Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
1.
Zh Nevrol Psikhiatr Im S S Korsakova ; 123(12): 116-123, 2023.
Artículo en Ruso | MEDLINE | ID: mdl-38147391

RESUMEN

OBJECTIVE: To investigate the associations of OPRM1 gene rs179971, OPRK1 gene rs6473797 and DCC gene rs8084280 polymorphisms with non-suicidal self-injury (NSSI) characteristics and motivations in adults. MATERIAL AND METHODS: A pilot sample included 28 adult patients with history of NSSI (89.3% (n=25) women, median age (Q1-Q3) - 23 (21.25-25) years). Most patients (78.6%, n=20) had a diagnosis of bipolar disorder. NSSI characteristics and motivations were assessed using the Inventory of Statements about Self-Injury (ISAS) scale. The Childhood Trauma Questionnaire (CTQ) was used to control for childhood trauma - one of the most important environmental factors associated with NSSI. The Baratt Impulsivity Scale (BIS) and the Buss-Perry Aggression Questionnaire (BPAQ) were also used to assess impulsivity and aggression, respectively. RT-PCR was used for genotyping, a genetic effect was assessed using the dominant model. Mann-Whitney U-test, Pearson χ2-test and multiple linear regression were used for statistical analysis. RESULTS: Carriers of the minor G allele of OPRM1 gene rs1779971 had a higher level of aggression assessed by BPAQ (p=0.02). The minor C allele of OPRK1 gene rs6473797 was associated with an increase of the subjective importance of «Affect regulation¼ (B=2.23; CI 95% [0.39-4.06]; p=0.022) and «Anti-dissociation¼ (B=3.31; CI 95% [0.18-6.44]; p=0.039) motivations, whereas the minor T allele of DCC gene rs8084280, on the contrary, was associated with a decrease of the importance of «Affect regulation¼ (B=-1.74; CI 95% [-3.30 - -0.18]; p=0.032). Moreover, this effect was found after adjusting for diagnosis, sex, age, and the presence of childhood trauma. CONCLUSIONS: To our knowledge, this is the first study on the association of genetic markers with NSSI motivations. The results of this pilot study demonstrate that OPRK1 and DCC gene polymorphisms can determine differences in motivations for self-harm, however, these results require confirmation in large samples.


Asunto(s)
Receptor DCC , Polimorfismo Genético , Receptores Opioides kappa , Receptores Opioides mu , Conducta Autodestructiva , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptor DCC/genética , Receptor DCC/metabolismo , Conducta Autodestructiva/genética , Humanos , Masculino , Femenino , Adulto Joven , Adulto , Marcadores Genéticos/genética , Encuestas y Cuestionarios , Conducta Impulsiva , Agresión , Polimorfismo Genético/genética
2.
Nicotine Tob Res ; 25(12): 1856-1864, 2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-37455648

RESUMEN

INTRODUCTION: The kappa-opioid receptor (KOR) has been implicated in mediating the behavioral and biochemical effects associated with nicotine reward and withdrawal; however, its underlying mechanisms remain to be further explored. METHODS: Adult male Sprague-Dawley rats were used to establish a nicotine dependence and withdrawal model by injecting nicotine (3 mg/kg/day, s.c.) or vehicle for 14 days, followed by the termination of nicotine for 7 days. Body weight gain, pain behaviors, and withdrawal scores were assessed in succession. MicroRNA (miRNA) sequencing was performed, and quantitative real-time PCR was used to detect the expression of candidate miRNAs and Oprk1. Western blotting was performed to examine KOR protein expression of KOR. Luciferase assay was conducted to validate the relationship of certain miRNAs/Oprk1. RESULTS: The behavioral results showed that nicotine dependence and withdrawal induced behavioral changes. Biochemical analyses demonstrated that miR-144-3p expression decreased and Oprk1/KOR expression increased in the prefrontal cortex, nucleus accumben, and hippocampus. Further investigation suggested that miR-144-3p exerted an inhibitory effect on Oprk1 expression in PC12 cells. CONCLUSIONS: This study revealed that miR-144-3p/Oprk1/KOR might be a potential pathway underlying the adverse effects induced by nicotine dependence and withdrawal, and might provide a novel therapeutic target for smoking cessation. IMPLICATIONS: This study demonstrates an impact of nicotine dependence and nicotine withdrawal on behavioral outcomes and the expressions of miR-144-3p/Oprk1/KOR in male rats. These findings have important translational implications given the continued use of nicotine and the difficulty in smoking cessation worldwide, which can be applied to alleviated the adverse effects induced by nicotine dependence and withdrawal, thus assist smokers to quit smoking.


