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

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Tipo del documento
Intervalo de año de publicación
1.
Cells ; 12(20)2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37887313

RESUMEN

De novo mutations in GNAO1, the gene encoding the major neuronal G protein Gαo, cause a spectrum of pediatric encephalopathies with seizures, motor dysfunction, and developmental delay. Of the >80 distinct missense pathogenic variants, many appear to uniformly destabilize the guanine nucleotide handling of the mutant protein, speeding up GTP uptake and deactivating GTP hydrolysis. Zinc supplementation emerges as a promising treatment option for this disease, as Zn2+ ions reactivate the GTP hydrolysis on the mutant Gαo and restore cellular interactions for some of the mutants studied earlier. The molecular etiology of GNAO1 encephalopathies needs further elucidation as a prerequisite for the development of efficient therapeutic approaches. In this work, we combine clinical and medical genetics analysis of a novel GNAO1 mutation with an in-depth molecular dissection of the resultant protein variant. We identify two unrelated patients from Norway and France with a previously unknown mutation in GNAO1, c.509C>G that results in the production of the Pro170Arg mutant Gαo, leading to severe developmental and epileptic encephalopathy. Molecular investigations of Pro170Arg identify this mutant as a unique representative of the pathogenic variants. Its 100-fold-accelerated GTP uptake is not accompanied by a loss in GTP hydrolysis; Zn2+ ions induce a previously unseen effect on the mutant, forcing it to lose the bound GTP. Our work combining clinical and molecular analyses discovers a novel, biochemically distinct pathogenic missense variant of GNAO1 laying the ground for personalized treatment development.


Asunto(s)
Encefalopatías , Humanos , Niño , Mutación/genética , Proteínas de Unión al GTP/metabolismo , Iones/metabolismo , Guanosina Trifosfato , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo
2.
Med ; 4(5): 311-325.e7, 2023 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-37001522

RESUMEN

BACKGROUND: The GNAO1 gene, encoding the major neuronal G protein Gαo, is mutated in a subset of pediatric encephalopathies. Most such mutations consist of missense variants. METHODS: In this study, we present a precision medicine workflow combining next-generation sequencing (NGS) diagnostics, molecular etiology analysis, and personalized drug discovery. FINDINGS: We describe a patient carrying a de novo intronic mutation (NM_020988.3:c.724-8G>A), leading to epilepsy-negative encephalopathy with motor dysfunction from the second decade. Our data show that this mutation creates a novel splice acceptor site that in turn causes an in-frame insertion of two amino acid residues, Pro-Gln, within the regulatory switch III region of Gαo. This insertion misconfigures the switch III loop and creates novel interactions with the catalytic switch II region, resulting in increased GTP uptake, defective GTP hydrolysis, and aberrant interactions with effector proteins. In contrast, intracellular localization, Gßγ interactions, and G protein-coupled receptor (GPCR) coupling of the Gαo[insPQ] mutant protein remain unchanged. CONCLUSIONS: This in-depth analysis characterizes the heterozygous c.724-8G>A mutation as partially dominant negative, providing clues to the molecular etiology of this specific pathology. Further, this analysis allows us to establish and validate a high-throughput screening platform aiming at identifying molecules that could correct the aberrant biochemical functions of the mutant Gαo. FUNDING: This work was supported by the Joint Seed Money Funding scheme between the University of Geneva and the Hebrew University of Jerusalem.


Asunto(s)
Proteínas de Unión al GTP , Ensayos Analíticos de Alto Rendimiento , Humanos , Niño , Evaluación Preclínica de Medicamentos , Mutación/genética , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Guanosina Trifosfato , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo
3.
J Cell Mol Med ; 26(14): 3816-3827, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35678269

RESUMEN

Radix puerariae, a traditional Chinese herbal medication, has been used to treat patients with diabetic kidney disease (DKD). Our previous studies demonstrated that puerarin, the active compound of radix puerariae, improves podocyte injury in type 1 DKD mice. However, the direct molecular target of puerarin and its underlying mechanisms in DKD remain unknown. In this study, we confirmed that puerarin also improved DKD in type 2 diabetic db/db mice. Through RNA-sequencing odf isolated glomeruli, we found that differentially expressed genes (DEGs) that were altered in the glomeruli of these diabetic mice but reversed by puerarin treatment were involved mostly in oxidative stress, inflammatory and fibrosis. Further analysis of these reversed DEGs revealed protein kinase A (PKA) was among the top pathways. By utilizing the drug affinity responsive target stability method combined with mass spectrometry analysis, we identified guanine nucleotide-binding protein Gi alpha-1 (Gnai1) as the direct binding partner of puerarin. Gnai1 is an inhibitor of cAMP production which is known to have protection against podocyte injury. In vitro, we showed that puerarin not only interacted with Gnai1 but also increased cAMP production in human podocytes and mouse diabetic kidney in vivo. Puerarin also enhanced CREB phosphorylation, a downstream transcription factor of cAMP/PKA. Overexpression of CREB reduced high glucose-induced podocyte apoptosis. Inhibition of PKA by Rp-cAMP also diminished the effects of puerarin on high glucose-induced podocyte apoptosis. We conclude that the renal protective effects of puerarin are likely through inhibiting Gnai1 to activate cAMP/PKA/CREB pathway in podocytes.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Podocitos , Animales , Apoptosis , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/farmacología , Glucosa/metabolismo , Guanidina/metabolismo , Guanidina/farmacología , Guanidina/uso terapéutico , Humanos , Isoflavonas , Ratones , Nucleótidos/metabolismo , Podocitos/metabolismo
4.
Cells ; 11(4)2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35203289

RESUMEN

Glucose provides vital energy for cells and contributes to gene expression. The hypothalamus is key for metabolic homeostasis, but effects of glucose on hypothalamic gene expression have not yet been investigated in detail. Thus, herein, we monitored the glucose-dependent transcriptome in murine hypothalamic mHypoA-2/10 cells by total RNA-seq analysis. A total of 831 genes were up- and 1390 genes were downregulated by at least 50%. Key genes involved in the cholesterol biosynthesis pathway were upregulated, and total cellular cholesterol levels were significantly increased by glucose. Analysis of single genes involved in fundamental cellular signaling processes also suggested a significant impact of glucose. Thus, we chose ≈100 genes involved in signaling and validated the effects of glucose on mRNA levels by qRT-PCR. We identified Gnai1-3, Adyc6, Irs1, Igfr1, Hras, and Elk3 as new glucose-dependent genes. In line with this, cAMP measurements revealed enhanced noradrenalin-induced cAMP levels, and reporter gene assays elevated activity of the insulin-like growth factor at higher glucose levels. Key data of our studies were confirmed in a second hypothalamic cell line. Thus, our findings link extra cellular glucose levels with hypothalamic lipid synthesis and pivotal intracellular signaling processes, which might be of particular interest in situations of continuously increased glucose levels.


Asunto(s)
Glucosa , Transcriptoma , Animales , Colesterol/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Glucosa/metabolismo , Hipotálamo/metabolismo , Ratones , Transducción de Señal , Transcriptoma/genética
5.
Hum Mol Genet ; 31(6): 929-941, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-34622282

RESUMEN

Dominant GNAO1 mutations cause an emerging group of childhood-onset neurological disorders characterized by developmental delay, intellectual disability, movement disorders, drug-resistant seizures and neurological deterioration. GNAO1 encodes the α-subunit of an inhibitory GTP/GDP-binding protein regulating ion channel activity and neurotransmitter release. The pathogenic mechanisms underlying GNAO1-related disorders remain largely elusive and there are no effective therapies. Here, we assessed the functional impact of two disease-causing variants associated with distinct clinical features, c.139A > G (p.S47G) and c.662C > A (p.A221D), using Caenorhabditis elegans as a model organism. The c.139A > G change was introduced into the orthologous position of the C. elegans gene via CRISPR/Cas9, whereas a knock-in strain carrying the p.A221D variant was already available. Like null mutants, homozygous knock-in animals showed increased egg laying and were hypersensitive to aldicarb, an inhibitor of acetylcholinesterase, suggesting excessive neurotransmitter release by different classes of motor neurons. Automated analysis of C. elegans locomotion indicated that goa-1 mutants move faster than control animals, with more frequent body bends and a higher reversal rate and display uncoordinated locomotion. Phenotypic profiling of heterozygous animals revealed a strong hypomorphic effect of both variants, with a partial dominant-negative activity for the p.A221D allele. Finally, caffeine was shown to rescue aberrant motor function in C. elegans harboring the goa-1 variants; this effect is mainly exerted through adenosine receptor antagonism. Overall, our findings establish a suitable platform for drug discovery, which may assist in accelerating the development of new therapies for this devastating condition, and highlight the potential role of caffeine in controlling GNAO1-related dyskinesia.


Asunto(s)
Proteínas de Caenorhabditis elegans , Discinesias , Acetilcolinesterasa/metabolismo , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Cafeína/farmacología , Evaluación Preclínica de Medicamentos , Discinesias/tratamiento farmacológico , Discinesias/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/farmacología , Proteínas de Unión al GTP/genética , Mutación , Neurotransmisores/metabolismo
6.
Cell Rep ; 37(7): 109997, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34788630

RESUMEN

The anorexigenic effect of serotonergic compounds has largely been attributed to activation of serotonin 2C receptors (Htr2cs). Using mouse genetic models in which Htr2c can be selectively deleted or restored (in Htr2c-null mice), we investigate the role of Htr2c in forebrain Sim1 neurons. Unexpectedly, we find that Htr2c acts in these neurons to promote food intake and counteract the anorectic effect of serotonergic appetite suppressants. Furthermore, Htr2c marks a subset of Sim1 neurons in the paraventricular nucleus of the hypothalamus (PVH). Chemogenetic activation of these neurons in adult mice suppresses hunger, whereas their silencing promotes feeding. In support of an orexigenic role of PVH Htr2c, whole-cell patch-clamp experiments demonstrate that activation of Htr2c inhibits PVH neurons. Intriguingly, this inhibition is due to Gαi/o-dependent activation of ATP-sensitive K+ conductance, a mechanism of action not identified previously in the mammalian nervous system.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo , Animales , Anorexia , Depresores del Apetito/metabolismo , Depresores del Apetito/farmacología , Metabolismo Energético/fisiología , Conducta Alimentaria/fisiología , Hambre/fisiología , Hipotálamo/metabolismo , Hipotálamo/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/fisiología , Potasio/metabolismo , Receptor de Serotonina 5-HT2C/genética , Serotonina/metabolismo , Serotonina/farmacología , Serotoninérgicos
7.
Theranostics ; 11(17): 8535-8549, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34373757

RESUMEN

Neuroligin-3 (NLGN3) is necessary and sufficient to promote glioma cell growth. The recruitment of Gαi1/3 to the ligand-activated receptor tyrosine kinases (RTKs) is essential for mediating oncogenic signaling. Methods: Various genetic strategies were utilized to examine the requirement of Gαi1/3 in NLGN3-driven glioma cell growth. Results: NLGN3-induced Akt-mTORC1 and Erk activation was inhibited by decreasing Gαi1/3 expression. In contrast ectopic Gαi1/3 overexpression enhanced NLGN3-induced signaling. In glioma cells, NLGN3-induced cell growth, proliferation and migration were attenuated by Gαi1/3 depletion with shRNA, but facilitated with Gαi1/3 overexpression. Significantly, Gαi1/3 silencing inhibited orthotopic growth of patient-derived glioma xenografts in mouse brain, whereas forced Gαi1/3-overexpression in primary glioma xenografts significantly enhanced growth. The growth of brain-metastatic human lung cancer cells in mouse brain was largely inhibited with Gαi1/3 silencing. It was however expedited with ectopic Gαi1/3 overexpression. In human glioma Gαi3 upregulation was detected, correlating with poor prognosis. Conclusion: Gαi1/3 mediation of NLGN3-induced signaling is essential for neuronal-driven glioma growth.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Glioma/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Anciano , Animales , Neoplasias Encefálicas/patología , Moléculas de Adhesión Celular Neuronal/fisiología , Línea Celular Tumoral , Proliferación Celular , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Glioma/genética , Glioma/patología , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Persona de Mediana Edad , Proteínas del Tejido Nervioso/fisiología , Neuronas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Extractos Vegetales , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal
8.
J Ethnopharmacol ; 235: 375-384, 2019 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-30738114

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Either Aconite Lateralis Radix Praeparata (Fuzi) or Pinelliae Rhizoma (Banxia) exerts anti-inflammatory activity and their combination has long been used in China for treating cardiovascular diseases. However, combination of two drugs is controversially prohibited in clinical prescriptions because it serves a representative incompatible pairs in "eighteen antagonisms". Up to date, whether the combination of Fuzi and Banxia could be used for treating heart failure with preserved ejection fraction (HFpEF) especially charactered by systemic inflammation and the potential mechanisms have not been elucidated. AIM OF THE STUDY: The pros and cons of Fuzi in combination with Banxia were evaluated in pressure overload (PO) rat models of HF in vivo. MATERIALS AND METHODS: Male Sprague Dawley rats were subjected to abdominal aorta constriction or sham-operated procedure. From week 12, rats were administered with low dose Fuzi (5.4 g kg-1 d-1), Banxia (5.4 g kg-1 d-1), combination (5.4 g kg-1 d-1 + 5.4 g kg-1 d-1), high dose Fuzi (10.8 g kg-1 d-1) or with vehicle (n = 15 per group) orally for additional 6 weeks. RESULTS: Fuzi alone treatment led to exaggerated cardiac-renal response to PO, and occurred dramatically at high dose as manifested by markedly exacerbated cardiac-renal inflammation and myocardial fibrosis. Further studies revealed that cardiotoxicity of Fuzi may be associated with highly expression levels of ß2-AR and PKA. In contrast, coadministration of Fuzi and Banxia restored cardiac function, as indicated by relieving inflammation and fibrosis as well as normalizing electrocardiogram parameters, which were accompanied by PKA down-regulation. More importantly, both high dose Fuzi and combination treatment enhanced induction of apoptosis, which could be partially associated with inhibition of ß2-AR-Gi signaling. CONCLUSION: Thus, combination of Fuzi and Banxia elicited concurrent protective and toxic effects in PO induced HF. The protective effect appeared to predominate and was associated with suppression of PKA/ß2-AR-Gs signaling pathway. Unlike the eighteen antagonisms theory where Fuzi and Banxia combination was considered incompatible, in the present study, this herb pairs appeared to be benefit, and probably had potential therapeutic prospect in treating HFpEF and diseases associated with inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Pinellia/química , Extractos Vegetales/farmacología , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/aislamiento & purificación , Apoptosis/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Diterpenos , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Inflamación/tratamiento farmacológico , Inflamación/patología , Masculino , Extractos Vegetales/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/efectos de los fármacos
9.
Brain Res ; 1694: 63-72, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29763576

RESUMEN

Orofacial pain includes neuronal pathways that project from the trigeminal nucleus to and through the thalamus. What role the ventroposterior thalamic complex (VP) has on orofacial pain transmission is not understood. To begin to address this question an inhibitory G protein (Gi) designer receptor exclusively activated by a designer drug (DREADD) was transfected in cells of the VP using adeno-associated virus isotype 8. Virus infected cells were identified by a fluorescent tag and immunostaining. Cells were silenced after injecting the designer drug clozapine-n-oxide, which binds the designer receptor activating Gi. Facial rubbing and local field potentials (LFP) in the VP were then recorded in awake, free moving Sprague Dawley rats after formalin injection of the masseter muscle to induce nociception. Formalin injection significantly increased LFP and the nociceptive behavioral response. Activation of DREADD Gi with clozapine-n-oxide significantly reduced LFP in the VP and reduced the orofacial nociceptive response. Because DREADD silencing can result from Gi-coupled inwardly-rectifying potassium channels (GIRK), the GIRK channel blocker tertiapin-Q was injected. Injection of GIRK blocker resulted in an increase in the nociceptive response and increased LFP activity. Immunostaining of the VP for glutamate vesicular transporter (VGLUT2) and gamma-aminobutyric acid vesicular transporter (VGAT) indicated a majority of the virally transfected cells were excitatory (VGLUT2 positive) and a minority were inhibitory (VGAT positive). We conclude first, that inhibition of the excitatory neurons within the VP reduced electrical activity and the orofacial nociceptive response and that the effect on excitatory neurons overwhelmed any change resulting from inhibitor neurons. Second, inhibition of LFP and nociception was due, in part, to GIRK activation.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Neuronas/metabolismo , Tálamo/metabolismo , Animales , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Proteínas de Unión al GTP/metabolismo , Masculino , Ratas Sprague-Dawley , Sinapsinas/metabolismo
10.
Proc Natl Acad Sci U S A ; 113(50): E8041-E8050, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27911799

RESUMEN

Heterotrimeric G proteins are crucial molecular switches that maintain a large number of physiological processes in cells. The signal is encoded into surface alterations of the Gα subunit that carries GTP in its active state and GDP in its inactive state. The ability of the Gα subunit to hydrolyze GTP is essential for signal termination. Regulator of G protein signaling (RGS) proteins accelerates this process. A key player in this catalyzed reaction is an arginine residue, Arg178 in Gαi1, which is already an intrinsic part of the catalytic center in Gα in contrast to small GTPases, at which the corresponding GTPase-activating protein (GAP) provides the arginine "finger." We applied time-resolved FTIR spectroscopy in combination with isotopic labeling and site-directed mutagenesis to reveal the molecular mechanism, especially of the role of Arg178 in the intrinsic Gαi1 mechanism and the RGS4-catalyzed mechanism. Complementary biomolecular simulations (molecular mechanics with molecular dynamics and coupled quantum mechanics/molecular mechanics) were performed. Our findings show that Arg178 is bound to γ-GTP for the intrinsic Gαi1 mechanism and pushed toward a bidentate α-γ-GTP coordination for the Gαi1·RGS4 mechanism. This movement induces a charge shift toward ß-GTP, increases the planarity of γ-GTP, and thereby catalyzes the hydrolysis.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/química , Arginina/química , Dominio Catalítico , Estabilidad de Enzimas , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Hidrólisis , Modelos Moleculares , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Neurofibromina 1/química , Neurofibromina 1/metabolismo , Proteínas RGS/química , Proteínas RGS/genética , Proteínas RGS/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier
11.
J Neurophysiol ; 115(5): 2376-88, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26912590

RESUMEN

Whereas the activation of Gαi/o-coupled receptors commonly results in postsynaptic responses that show acute desensitization, the presynaptic inhibition of transmitter release caused by many Gαi/o-coupled receptors is maintained during agonist exposure. However, an exception has been noted where GABAB receptor (GABABR)-mediated inhibition of inhibitory postsynaptic currents (IPSCs) recorded in mouse proopiomelanocortin (POMC) neurons exhibit acute desensitization in ∼25% of experiments. To determine whether differential effector coupling confers sensitivity to desensitization, voltage-clamp recordings were made from POMC neurons to compare the mechanism by which µ-opioid receptors (MORs) and GABABRs inhibit transmitter release. Neither MOR- nor GABABR-mediated inhibition of release relied on the activation of presynaptic K(+) channels. Both receptors maintained the ability to inhibit release in the absence of external Ca(2+) or in the presence of ionomycin-induced Ca(2+) influx, indicating that inhibition of release can occur through a Ca(2+)-independent mechanism. Replacing Ca(2+) with Sr(2+) to disrupt G-protein-mediated inhibition of release occurring directly at the release machinery did not alter MOR- or GABAB -mediated inhibition of IPSCs, suggesting that reductions in evoked release can occur through the inhibition of Ca(2+) channels. Additionally, both receptors inhibited evoked IPSCs in the presence of selective blockers of N- or P/Q-type Ca(2+) channels. Altogether, the results show that MORs and GABABRs can inhibit transmitter release through the inhibition of calcium influx and by direct actions at the release machinery. Furthermore, since both the desensitizing and nondesensitizing presynaptic receptors are similarly coupled, differential effector coupling is unlikely responsible for differential desensitization of the inhibition of release.


Asunto(s)
Calcio/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Hipotálamo/fisiología , Potenciales Postsinápticos Inhibidores , Receptores de GABA-B/metabolismo , Sinapsis/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Canales de Calcio/metabolismo , Señalización del Calcio , Ratones , Ratones Endogámicos C57BL , Receptores Opioides mu/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
12.
Curr Biol ; 25(10): 1340-6, 2015 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-25936549

RESUMEN

The existence of innate predator aversion evoked by predator-derived chemostimuli called kairomones offers a strong selective advantage for potential prey animals. However, it is unclear how chemically diverse kairomones can elicit similar avoidance behaviors. Using a combination of behavioral analyses and single-cell Ca(2+) imaging in wild-type and gene-targeted mice, we show that innate predator-evoked avoidance is driven by parallel, non-redundant processing of volatile and nonvolatile kairomones through the activation of multiple olfactory subsystems including the Grueneberg ganglion, the vomeronasal organ, and chemosensory neurons within the main olfactory epithelium. Perturbation of chemosensory responses in specific subsystems through disruption of genes encoding key sensory transduction proteins (Cnga3, Gnao1) or by surgical axotomy abolished avoidance behaviors and/or cellular Ca(2+) responses to different predator odors. Stimulation of these different subsystems resulted in the activation of widely distributed target regions in the olfactory bulb, as assessed by c-Fos expression. However, in each case, this c-Fos increase was observed within the same subnuclei of the medial amygdala and ventromedial hypothalamus, regions implicated in fear, anxiety, and defensive behaviors. Thus, the mammalian olfactory system has evolved multiple, parallel mechanisms for kairomone detection that converge in the brain to facilitate a common behavioral response. Our findings provide significant insights into the genetic substrates and circuit logic of predator-driven innate aversion and may serve as a valuable model for studying instinctive fear and human emotional and panic disorders.


Asunto(s)
Reacción de Prevención/fisiología , Hipotálamo/fisiología , Odorantes , Bulbo Olfatorio/fisiología , Animales , Conducta Animal/fisiología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Ganglios/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Fenetilaminas , Feromonas , Conducta Predatoria , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal , Órgano Vomeronasal/fisiología
13.
J Mol Endocrinol ; 54(3): 205-16, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25878061

RESUMEN

Most in vivo effects of 3-iodothyronamine (3-T1AM) have been thus far thought to be mediated by binding at the trace amine-associated receptor 1 (TAAR1). Inconsistently, the 3-T1AM-induced hypothermic effect still persists in Taar1 knockout mice, which suggests additional receptor targets. In support of this general assumption, it has previously been reported that 3-T1AM also binds to the α-2A-adrenergic receptor (ADRA2A), which modulates insulin secretion. However, the mechanism of this effect remains unclear. We tested two different scenarios that may explain the effect: the sole action of 3-T1AM at ADRA2A and a combined action of 3-T1AM at ADRA2A and TAAR1, which is also expressed in pancreatic islets. We first investigated a potential general signaling modification using the label-free EPIC technology and then specified changes in signaling by cAMP inhibition and MAPKs (ERK1/2) determination. We found that 3-T1AM induced Gi/o activation at ADRA2A and reduced the norepinephrine (NorEpi)-induced MAPK activation. Interestingly, in ADRA2A/TAAR1 hetero-oligomers, application of NorEpi resulted in uncoupling of the Gi/o signaling pathway, but it did not affect MAPK activation. However, 3-T1AM application in mice over a period of 6 days at a daily dose of 5 mg/kg had no significant effects on glucose homeostasis. In summary, we report an agonistic effect of 3-T1AM on the ADRA2A-mediated Gi/o pathway but an antagonistic effect on MAPK induced by NorEpi. Moreover, in ADRA2A/TAAR1 hetero-oligomers, the capacity of NorEpi to stimulate Gi/o signaling is reduced by co-stimulation with 3-T1AM. The present study therefore points to a complex spectrum of signaling modification mediated by 3-T1AM at different G protein-coupled receptors.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Receptores Adrenérgicos alfa 2/metabolismo , Tironinas/farmacología , Animales , Evaluación Preclínica de Medicamentos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Expresión Génica , Glucosa/metabolismo , Células HEK293 , Humanos , Islotes Pancreáticos/metabolismo , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Norepinefrina/antagonistas & inhibidores , Norepinefrina/farmacología , Receptores Adrenérgicos alfa 2/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
14.
Clinics ; 70(3): 157-161, 03/2015. tab
Artículo en Inglés | LILACS | ID: lil-747105

RESUMEN

OBJECTIVE: To determine whether Tai Chi Chuan or ballroom dancing promotes better performance with respect to postural balance, gait, and postural transfer among elderly people. METHODS: We evaluated 76 elderly individuals who were divided into two groups: the Tai Chi Chuan Group and the Dance Group. The subjects were tested using the NeuroCom Balance Master¯ force platform system with the following protocols: static balance tests (the Modified Clinical Tests of Sensory Interaction on Balance and Unilateral Stance) and dynamic balance tests (the Walk Across Test and Sit-to-stand Transfer Test). RESULTS: In the Modified Clinical Test of Sensory Interaction on Balance, the Tai Chi Chuan Group presented a lower sway velocity on a firm surface with open and closed eyes, as well as on a foam surface with closed eyes. In the Modified Clinical Test of Sensory Interaction on Unilateral Stance, the Tai Chi Chuan Group presented a lower sway velocity with open eyes, whereas the Dance Group presented a lower sway velocity with closed eyes. In the Walk Across Test, the Tai Chi Chuan Group presented faster walking speeds than those of the Dance Group. In the Sit-to-stand Transfer Test, the Tai Chi Chuan Group presented shorter transfer times from the sitting to the standing position, with less sway in the final standing position. CONCLUSION: The elderly individuals who practiced Tai Chi Chuan had better bilateral balance with eyes open on both types of surfaces compared with the Dance Group. The Dance Group had better unilateral postural balance with eyes closed. The Tai Chi Chuan Group had faster walking speeds, shorter transfer times, and better postural balance in the final standing position during the Sit-to-stand Test. .


Asunto(s)
/metabolismo , AMP Cíclico/metabolismo , Dictyostelium/enzimología , Dictyostelium/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteínas Protozoarias/metabolismo , /genética , Dictyostelium/crecimiento & desarrollo , Dictyostelium/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Ácido Fólico/farmacología , /deficiencia , /genética , /metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Mutación , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Protozoarias/genética , Transducción de Señal , Esporas Protozoarias/enzimología , Esporas Protozoarias/genética , Complejo Vitamínico B/farmacología
15.
Nat Cell Biol ; 15(9): 1107-15, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23934215

RESUMEN

In ciliated mammalian cells, the precise migration of the primary cilium at the apical surface of the cells, also referred to as translational polarity, defines planar cell polarity (PCP) in very early stages. Recent research has revealed a co-dependence between planar polarization of some cell types and cilium positioning at the surface of cells. This important role of the primary cilium in mammalian cells is in contrast with its absence from Drosophila melanogaster PCP establishment. Here, we show that deletion of GTP-binding protein alpha-i subunit 3 (Gαi3) and mammalian Partner of inscuteable (mPins) disrupts the migration of the kinocilium at the surface of cochlear hair cells and affects hair bundle orientation and shape. Inhibition of G-protein function in vitro leads to kinocilium migration defects, PCP phenotype and abnormal hair bundle morphology. We show that Gαi3/mPins are expressed in an apical and distal asymmetrical domain, which is opposite and complementary to an aPKC/Par-3/Par-6b expression domain, and non-overlapping with the core PCP protein Vangl2. Thus G-protein-dependent signalling controls the migration of the cilium cell autonomously, whereas core PCP signalling controls long-range tissue PCP.


Asunto(s)
Cilios/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Proteínas de Ciclo Celular , Movimiento Celular , Polaridad Celular , Forma de la Célula , Cilios/genética , Citoesqueleto/genética , Citoesqueleto/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Regulación de la Expresión Génica , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Externas/citología , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo
16.
Biomed Res Int ; 2013: 212801, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24386635

RESUMEN

Myocardial ischemia/reperfusion (MIR) injury easily occurrs during cardiopulmonary bypass surgery in elderly patients. In an attempt to develop an effective strategy, we employed a pig model of MIR injury to investigate the maximum rate of change of left ventricular pressure, left ventricular enddiastolic pressure, and left intraventricular pressure. Coronary sinus cardiac troponin T (TnT) and adenosine-triphosphate (ATP) content in myocardium were measured. The ultrastructures for MIR injury were visualized by transmission electron microscopy (TEM). The role of δ-opioid receptor activation using D-Ala2, D-Leu5-enkephalin (DADLE) in both early (D1) and late (D2) phases of cardioprotection was identified. Also, the merit of cardioprotection by DADLE in combination with anisodamine, the muscarinic receptor antagonist (D+M), was evaluated. Glibenclamide was employed at the dose sufficient to block ATP-sensitive potassium channels. Significant higher cardiac indicators, reduced TnT and increased ATP contents, were observed in D1, D2, and D+M groups compared with the control group. DADLE induced protection was better in later phase of ischemia that was attenuated by glibenclamide. DADLE after the ischemia showed no benefit, but combined treatment with anisodamine showed a marked postischemic cardioprotection. Thus, anisodamine is helpful in combination with DADLE for postischemic cardioprotection.


Asunto(s)
Puente Cardiopulmonar/efectos adversos , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/prevención & control , Receptores Opioides delta/metabolismo , Alcaloides Solanáceos/uso terapéutico , Adenosina Trifosfato/metabolismo , Animales , Forma de la Célula/efectos de los fármacos , Seno Coronario/efectos de los fármacos , Seno Coronario/metabolismo , Seno Coronario/patología , Seno Coronario/fisiopatología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Pruebas de Función Cardíaca , Daño por Reperfusión Miocárdica/sangre , Miocardio/enzimología , Miocardio/patología , Miocardio/ultraestructura , Proteína Quinasa C/metabolismo , Alcaloides Solanáceos/farmacología , Sus scrofa , Troponina/sangre
17.
J Pharmacol Exp Ther ; 344(1): 179-88, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23097213

RESUMEN

Drugs targeting G-protein-coupled receptors (GPCRs) make up more than 25% of all prescribed medicines. The ability of GPCRs to form heteromers with unique signaling properties suggests an entirely new and unexplored pool of drug targets. However, current in vitro assays are ill equipped to detect heteromer-selective compounds. We have successfully adapted an approach, using fusion proteins of GPCRs and chimeric G proteins, to create an in vitro screening assay (in human embryonic kidney cells) in which only activated heteromers are detectable. Here we show that this assay can demonstrate heteromer-selective G-protein bias as well as measure transinhibition. Using this assay, we reveal that the δ-opioid receptor agonist ADL5859, which is currently in clinical trials, has a 10-fold higher potency against δ-opioid receptor homomers than δ/µ-opioid receptor heteromers (pEC(50) = 6.7 ± 0.1 versus 5.8 ± 0.2). The assay enables the screening of large compound libraries to identify heteromer-selective compounds that could then be used in vivo to determine the functional role of heteromers and develop potential therapeutic agents.


Asunto(s)
Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/efectos de los fármacos , Secuencia de Aminoácidos , Benzamidas/farmacología , Calcio/metabolismo , Señalización del Calcio , Clonación Molecular , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Encefalina Leucina/farmacología , Ensayo de Inmunoadsorción Enzimática , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Células HEK293 , Humanos , Microscopía Fluorescente , Datos de Secuencia Molecular , Proteínas Mutantes Quiméricas/química , Proteínas Mutantes Quiméricas/genética , Piperazinas/farmacología , Reacción en Cadena de la Polimerasa , Receptores Acoplados a Proteínas G/genética , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides delta/genética , Proteínas Recombinantes de Fusión
18.
FASEB J ; 27(2): 832-42, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23150526

RESUMEN

Pasteurella multocida is the causative agent of a number of epizootic and zoonotic diseases. Its major virulence factor associated with atrophic rhinitis in animals and dermonecrosis in bite wounds is P. multocida toxin (PMT). PMT stimulates signal transduction pathways downstream of heterotrimeric G proteins, leading to effects such as mitogenicity, blockade of apoptosis, or inhibition of osteoblast differentiation. On the basis of Gα(i2), it was demonstrated that the toxin deamidates an essential glutamine residue of the Gα(i2) subunit, leading to constitutive activation of the G protein. Here, we studied the specificity of PMT for its G-protein targets by mass spectrometric analyses and by utilizing a monoclonal antibody, which recognizes specifically G proteins deamidated by PMT. The studies revealed deamidation of 3 of 4 families of heterotrimeric G proteins (Gα(q/11), Gα(i1,2,3), and Gα(12/13) of mouse or human origin) by PMT but not by a catalytic inactive toxin mutant. With the use of G-protein fragments and chimeras of responsive or unresponsive G proteins, the structural basis for the discrimination of heterotrimeric G proteins was studied. Our results elucidate substrate specificity of PMT on the molecular level and provide evidence for the underlying structural reasons of substrate discrimination.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidad , Subunidades alfa de la Proteína de Unión al GTP/química , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Pasteurella multocida/metabolismo , Pasteurella multocida/patogenicidad , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Secuencia de Bases , Sitios de Unión , Células Cultivadas , ADN Complementario/genética , Subunidades alfa de la Proteína de Unión al GTP/deficiencia , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Glutamina/química , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Pasteurella multocida/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato
19.
J Thromb Haemost ; 8(9): 2032-41, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20586915

RESUMEN

BACKGROUND: The serine-threonine kinase Akt plays an important role in regulating platelet activation. Stimulation of platelets with various agonists results in Akt activation as indicated by Akt phosphorylation. However, the mechanisms of Akt phosphorylation in platelets are not completely understood. OBJECTIVES AND METHODS: We used P2Y1 knockout mice to address the role of P2Y12 in Akt phosphorylation in response to thrombin receptors in platelets. RESULTS: Thrombin or the PAR4 thrombin receptor peptide AYPGKF at high concentrations stimulated substantial phosphorylation of Akt residues Thr³°8 and Ser47³ in P2Y12-deficient platelets. AYPGKF-induced Akt phosphorylation is enhanced by expression of recombinant human PAR4 cDNA in Chinese hamster ovary (CHO) cells. P2Y12 -independent Akt phosphorylation was not inhibited by integrin inhibitor peptide RGDS or integrin ß3 deficiency. Akt phosphorylation induced by thrombin or AYPGKF in P2Y12-deficient platelets was inhibited by the calcium chelator dimethyl-BAPTA, the Src family kinase inhibitor PP2, and PI3K inhibitors, respectively. CONCLUSIONS: Our results reveal a novel P2Y12-independent signaling pathway mediating Akt phosphorylation in response to thrombin receptors.


Asunto(s)
Plaquetas/citología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Plaquetas/metabolismo , Células CHO , Quelantes/farmacología , Cricetinae , Cricetulus , ADN Complementario/metabolismo , Humanos , Ratones , Ratones Noqueados , Péptidos/química , Fosforilación , Receptores Purinérgicos P2Y12/genética , Proteínas Recombinantes/química , Serina/química , Trombina/química
20.
Lipids ; 45(1): 53-61, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19924462

RESUMEN

Atherosclerosis contributes to disruption of neuronal signaling pathways by producing lipid-dependent modifications of brain plasma membranes, neuroinflammation and oxidative stress. We investigated whether long-term (11 weeks) consumption of refined- (ROO) and pomace- (POO) olive oil modulated the fatty acid composition and the levels of membrane signaling proteins in the brain of apolipoprotein E (apoE) knockout (KO) mice, an animal model of atherosclerosis. Both of these oils are rich in bioactive molecules with anti-inflammatory and antioxidant effects. ROO and POO long-term consumption increased the proportion of monounsaturated fatty acids (MUFAs), particularly of oleic acid, while reducing the level of the saturated fatty acids (SFAs) palmitic and stearic acid. As a result, the MUFA:SFA ratio was higher in apoE KO mice brain fed with ROO and POO. Furthermore, both oils reduced the level of arachidonic and eicosapentaenoic acid, suggesting a decrease in the generation of pro- and anti-inflammatory eicosanoids. Finally, ROO and POO induced an increase in the density of membrane proteins implicated in both the Galphas/PKA and Galphaq/PLCbeta1/PKCalpha signaling pathways. The combined effects of long-term ROO and POO consumption on fatty acid composition and the level of signaling proteins involved in PKA and PKC activation, suggest positive effects on neuroinflammation and brain function in apoE KO mice brain, and convert these oils into promising functional foods in diseases involving apoE deficiency.


Asunto(s)
Apolipoproteínas E/deficiencia , Química Encefálica/efectos de los fármacos , Grasas Insaturadas en la Dieta/farmacología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Aceites de Plantas/farmacología , Animales , Apolipoproteínas E/fisiología , Encéfalo/metabolismo , Subunidad RIIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Subunidad RIIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ácidos Grasos/análisis , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Masculino , Ratones , Ratones Noqueados , Aceite de Oliva , Fosfolipasa C beta/metabolismo , Proteína Quinasa C-alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA