA in OFC and dynorphins in hippocampus were up-regulated in alcoholics. No significant changes in expression of proenkephalin, and µ- and δ-opioid receptors were evident; pro-opiomelanocortin mRNA levels were below the detection limit. Activation of the κ-opioid receptor by up-regulated dynorphins in alcoholics may underlie in part neurocognitive dysfunctions relevant for addiction and disrupted inhibitory control.


Asunto(s)
Alcoholismo/metabolismo , Conducta Adictiva/metabolismo , Péptidos Opioides/metabolismo , Corteza Prefrontal/metabolismo , ARN Mensajero/metabolismo , Receptores Opioides/metabolismo , Adaptación Fisiológica/genética , Adulto , Alcoholismo/genética , Alcoholismo/fisiopatología , Análisis de Varianza , Animales , Conducta Adictiva/genética , Conducta Adictiva/fisiopatología , Estudios de Casos y Controles , Dinorfinas/genética , Dinorfinas/metabolismo , Encefalinas/genética , Encefalinas/metabolismo , Hipocampo/metabolismo , Humanos , Masculino , Péptidos Opioides/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , ARN Mensajero/genética , Radioinmunoensayo/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Recompensa , Estadísticas no Paramétricas , Regulación hacia Arriba/fisiología
3.
J Neurotrauma ; 29(9): 1785-93, 2012 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-22468884

RESUMEN

Traumatic brain injury (TBI) induces a cascade of primary and secondary events resulting in impairment of neuronal networks that eventually determines clinical outcome. The dynorphins, endogenous opioid peptides, have been implicated in secondary injury and neurodegeneration in rodent and human brain. To gain insight into the role of dynorphins in the brain's response to trauma, we analyzed short-term (1-day) and long-term (7-day) changes in dynorphin A (Dyn A) levels in the frontal cortex, hippocampus, and striatum, induced by unilateral left-side or right-side cortical TBI in mice. The effects of TBI were significantly different from those of sham surgery (Sham), while the sham surgery also produced noticeable effects. Both sham and TBI induced short-term changes and long-term changes in all three regions. Two types of responses were generally observed. In the hippocampus, Dyn A levels were predominantly altered ipsilateral to the injury. In the striatum and frontal cortex, injury to the right (R) hemisphere affected Dyn A levels to a greater extent than that seen in the left (L) hemisphere. The R-TBI but not L-TBI produced Dyn A changes in the striatum and frontal cortex at 7 days after injury. Effects of the R-side injury were similar in the two hemispheres. In naive animals, Dyn A was symmetrically distributed between the two hemispheres. Thus, trauma may reveal a lateralization in the mechanism mediating the response of Dyn A-expressing neuronal networks in the brain. These networks may differentially mediate effects of left and right brain injury on lateralized brain functions.


Asunto(s)
Lesiones Encefálicas/metabolismo , Dinorfinas/metabolismo , Análisis de Varianza , Animales , Cuerpo Estriado/metabolismo , Lateralidad Funcional/fisiología , Hipocampo/metabolismo , Masculino , Ratones , Corteza Prefrontal/metabolismo , Radioinmunoensayo
4.
Am J Psychiatry ; 168(10): 1090-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21828288

RESUMEN

OBJECTIVE: Circadian and stress-response systems mediate environmental changes that affect alcohol drinking. Psychosocial stress is an environmental risk factor for alcohol abuse. Circadian rhythm gene period 1 (Per1) is targeted by stress hormones and is transcriptionally activated in corticotropin releasing factor-expressing cells. The authors hypothesized that Per1 is involved in integrating stress response and circadian rhythmicity and explored its relevance to alcohol drinking. METHOD: In mice, the effects of stress on ethanol intake in mPer1-mutant and wild-type mice were assessed. In humans, single nucleotide polymorphisms (SNPs) in hPer1 were tested for association with alcohol drinking behavior in 273 adolescents and an adult case-control sample of 1,006 alcohol-dependent patients and 1,178 comparison subjects. In vitro experiments were conducted to measure genotype-specific expression and transcription factor binding to hPer1. RESULTS: The mPer1-mutant mice showed enhanced alcohol consumption in response to social defeat stress relative to their wild-type littermates. An association with the frequency of heavy drinking in adolescents with the hPer1 promoter SNP rs3027172 and with psychosocial adversity was found. There was significant interaction between the rs3027172 genotype and psychosocial adversity on this drinking measure. In a confirmatory analysis, association of hPer1 rs3027172 with alcohol dependence was shown. Cortisol-induced transcriptional activation of hPer1 was reduced in human B-lymphoblastoid cells carrying the risk genotype of rs3027172. Binding affinity of the transcription factor Snail1 to the risk allele of the hPer1 SNP rs3027172 was also reduced. CONCLUSIONS: The findings indicate that the hPer1 gene regulates alcohol drinking behavior during stressful conditions and provide evidence for underlying neurobiological mechanisms.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Proteínas Circadianas Period/genética , Estrés Psicológico/genética , Adolescente , Adulto , Consumo de Bebidas Alcohólicas/psicología , Alelos , Animales , Estudios de Casos y Controles , Femenino , Genotipo , Humanos , Masculino , Ratones , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Estrés Psicológico/complicaciones
5.
Addict Biol ; 16(3): 499-509, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21521424

RESUMEN

The genetic, epigenetic and environmental factors may influence the risk for neuropsychiatric disease through their effects on gene transcription. Mechanistically, these effects may be integrated through regulation of methylation of CpG dinucleotides overlapping with single-nucleotide polymorphisms (SNPs) associated with a disorder. We addressed this hypothesis by analyzing methylation of prodynorphin (PDYN) CpG-SNPs associated with alcohol dependence, in human alcoholics. Postmortem specimens of the dorsolateral prefrontal cortex (dl-PFC) involved in cognitive control of addictive behavior were obtained from 14 alcohol-dependent and 14 control subjects. Methylation was measured by pyrosequencing after bisulfite treatment of DNA. DNA binding proteins were analyzed by electromobility shift assay. Three PDYN CpG-SNPs associated with alcoholism were found to be differently methylated in the human brain. In the dl-PFC of alcoholics, methylation levels of the C, non-risk variant of 3'-untranslated region (3'-UTR) SNP (rs2235749; C > T) were increased, and positively correlated with dynorphins. A DNA-binding factor that differentially targeted the T, risk allele and methylated and unmethylated C allele of this SNP was identified in the brain. The findings suggest a causal link between alcoholism-associated PDYN 3'-UTR CpG-SNP methylation, activation of PDYN transcription and vulnerability of individuals with the C, non-risk allele(s) to develop alcohol dependence.


Asunto(s)
Alcoholismo/genética , Islas de CpG/genética , Metilación de ADN/genética , Encefalinas/genética , Polimorfismo de Nucleótido Simple/genética , Corteza Prefrontal/metabolismo , Precursores de Proteínas/genética , Regiones no Traducidas 3'/genética , Adulto , Anciano , Alcoholismo/patología , Alelos , Epigenómica , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Masculino
6.
Brain Res ; 1385: 18-25, 2011 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-21338584

RESUMEN

Single nucleotide polymorphism (rs1997794) in promoter of the prodynorphin gene (PDYN) associated with alcohol-dependence may impact PDYN transcription in human brain. To address this hypothesis we analyzed PDYN mRNA levels in the dorsolateral prefrontal cortex (dl-PFC) and hippocampus, both involved in cognitive control of addictive behavior and PDYN promoter SNP genotype in alcohol-dependent and control human subjects. The principal component analysis suggested that PDYN expression in the dl-PFC may be related to alcoholism, while in the hippocampus may depend on the genotype. We also demonstrated that the T, low risk SNP allele resides within noncanonical AP-1-binding element that may be targeted by JUND and FOSB proteins, the dominant AP-1 constituents in the human brain. The T to C transition abrogated AP-1 binding. The impact of genetic variations on PDYN transcription may be relevant for diverse adaptive responses of this gene to alcohol.


Asunto(s)
Alcoholismo/genética , Alcoholismo/metabolismo , Encefalinas/genética , Hipocampo/metabolismo , Polimorfismo de Nucleótido Simple/genética , Corteza Prefrontal/metabolismo , Regiones Promotoras Genéticas/genética , Precursores de Proteínas/genética , Factor de Transcripción AP-1/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Alcoholismo/patología , Sitios de Unión/genética , Encefalinas/biosíntesis , Regulación de la Expresión Génica , Genotipo , Células HeLa , Hipocampo/patología , Humanos , Masculino , Persona de Mediana Edad , Corteza Prefrontal/patología , Precursores de Proteínas/biosíntesis , Factor de Transcripción AP-1/biosíntesis , Factor de Transcripción AP-1/genética
7.
Brain Behav Immun ; 25 Suppl 1: S29-38, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21195164

RESUMEN

Alcohol dependence and associated cognitive impairment appear to result from maladaptive neuroplasticity in response to chronic alcohol consumption, neuroinflammation and neurodegeneration. The inherent stability of behavioral alterations associated with the addicted state suggests that transcriptional and epigenetic mechanisms are operative. NF-κB transcription factors are regulators of synaptic plasticity and inflammation, and responsive to a variety of stimuli including alcohol. These factors are abundant in the brain where they have diverse functions that depend on the composition of the NF-κB complex and cellular context. In neuron cell bodies, NF-κB is constitutively active, and involved in neuronal injury and neuroprotection. However, at the synapse, NF-κB is present in a latent form and upon activation is transported to the cell nucleus. In glia, NF-κB is inducible and regulates inflammatory processes that exacerbate alcohol-induced neurodegeneration. Animal studies demonstrate that acute alcohol exposure transiently activates NF-κB, which induces neuroinflammatory responses and neurodegeneration. Postmortem studies of brains of human alcoholics suggest that repeated cycles of alcohol consumption and withdrawal cause adaptive changes in the NF-κB system that may permit the system to better tolerate excessive stimulation. This type of tolerance, ensuring a low degree of responsiveness to applied stimuli, apparently differs from that in the immune system, and may represent a compensatory response that protects brain cells against alcohol neurotoxicity. This view is supported by findings showing preferential downregulation of pro-apoptotic gene expression in the affected brain areas in human alcoholics. Although further verification is needed, we speculate that NF-κB-driven neuroinflammation and disruption to neuroplasticity play a significant role in regulating alcohol dependence and cognitive impairment.


Asunto(s)
Alcoholismo/genética , Encéfalo/metabolismo , FN-kappa B/genética , Neuronas/metabolismo , Alcoholismo/metabolismo , Humanos , FN-kappa B/metabolismo , Transducción de Señal/genética
8.
Am J Hum Genet ; 87(5): 593-603, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-21035104

RESUMEN

Spinocerebellar ataxias (SCAs) are dominantly inherited neurodegenerative disorders characterized by progressive cerebellar ataxia and dysarthria. We have identified missense mutations in prodynorphin (PDYN) that cause SCA23 in four Dutch families displaying progressive gait and limb ataxia. PDYN is the precursor protein for the opioid neuropeptides, α-neoendorphin, and dynorphins A and B (Dyn A and B). Dynorphins regulate pain processing and modulate the rewarding effects of addictive substances. Three mutations were located in Dyn A, a peptide with both opioid activities and nonopioid neurodegenerative actions. Two of these mutations resulted in excessive generation of Dyn A in a cellular model system. In addition, two of the mutant Dyn A peptides induced toxicity above that of wild-type Dyn A in cultured striatal neurons. The fourth mutation was located in the nonopioid PDYN domain and was associated with altered expression of components of the opioid and glutamate system, as evident from analysis of SCA23 autopsy tissue. Thus, alterations in Dyn A activities and/or impairment of secretory pathways by mutant PDYN may lead to glutamate neurotoxicity, which underlies Purkinje cell degeneration and ataxia. PDYN mutations are identified in a small subset of ataxia families, indicating that SCA23 is an infrequent SCA type (∼0.5%) in the Netherlands and suggesting further genetic SCA heterogeneity.


Asunto(s)
Encefalinas/genética , Mutación Missense , Precursores de Proteínas/genética , Degeneraciones Espinocerebelosas/genética , Cerebelo/química , Cerebelo/citología , Dinorfinas/análisis , Encefalinas/análisis , Femenino , Proteínas de Transporte de Glutamato en la Membrana Plasmática/análisis , Humanos , Masculino , Linaje , Precursores de Proteínas/análisis , Células de Purkinje/química
9.
Addict Biol ; 14(3): 294-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19523044

RESUMEN

The transcription factor DeltaFosB is accumulated in the addiction circuitry, including the orbitofrontal and medial prefrontal cortices of rodents chronically exposed to ethanol or other drugs of abuse, and has been suggested to play a direct role in addiction maintenance. To address this hypothesis in the context of substance dependence in humans, we compared the immunoreactivities of FOSB proteins in the orbitofrontal and dorsolateral prefrontal cortices (OFC and DLPFC respectively) between controls and alcoholics using semiquantitative immunoblotting. In both structures, we detected three forms of FOSB, one of which was DeltaFOSB, but in neither case did their immunoreactivities differ between the groups. Our results indicate that the DeltaFOSB immunoreactivity in the human brain is very low, and that it is not accumulated in the OFC and DLPFC of human alcoholics, suggesting that it may not be directly involved in addiction maintenance, at least not in ethanol dependence.


Asunto(s)
Alcoholismo/patología , Lóbulo Frontal/patología , Corteza Prefrontal/patología , Proteínas Proto-Oncogénicas c-fos/análisis , Células HeLa , Humanos , Immunoblotting , Peso Molecular , Red Nerviosa/patología , Valores de Referencia
10.
Int J Neuropsychopharmacol ; 12(1): 109-15, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18937880

RESUMEN

In human alcoholics, the cell density is decreased in the prefrontal cortex (PFC) and other brain areas. This may be due to persistent activation of cell death pathways. To address this hypothesis, we examined the status of cell death machinery in the dorsolateral PFC in alcoholics. Protein and mRNA expression levels of several key pro- and anti-apoptotic genes were compared in post-mortem samples of 14 male human alcoholics and 14 male controls. The findings do not support the hypothesis. On the contrary, they show that several components of intrinsic apoptotic pathway are decreased in alcoholics. No differences were evident in the motor cortex, which is less damaged in alcoholics and was analysed for comparison. Thus, cell death mechanisms may be dysregulated by inhibition of intrinsic apoptotic pathway in the PFC in human alcoholics. This inhibition may reflect molecular adaptations that counteract alcohol neurotoxicity in cells that survive after many years of alcohol exposure and withdrawal.


Asunto(s)
Alcoholismo/patología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Corteza Prefrontal/patología , Adulto , Anciano , Anciano de 80 o más Años , Alcoholismo/genética , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Western Blotting , Muerte Celular/genética , Humanos , Masculino , Persona de Mediana Edad , Corteza Motora/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Synapse ; 62(11): 829-33, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18720419

RESUMEN

Convergent lines of evidence suggest potentiation of glutamatergic synapses after chronic ethanol exposure, and indicate that the presynaptic effect hereof is on modulators of synaptic strength rather than on executors of glutamate release. To address this hypothesis in the context of ethanol dependence in humans, we used semiquantitative immunoblotting to compare the immunoreactivities of synaptophysin I, syntaxin 1A, synaptosome-associated protein 25, and vesicle-associated membrane protein in the prefrontal and motor cortices between chronic alcoholics and control subjects. We found a region-specific elevation in synaptophysin I immunoreactivity in the prefrontal cortex of alcoholics, but detected no significant differences between the groups in the immunoreactivities of the other three proteins. Our findings are consistent with an effect of repeated ethanol exposure on modulators of synaptic strength but not on executors of glutamate release, and suggest a role for synaptophysin I in the enduring neuroplasticity in the prefrontal cortical glutamate circuitry that is associated with ethanol dependence.


Asunto(s)
Alcoholismo/metabolismo , Glicoproteínas de Membrana/metabolismo , Corteza Prefrontal/metabolismo , Sinaptofisina/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Alcoholismo/fisiopatología , Ácido Glutámico/metabolismo , Humanos , Masculino , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/fisiología , Persona de Mediana Edad , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Corteza Prefrontal/química , Sinaptofisina/biosíntesis , Sinaptofisina/fisiología
12.
PLoS One ; 2(9): e930, 2007 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-17895971

RESUMEN

BACKGROUND: Alcohol dependence and associated cognitive impairments apparently result from neuroadaptations to chronic alcohol consumption involving changes in expression of multiple genes. Here we investigated whether transcription factors of Nuclear Factor-kappaB (NF-kappaB) family, controlling neuronal plasticity and neurodegeneration, are involved in these adaptations in human chronic alcoholics. METHODS AND FINDINGS: Analysis of DNA-binding of NF-kappaB (p65/p50 heterodimer) and the p50 homodimer as well as NF-kappaB proteins and mRNAs was performed in postmortem human brain samples from 15 chronic alcoholics and 15 control subjects. The prefrontal cortex involved in alcohol dependence and cognition was analyzed and the motor cortex was studied for comparison. The p50 homodimer was identified as dominant kappaB binding factor in analyzed tissues. NF-kappaB and p50 homodimer DNA-binding was downregulated, levels of p65 (RELA) mRNA were attenuated, and the stoichiometry of p65/p50 proteins and respective mRNAs was altered in the prefrontal cortex of alcoholics. Comparison of a number of p50 homodimer/NF-kappaB target DNA sites, kappaB elements in 479 genes, down- or upregulated in alcoholics demonstrated that genes with kappaB elements were generally upregulated in alcoholics. No significant differences between alcoholics and controls were observed in the motor cortex. CONCLUSIONS: We suggest that cycles of alcohol intoxication/withdrawal, which may initially activate NF-kappaB, when repeated over years downregulate RELA expression and NF-kappaB and p50 homodimer DNA-binding. Downregulation of the dominant p50 homodimer, a potent inhibitor of gene transcription apparently resulted in derepression of kappaB regulated genes. Alterations in expression of p50 homodimer/NF-kappaB regulated genes may contribute to neuroplastic adaptation underlying alcoholism.


Asunto(s)
Alcoholismo/patología , Encéfalo/patología , FN-kappa B/genética , Adaptación Fisiológica/genética , Adulto , Anciano , Anciano de 80 o más Años , Alcoholismo/genética , Alcoholismo/metabolismo , Western Blotting , Encéfalo/metabolismo , Enfermedad Crónica , ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Masculino , Persona de Mediana Edad , FN-kappa B/metabolismo , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/metabolismo
13.
J Neurochem ; 97(1): 292-301, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16515546

RESUMEN

The diversity of peptide ligands for a particular receptor may provide a greater dynamic range of functional responses, while maintaining selectivity in receptor activation. Dynorphin A (Dyn A), and dynorphin B (Dyn B) are endogenous opioid peptides that activate the kappa-opioid receptor (KOR). Here, we characterized interactions of big dynorphin (Big Dyn), a 32-amino acid prodynorphin-derived peptide consisting of Dyn A and Dyn B, with human KOR, mu- (hMOR) and delta- (hDOR) opioid receptors and opioid receptor-like receptor 1 (hORL1) expressed in cells transfected with respective cDNA. Big Dyn and Dyn A demonstrated roughly similar affinity for binding to hKOR that was higher than that of Dyn B. Dyn A was more selective for hKOR over hMOR, hDOR and hORL1 than Big Dyn, while Dyn B demonstrated low selectivity. In contrast, Big Dyn activated G proteins through KOR with much greater potency, efficacy and selectivity than other dynorphins. There was no correlation between the rank order of the potency for the KOR-mediated activation of G proteins and the binding affinity of dynorphins for KOR. The rank of the selectivity for the activation of G proteins through hKOR and of the binding to this receptor also differed. Immunoreactive Big Dyn was detected using the combination of radioimmunoassay (RIA) and HPLC in the human nucleus accumbens, caudate nucleus, hippocampus and cerebrospinal fluid (CSF) with the ratio of Big Dyn and Dyn B being approximately 1:3. The presence in the brain implies that Big Dyn, along with other dynorphins, is processed from prodynorphin and secreted from neurons. Collectively, the high potency and efficacy and the relative abundance suggest that Big Dyn may play a role in the KOR-mediated activation of G proteins.


Asunto(s)
Unión Competitiva/fisiología , Sistema Nervioso Central/metabolismo , Dinorfinas/líquido cefalorraquídeo , Receptores Opioides kappa/metabolismo , Animales , Unión Competitiva/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Líquido Cefalorraquídeo/metabolismo , Dinorfinas/química , Dinorfinas/genética , Endorfinas/líquido cefalorraquídeo , Endorfinas/química , Endorfinas/genética , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Ligandos , Ratones , Ratones Noqueados , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Dolor/genética , Dolor/metabolismo , Dolor/fisiopatología , Radioinmunoensayo , Ensayo de Unión Radioligante , Receptores Opioides/efectos de los fármacos , Receptores Opioides/metabolismo , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/metabolismo , Receptor de Nociceptina
14.
Pain ; 113(3): 301-309, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15661437

RESUMEN

Intrathecal (i.t.) administration into mice of N-ethylmaleimide (NEM), a cysteine protease inhibitor, produced a characteristic behavioral response, the biting and/or licking of the hindpaw and the tail along with slight hindlimb scratching directed toward the flank. The behavior induced by NEM was inhibited by the intraperitoneal injection of morphine. We have recently reported that dynorphin A and, more potently big dynorphin, consisting of dynorphins A and B, produce the same type of nociceptive response whereas dynorphin B does not [Tan-No K, Esashi A, Nakagawasai O, Niijima F, Tadano T, Sakurada C, Sakurada T, Bakalkin G, Terenius L, Kisara K. Intrathecally administered big dynorphin, a prodynorphin-derived peptide, produces nociceptive behavior through an N-methyl-d-aspartate receptor mechanism. Brain Res 2002;952:7-14]. The NEM-induced nociceptive behavior was inhibited by pretreatment with dynorphin A- or dynorphin B-antiserum and each antiserum also reduced the nociceptive effects of i.t.-injected synthetic big dynorphin. The characteristic NEM-evoked response was not observed in prodynorphin knockout mice. Naloxone, an opioid receptor antagonist, had no effects on the NEM-induced behavior. Ifenprodil, arcaine and agmatine, antagonists at the polyamine recognition site on the N-methyl-D-aspartate (NMDA) receptor ion-channel complex, and MK-801, an NMDA ion-channel blocker inhibited the NEM-induced effects. Ro25-6981, an antagonist of the NMDA receptor subtype containing NR2B subunit was not active. NEM completely inhibited degradation of dynorphin A by soluble and particulate fractions of mouse spinal cord. Collectively, the results demonstrate that endogenous prodynorphin-derived peptides are pronociceptive in uninjured animals, and required for the NEM-induced behavior. The NEM effects may be mediated through inhibition of the degradation of endogenous dynorphins, presumably big dynorphin that in turn activates the NMDA receptor ion-channel complex by acting on the polyamine recognition site.


Asunto(s)
Conducta Animal/efectos de los fármacos , Dinorfinas/metabolismo , Inhibidores Enzimáticos/toxicidad , Etilmaleimida/toxicidad , Receptores Opioides/efectos de los fármacos , Agmatina/farmacología , Análisis de Varianza , Animales , Biguanidas/farmacología , Maleato de Dizocilpina/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Dinorfinas/inmunología , Encefalinas/genética , Antagonistas de Aminoácidos Excitadores , Sueros Inmunes/farmacología , Inyecciones Espinales/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Narcóticos/farmacología , Piperidinas/farmacología , Precursores de Proteínas/genética , Receptores Opioides/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Factores de Tiempo , Receptor de Nociceptina
15.
Biochem Biophys Res Commun ; 321(1): 202-9, 2004 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-15358236

RESUMEN

A novel soluble non-opioid dynorphin A-binding factor (DABF) was identified and characterized in neuronal cell lines, rat spinal cord, and brain. DABF binds dynorphin A(1-17), dynorphin A(2-17), and the 32 amino acid prodynorphin fragment big dynorphin consisting of dynorphin A and B, but not other opioid and non-opioid peptides, opiates, and benzomorphans. The IC50 for dynorphin A(1-17), dynorphin A(2-17), and big dynorphin is in the 5-10 nM range. Using dynorphin A and big dynorphin fragments a binding epitope was mapped to dynorphin A(6-13). DABF has a molecular mass of about 70 kDa. SH-groups are apparently involved in the binding of dynorphin A since p-hydroxy-mercuribenzoic acid inhibited this process. Upon interaction with DABF dynorphin A was converted into Leu-enkephalin, which remained bound to the protein. These data suggest that DABF functions as an oligopeptidase that forms stable and specific complexes with dynorphin A. The presence of DABF in brain structures and other tissues with low level of prodynorphin expression suggests that DABF as an oligopeptidase may degrade other peptides. Dynorphin A at the sites of its release in the CNS may attenuate this degradation as a competitor when it specifically binds to the enzyme.


Asunto(s)
Dinorfinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Encéfalo/metabolismo , Carcinoma de Células Pequeñas , Proteínas Portadoras/metabolismo , Línea Celular , Línea Celular Tumoral , Coriocarcinoma , DDT/farmacología , Humanos , Hidroximercuribenzoatos/farmacología , Cinética , Neoplasias Pulmonares , Ratones , Ratones Endogámicos , Neuroblastoma , Neuronas/metabolismo , Inhibidores de Proteasas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...