Asunto(s)
MicroARNs , Receptores Opioides kappa , Síndrome de Abstinencia a Sustancias , Tabaquismo , Animales , Masculino , Ratas , MicroARNs/genética , MicroARNs/uso terapéutico , Nicotina/farmacología , Ratas Sprague-Dawley , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Síndrome de Abstinencia a Sustancias/genética , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Tabaquismo/genética , Tabaquismo/tratamiento farmacológico
3.
Mol Psychiatry ; 28(1): 434-447, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36460726

RESUMEN

Modulation of corticostriatal plasticity alters the information flow throughout basal ganglia circuits and represents a fundamental mechanism for motor learning, action selection, and reward. Synaptic plasticity in the striatal direct- and indirect-pathway spiny projection neurons (dSPNs and iSPNs) is regulated by two distinct networks of GPCR signaling cascades. While it is well-known that dopamine D2 and adenosine A2a receptors bi-directionally regulate iSPN plasticity, it remains unclear how D1 signaling modulation of synaptic plasticity is counteracted by dSPN-specific Gi signaling. Here, we show that striatal dynorphin selectively suppresses long-term potentiation (LTP) through Kappa Opioid Receptor (KOR) signaling in dSPNs. Both KOR antagonism and conditional deletion of dynorphin in dSPNs enhance LTP counterbalancing with different levels of D1 receptor activation. Behaviorally, mice lacking dynorphin in D1 neurons show comparable motor behavior and reward-based learning, but enhanced flexibility during reversal learning. These findings support a model in which D1R and KOR signaling bi-directionally modulate synaptic plasticity and behavior in the direct pathway.


Asunto(s)
Cuerpo Estriado , Dinorfinas , Ratones , Animales , Dinorfinas/metabolismo , Cuerpo Estriado/metabolismo , Ganglios Basales , Potenciación a Largo Plazo , Plasticidad Neuronal/fisiología , Receptores Opioides kappa/genética , Receptores de Dopamina D1/metabolismo
4.
Cell Commun Signal ; 20(1): 35, 2022 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-35305679

RESUMEN

BACKGROUND: As a class of the opioid receptors, the kappa opioid receptor (KOR) has been verified to be a potential biomarker and therapeutic target for human malignant tumors. However, a thorough understanding of whether KOR affects progression of esophageal squamous cell carcinoma (ESCC) is still lacking. This study focused on exploring the effect of knocking down KOR in ESCC and its underlying mechanism. METHODS: Bioinformatics analysis was used to compare the different expression level of OPRK1 (KOR gene) in tumor and adjacent normal tissues, and predict the relationship between KOR expression and overall survival. RNA-sequence analysis was performed to detect the altered functions and mechanisms after down regulating KOR. The in vitro and in vivo assays were used to detect the effects of down-regulated KOR on cell proliferation, migration and invasion. Substrate gel zymography and 3D cell culture assays were used to find the effect of KOR knockdown on the degradation of extracellular matrix (ECM), and immunefluorescence was performed to detect the altered cytoskeleton. Western blotting and immunohistochemistry were used to explore the underlying mechanism pathway. RESULTS: Bioinformatics analysis revealed that the expression of OPRK1 was lower in tumor tissue than that in adjacent normal tissues, and lowered expression of KOR was associated with poorer overall survival. The in vitro assays demonstrated that down-regulation of KOR enhanced ESCC proliferation, metastasis and invasion. Western blotting revealed that down-regulation of KOR could activate PDK1-AKT signaling pathway, which actively regulated the cancer progression. Down-regulation of KOR enhanced the formation of invadopodia, secretion of matrix metalloproteinase-2 (MMP2) and rearrangement of cytoskeleton, which were positively related with the invasion of ESCC. KOR knockdown enhanced the tumor invasion and elevated the AKT phosphorylation in nude mice. The AKT kinase inhibition could reverse the effect of down-regulation of KOR. CONCLUSION: KOR might act as a tumor suppressor in ESCC and down-regulation of KOR could enhance the ESCC tumor phenotype. Video Abstract.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Regulación hacia Abajo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/genética , Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Desnudos , Invasividad Neoplásica/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Transducción de Señal/genética
5.
Handb Exp Pharmacol ; 271: 419-433, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33580386

RESUMEN

The kappa opioid receptor (KOR) is expressed on a number of hematopoietic cell populations, based on both protein binding analysis and the detection of kappa opioid receptor gene (Oprk1) transcripts. There are prominent Oprk1 splice variants that are expressed in the mouse and human brain cells and leukocytes. The activation of KOR results in reduced antibody production, an inhibition of phagocytic cell activity, an inhibition of T cell development, alterations in the production of various pro-inflammatory cytokines, chemokines, and the receptors for these mediators. Finally, the activation of KOR also leads to the regulation of receptor functional activity of chemokine receptors through the process of heterologous desensitization. The functional activity of KOR is important for the regulation of inflammatory responses and may provide opportunities for the development of therapeutics for the treatment of inflammatory disease states.


Asunto(s)
Citocinas , Receptores Opioides kappa , Animales , Sistema Inmunológico , Ratones , Receptores Opioides kappa/genética
6.
BMC Anesthesiol ; 21(1): 210, 2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34461834

RESUMEN

BACKGROUND: Opioid receptors are implicated in cell proliferation and cancer migration. However, the effects and underlying mechanisms of opioid receptor κ (OPRK1) in breast cancer remain unknown. METHODS: Small interfering RNA (siRNAs) was used to knockdown the expression of OPRK1. Western blot was used to determine the protein expression and reverse transcription-quantitative PCR (RT-qPCR) determined the genes transcription. Cell viability was detected by MTT assay and cell death rates were determined by Annexin V/PI and flow cytometry. Cell migration and invasion were detected by wound healing analysis and transwell assay, respectively. RESULTS: Our research demonstrated that OPRK1 was overexpressed in breast cancer cells compared with the normal human mammary epithelial cells. OPRK1 knockdown could inhibited cell viability and migration in cancer cells, accompanied with the decreased proteins and genes expression of N-cadherin, Snail, MMP2 and Vimentin, while the E-cadherin expression was increased. Additionally, OPRK1 knockdown also promoted PI3K/AKT signaling inactivation. Activation of AKT reversed the OPRK1 knockdown-induced cell viability inhibition and migration suppression, while inhibition of AKT reduced cell viability and promoted cell death. CONCLUSIONS: Our findings illustrated the role of OPRK1 played on promoting migration in vitro, and we also provided the therapeutic research of OPRK1 knockdown combined with AKT inhibition.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Receptores Opioides kappa/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Femenino , Silenciador del Gen , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño , Receptores Opioides kappa/genética , Transducción de Señal
7.
Brain Res Bull ; 175: 158-167, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34339779

RESUMEN

Fabry disease (FD) is an X-linked inherited disorder characterized by glycosphingolipid accumulation due to deficiency of α-galactosidase A (α-Gal A) enzyme. Chronic pain and mood disorders frequently coexist in FD clinical setting, however underlying pathophysiologic mechanisms are still unclear. Here we investigated the mechanical and thermal sensitivity in α-Gal A (-/0) hemizygous male and the α-Gal A (-/-) homozygous female mice. We also characterized the gene expression of dynorphinergic, nociceptinergic and CRFergic systems, known to be involved in pain control and mood disorders, in the prefrontal cortex, amygdala and thalamus of α-Gal A (-/0) hemizygous male and the α-Gal A (-/-) homozygous female mice. Moreover, KOP receptor protein levels were evaluated in the same areas. Fabry knock-out male, but not female, mice displayed a decreased pain threshold in both mechanical and thermal tests compared to their wild type littermates. In the amygdala and prefrontal cortex, we observed a decrease of pDYN mRNA levels in males, whereas an increase was assessed in females, thus suggesting sex-related dysregulation of stress coping and pain mechanisms. Elevated mRNA levels for pDYN/KOP and CRF/CRFR1 systems were observed in male and female thalamus, a critical crossroad for both painful signals and cognitive/emotional processes. KOP receptor protein level changes assessed in the investigated areas, appeared mostly in agreement with KOP gene expression alterations. Our data suggest that α-Gal A enzyme deficiency in male and female mice is associated with distinct neuropeptide gene and protein expression dysregulations of investigated systems, possibly related to the neuroplasticity underlying the neurological features of FD.


Asunto(s)
Conducta Animal , Enfermedad de Fabry/psicología , Neuropéptidos/metabolismo , Nocicepción , Animales , Química Encefálica/genética , Hormona Liberadora de Corticotropina , Dinorfinas/genética , Femenino , Expresión Génica , Masculino , Ratones , Ratones Noqueados , Nociceptores , Umbral del Dolor , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Opioides kappa/genética , Caracteres Sexuales
8.
Elife ; 102021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33908346

RESUMEN

Many signal transduction systems have an apparent redundancy built into them, where multiple physiological agonists activate the same receptors. Whether this is true redundancy, or whether this provides an as-yet unrecognized specificity in downstream signaling, is not well understood. We address this question using the kappa opioid receptor (KOR), a physiologically relevant G protein-coupled receptor (GPCR) that is activated by multiple members of the Dynorphin family of opioid peptides. We show that two related peptides, Dynorphin A and Dynorphin B, bind and activate KOR to similar extents in mammalian neuroendocrine cells and rat striatal neurons, but localize KOR to distinct intracellular compartments and drive different post-endocytic fates of the receptor. Strikingly, localization of KOR to the degradative pathway by Dynorphin A induces sustained KOR signaling from these compartments. Our results suggest that seemingly redundant endogenous peptides can fine-tune signaling by regulating the spatiotemporal profile of KOR signaling.


Asunto(s)
Dinorfinas/metabolismo , Receptores Opioides kappa/metabolismo , Animales , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Células Neuroendocrinas/metabolismo , Neuronas/metabolismo , Células PC12 , Ratas , Receptores Opioides kappa/genética , Transducción de Señal
10.
Cell Mol Neurobiol ; 41(5): 1039-1055, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33010019

RESUMEN

The opioid receptor (OPR) family comprises the mu-, delta-, and kappa-opioid, and nociceptin receptors that belong to the superfamily of 7-transmembrane spanning G protein-coupled receptors (GPCRs). The mu-opioid receptor is the main target for clinically used opioid analgesics, and its biology has been extensively studied. The N-terminally truncated 6TM receptors isoform produced through alternative splicing of the OPRM1 gene displays unique signaling and analgesic properties, but it is unclear if other OPRs have the same ability. In this study, we have built a comprehensive map of alternative splicing events that produce 6TM receptor variants in all the OPRs and demonstrated their evolutionary conservation. We then obtained evidence for their translation through ribosomal footprint analysis. We discovered that N-terminally truncated 6TM GPCRs are rare in the human genome and OPRs are overrepresented in this group. Finally, we also observed a significant enrichment of 6TM GPCR genes among genes associated with pain, psychiatric disorders, and addiction. Understanding the biology of 6TM receptors and leveraging this knowledge for drug development should pave the way for novel therapies.


Asunto(s)
Empalme Alternativo/genética , Secuencia Conservada/genética , Receptores Opioides delta/genética , Receptores Opioides kappa/genética , Receptores Opioides mu/genética , Receptores Opioides/genética , Animales , Línea Celular Tumoral , Bases de Datos Genéticas , Variación Genética/genética , Humanos , Macaca , Ratones , Especificidad de la Especie , Receptor de Nociceptina
11.
Brain Res ; 1748: 147083, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32871137

RESUMEN

Thienorphine hydrochloride is a new anti-relapse drug for opioid abusers that is currently in phase II clinical trial. In the present study, the antinociception, dependence, and signal transduction induced by thienorphine were examined. Thienorphine showed a potent antinociception effect in acetic acid-induced writhing test and formalin test. In the hot plate test and tail-flick test, thienorphine presented the typical partial opioid agonist character with a ceiling dose-response curve in addition to a bell-shaped curve. The hot plate test revealed that thienorphine induced approximately 50% of antinociception in µ receptor knockout (µ-KO) mice compared to wild-type controls (P < 0.05). The κ, δ selective antagonist nor-binaltorphimine (nor-BNI), and naltrindole decreased approximately 50-60% of theinorphine antinociception in µ-KO mice, respectively. The ORL1 receptor-selective antagonist J113397 did not affect theinorphine antinociception in µ-KO mice. Chronic treatment with thienorphine (1.5 mg/kg) induced some tolerance that was lower compared to buprenorphine or morphine addition. In contrast to buprenorphine or morphine, thienorphine did not lead to psychological dependence by conditioned place preference (CPP). The maximum inhibition of thienorphine on protein kinase A (PKA) activity was about 36%, 100%, 100%, and 12% in CHO-µ/κ/δ/ORL1-PKAcatEGFP cells, respectively. Similar results were observed in cyclic adenosine monophosphate (cAMP) accumulation inhibited by thienorphine in cells. Thienorphine significantly increased pERK1/2 in CHO-κ/δ-PKAcatEGFP cells. These results indicated that thienorphine induced analgesia through activation of κ- and δ-, partial activation of µ- opioid receptor without a bias between G-protein- and ß-arrestin-mediated pathways. Thienorphine might be used for antinociception with minimal adverse effects.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Encéfalo/efectos de los fármacos , Buprenorfina/análogos & derivados , Nocicepción/efectos de los fármacos , Dolor/tratamiento farmacológico , Analgésicos Opioides/farmacología , Animales , Encéfalo/metabolismo , Buprenorfina/farmacología , Buprenorfina/uso terapéutico , Línea Celular Tumoral , AMP Cíclico/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Dolor/genética , Dolor/metabolismo , Ratas , Ratas Wistar , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo
12.
Nat Commun ; 11(1): 1145, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-32123179

RESUMEN

Recent studies show that GPCRs rapidly interconvert between multiple states although our ability to interrogate, monitor and visualize them is limited by a relative lack of suitable tools. We previously reported two nanobodies (Nb39 and Nb6) that stabilize distinct ligand- and efficacy-delimited conformations of the kappa opioid receptor. Here, we demonstrate via X-ray crystallography a nanobody-targeted allosteric binding site by which Nb6 stabilizes a ligand-dependent inactive state. As Nb39 stabilizes an active-like state, we show how these two state-dependent nanobodies can provide real-time reporting of ligand stabilized states in cells in situ. Significantly, we demonstrate that chimeric GPCRs can be created with engineered nanobody binding sites to report ligand-stabilized states. Our results provide both insights regarding potential mechanisms for allosterically modulating KOR with nanobodies and a tool for reporting the real-time, in situ dynamic range of GPCR activity.


Asunto(s)
Receptores Opioides kappa/química , Receptores Opioides kappa/metabolismo , Anticuerpos de Dominio Único/química , Sitio Alostérico , Sitios de Unión , Técnicas Biosensibles , Cristalografía por Rayos X , AMP Cíclico/metabolismo , Dinorfinas/química , Dinorfinas/farmacología , Células HEK293 , Humanos , Mediciones Luminiscentes/métodos , Piperazinas/química , Piperazinas/farmacología , Piperidinas/química , Piperidinas/farmacología , Conformación Proteica , Pirrolidinas/química , Pirrolidinas/farmacología , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Anticuerpos de Dominio Único/metabolismo , Tetrahidroisoquinolinas/química , Tetrahidroisoquinolinas/farmacología
13.
Genet Test Mol Biomarkers ; 24(1): 17-23, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31940240

RESUMEN

Background: Heroin use disorder (HUD) is a complex brain disease that includes multiple phenotypes. Heroin acts primarily as a mu-opioid receptor (OPRM1) agonist. The κ opioid receptor 1 (OPRK1) is critically involved in abstinence and remission. Multiple studies confirm that the OPRM1 and OPRK1 genes are associated with HUD. However, their relationship with the addictive phenotype is still unclear. This study was designed to identify the genetic polymorphisms within OPRM1 and OPRK1 with six HUD phenotypes. Methods: A total of 801 patients with HUD were recruited from the Methadone Maintenance Treatment Program in Xi'an. We identified eight potential functional single nucleotide polymorphisms (SNPs) in the two genes that were genotyped using SNaPshot SNP technology. We then performed a case-control association analysis, investigated particular disease phenotypes, and assessed the extent of epistasis among the variants of the two genes. Results: The OPRK1 rs3802279, rs3802281, and rs963549 genotypes were significantly associated with methadone dosage analyzed by Pearson's chi-square test or binary logistic regression to correct for covariates. The rs3802279 CC, rs3802281 TT, and rs963549 CC genotype carriers required a lower methadone maintenance dose per day. Multifactor dimensionality reduction analysis indicated strong interactions between sex and OPRK1 rs963549. The results of the OPRM1 genotyping did not reveal any associations with the various HUD phenotypes. Conclusion: These findings support an important role of the OPRK1 polymorphism in determining the daily methadone dose and may guide future studies in identifying additional genetic risk factors for HUD.


Asunto(s)
Dependencia de Heroína/genética , Receptores Opioides kappa/genética , Receptores Opioides mu/genética , Adulto , Pueblo Asiatico/genética , Biomarcadores Farmacológicos , Estudios de Casos y Controles , China/epidemiología , Relación Dosis-Respuesta a Droga , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Haplotipos , Humanos , Masculino , Metadona/farmacología , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Trastornos Relacionados con Sustancias/genética
14.
Artículo en Inglés | MEDLINE | ID: mdl-31689444

RESUMEN

The pathoetiology and pathophysiology of borderline personality disorder (BPD) have been relatively under-explored. Consequently, no targetted pharmaceutical treatments or preventative interventions are available. The current article reviews the available data on the biological underpinnings of BPD, highlighting a role for early developmental processes, including prenatal stress and maternal dysbiosis, in BPD pathoetiology. Such factors are proposed to drive alterations in the infant's gut microbiome, in turn modulating amygdala development and the amygdala's two-way interactions with other brain regions. Alterations in opioidergic activity, including variations in the ratio of the mu-and kappa-opioid receptors seem a significant aspect of BPD pathophysiology, contributing to its comorbidities with depression, anxiety, impulsivity and addiction. Stress and dysphoria are commonly experienced in people classed with BPD. A growing body of data, across a host of medical conditions, indicate that stress and mood dysregulation may be intimately associated with gut dysbiosis and increased gut permeability, coupled to heightened levels of oxidative stress and immune-inflammatory activity. It urgently requires investigation as to the relevance of such gut changes in the course of BPD symptomatology. Accumulating data indicates that BPD symptom exacerbations may be linked to cyclical variations in estrogen, in turn decreasing serotonin and local melatonin synthesis, and thereby overlapping with the pathophysiology of migraine and endometriosis, which also have a heightened association with BPD. Future research directions and treatment implications are indicated.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Trastorno de Personalidad Limítrofe/etiología , Trastorno de Personalidad Limítrofe/fisiopatología , Microbioma Gastrointestinal/fisiología , Corteza Prefrontal/fisiopatología , Receptores Opioides kappa/genética , Receptores Opioides mu/genética , Adulto , Amígdala del Cerebelo/metabolismo , Trastorno de Personalidad Limítrofe/mortalidad , Disbiosis/complicaciones , Disbiosis/psicología , Femenino , Humanos , Recién Nacido , Trastornos Mentales/complicaciones , Corteza Prefrontal/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Estrés Psicológico/complicaciones , Estrés Psicológico/psicología
15.
Nat Commun ; 10(1): 4037, 2019 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-31492869

RESUMEN

Increased body weight is a major factor that interferes with smoking cessation. Nicotine, the main bioactive compound in tobacco, has been demonstrated to have an impact on energy balance, since it affects both feeding and energy expenditure at the central level. Among the central actions of nicotine on body weight, much attention has been focused on its effect on brown adipose tissue (BAT) thermogenesis, though its effect on browning of white adipose tissue (WAT) is unclear. Here, we show that nicotine induces the browning of WAT through a central mechanism and that this effect is dependent on the κ opioid receptor (KOR), specifically in the lateral hypothalamic area (LHA). Consistent with these findings, smokers show higher levels of uncoupling protein 1 (UCP1) expression in WAT, which correlates with smoking status. These data demonstrate that central nicotine-induced modulation of WAT browning may be a target against human obesity.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Nicotina/farmacología , Receptores Opioides kappa/metabolismo , Termogénesis/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Adulto , Animales , Peso Corporal/efectos de los fármacos , Femenino , Estimulantes Ganglionares/administración & dosificación , Estimulantes Ganglionares/farmacología , Humanos , Hipotálamo/metabolismo , Masculino , Ratones Noqueados , Persona de Mediana Edad , Nicotina/administración & dosificación , Ratas Sprague-Dawley , Receptores Opioides kappa/genética , Proteína Desacopladora 1/metabolismo
16.
Sci Rep ; 9(1): 5550, 2019 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-30944388

RESUMEN

The tumour suppressor protein RASSF1A is phosphorylated by Aurora A kinase, thereby impairing its tumour suppressor function. Consequently, inhibiting the interaction between Aurora A and RASSF1A may be used for anti-tumour therapy. We used recombinant variants of RASSF1A to map the sites of interaction with Aurora A. The phosphorylation kinetics of three truncated RASSF1A variants has been analysed. Compared to the RASSF1A form lacking the 120 residue long N-terminal part, the Km value of the phosphorylation is increased from 10 to 45 µM upon additional deletion of the C-terminal SARAH domain. On the other hand, deletion of the flexible loop (Δ177-197) that precedes the phosphorylation site/s (T202/S203) results in a reduction of the kcat value from about 40 to 7 min-1. Direct physical interaction between the isolated SARAH domain and Aurora A was revealed by SPR. These data demonstrate that the SARAH domain of RASSF1A is involved in the binding to Aurora A kinase. Structural modelling confirms that a novel complex is feasible between the SARAH domain and the kinase domain of Aurora A. In addition, a regulatory role of the loop in the catalytic phosphorylation reaction has been demonstrated both experimentally and by structural modelling.


Asunto(s)
Aurora Quinasa A/metabolismo , Dominios y Motivos de Interacción de Proteínas , Receptores Opioides kappa/metabolismo , Aurora Quinasa A/química , Aurora Quinasa A/genética , Sitios de Unión , Cromatografía en Gel , Modelos Moleculares , Mutación , Fosforilación , Multimerización de Proteína , Receptores Opioides kappa/química , Receptores Opioides kappa/genética , Resonancia por Plasmón de Superficie
17.
Lipids Health Dis ; 18(1): 52, 2019 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-30764838

RESUMEN

BACKGROUND: This study was designed to test the hypothesis that κ-opioid receptor (κ-OR) stimulation reduces palmitate-induced HUVECs apoptosis and to investigate its mechanisms. METHODS: HUVECs were subjected to sodium palmitate, apoptosis and cell viability were determined, HUVECs were treated with specific inhibitors to PI3K, Akt, eNOS and siRNAs targeting κ-OR and Akt. Groups were divided as follows: the control group, the sodium palmitate group, the sodium palmitate+U50,488H (a selective κ-OR agonist) group and the sodium palmitate+U50,488H + nor-BNI (a selective κ-OR antagonist) group. RESULTS: Treatment with sodium palmitate significantly reduced cell viability and increased apoptosis rate which were significantly alleviated by pretreatment with U50,488H, the effect of U50,488H was abolished by nor-BNI. Phosphorylation of Akt and eNOS, as well as NO production were attenuated and accompanied by an increased expression of caspase 3 when HUVECs were subjected to sodium palmitate, and all these changes were restored by pretreatment with U50,488H, the effects of U50,488H were abolished by nor-BNI, and specific inhibitors to PI3K, Akt, eNOS, respectively. SiRNAs targeting κ-OR or Akt abolished the effects of U50,488H on phosphorylation of Akt and eNOS as well as the expressions of caspase 3, Bax and Bcl-2. SiRNAs targeting Akt elicited no effect on the expression of κ-OR. CONCLUSION: This study provides the evidence for the first time that κ-OR stimulation possesses anti-palmitate-induced apoptosis effect, which is mediated by PI3K/Akt/eNOS signaling pathway.


Asunto(s)
3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Apoptosis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/genética , Proteínas Proto-Oncogénicas c-akt/genética , Receptores Opioides kappa/genética , Analgésicos Opioides/farmacología , Caspasa 3/genética , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ácido Palmítico/antagonistas & inhibidores , Ácido Palmítico/farmacología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides kappa/metabolismo , Transducción de Señal , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
18.
Sci Signal ; 11(542)2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30087177

RESUMEN

Biased agonists of G protein-coupled receptors may present a means to refine receptor signaling in a way that separates side effects from therapeutic properties. Several studies have shown that agonists that activate the κ-opioid receptor (KOR) in a manner that favors G protein coupling over ß-arrestin2 recruitment in cell culture may represent a means to treat pain and itch while avoiding sedation and dysphoria. Although it is attractive to speculate that the bias between G protein signaling and ß-arrestin2 recruitment is the reason for these divergent behaviors, little evidence has emerged to show that these signaling pathways diverge in the neuronal environment. We further explored the influence of cellular context on biased agonism at KOR ligand-directed signaling toward G protein pathways over ß-arrestin-dependent pathways and found that this bias persists in striatal neurons. These findings advance our understanding of how a G protein-biased agonist signal differs between cell lines and primary neurons, demonstrate that measuring [35S]GTPγS binding and the regulation of adenylyl cyclase activity are not necessarily orthogonal assays in cell lines, and emphasize the contributions of the environment to assessing biased agonism.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Neuronas/metabolismo , Receptores Opioides kappa/agonistas , Transducción de Señal , Animales , Animales Recién Nacidos , Bencenoacetamidas/farmacología , Células CHO , Línea Celular Tumoral , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Cricetinae , Cricetulus , Células HEK293 , Humanos , Ratones Noqueados , Pirrolidinas/farmacología , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Arrestina beta 2/genética , Arrestina beta 2/metabolismo
19.
Neurosci Lett ; 685: 75-82, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30138645

RESUMEN

Prodynorphin (PDYN) binds to k-opioid receptors (KOPr; encoded by OPRK1) and is known to regulate dopaminergic tone, making this system important for drugs addiction. Dynorphin (Dyn)/KORr system are powerful effectors of stress-induced alterations in reward processing and dysphoric states. Thus, We identified 11 potential functional SNPs and one variable number of tandem repeat (VNTR) in this system, performed a case-control association analysis, investigated particular disease phenotypes, assessed the joint effect of variants in two genes, carried out a meta-analysis to analyze the association between this VNTR and Heroin dependence (HD) risk. Eleven single-nucleotide polymorphisms (SNPs) were genotyped using SNaPshot SNP technology. Participants included 566 healthy controls and 541 patients with HD. We found that PDYN polymorphisms modulate the susceptibility to HD. An increased risk of HD was significantly associated with H alleles of PDYN VNTR (χ2 = 10.824, p = 0.001, OR = 1.419, 95% CI = 1.151-1.748). In addition, the results revealed the patients with the HH genotype showed greater number of withdrawal instances (F(2538) = 7.987, p = 0.0004) compared to the patients with the LL genotype. The Meta-analysis showed the pooled effect of the H allele at this locus is a risk factor for HD in Chinese Han. Gene-gene interaction analysis indicated strong interactions between PDYN rs3830064, 68-bp VNTR and OPRK1 rs16918842, rs3802279. These findings support the important role of PDYN polymorphism in HD, and may guide future studies to identify genetic risk factors for HD.


Asunto(s)
Dinorfinas/genética , Predisposición Genética a la Enfermedad , Dependencia de Heroína/genética , Receptores Opioides kappa/genética , Analgésicos Opioides/farmacología , Pueblo Asiatico , Frecuencia de los Genes/genética , Pruebas Genéticas/métodos , Genotipo , Humanos , Polimorfismo de Nucleótido Simple/genética
20.
Science ; 360(6395)2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29930108

RESUMEN

A systems view of G protein-coupled receptor (GPCR) signaling in its native environment is central to the development of GPCR therapeutics with fewer side effects. Using the kappa opioid receptor (KOR) as a model, we employed high-throughput phosphoproteomics to investigate signaling induced by structurally diverse agonists in five mouse brain regions. Quantification of 50,000 different phosphosites provided a systems view of KOR in vivo signaling, revealing novel mechanisms of drug action. Thus, we discovered enrichment of the mechanistic target of rapamycin (mTOR) pathway by U-50,488H, an agonist causing aversion, which is a typical KOR-mediated side effect. Consequently, mTOR inhibition during KOR activation abolished aversion while preserving beneficial antinociceptive and anticonvulsant effects. Our results establish high-throughput phosphoproteomics as a general strategy to investigate GPCR in vivo signaling, enabling prediction and modulation of behavioral outcomes.


Asunto(s)
Encéfalo/metabolismo , Ensayos Analíticos de Alto Rendimiento , Fosfoproteínas/metabolismo , Proteómica/métodos , Receptores Opioides kappa/metabolismo , Transducción de Señal , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/metabolismo , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Analgésicos no Narcóticos/farmacología , Animales , Anticonvulsivantes/farmacología , Arrestinas/metabolismo , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Línea Celular Tumoral , Diterpenos de Tipo Clerodano/metabolismo , Diterpenos de Tipo Clerodano/farmacología , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenetilaminas/metabolismo , Fenetilaminas/farmacología , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética , Transducción de Señal/efectos de la radiación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA