Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biochem Pharmacol ; 225: 116264, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38710334

RESUMO

The retrosplenial cortex (RSC) plays a central role in processing contextual fear conditioning. In addition to corticocortical and thalamocortical projections, the RSC receives subcortical inputs, including a substantial projection from the nucleus incertus in the pontine tegmentum. This GABAergic projection contains the neuropeptide, relaxin-3 (RLN3), which inhibits target neurons via its Gi/o-protein-coupled receptor, RXFP3. To assess this peptidergic system role in contextual fear conditioning, we bilaterally injected the RSC of adult rats with an adeno-associated-virus (AAV), expressing the chimeric RXFP3 agonist R3/I5 or a control AAV, and subjected them to contextual fear conditioning. The R3/I5 injected rats did not display any major differences to control-injected and naïve rats but displayed a significantly delayed extinction. Subsequently, we employed acute bilateral injections of the specific RXFP3 agonist peptide, RXFP3-Analogue 2 (A2), into RSC. While the administration of A2 before each extinction trial had no impact on the extinction process, treatment with A2 before each acquisition trial resulted in delayed extinction. In related anatomical studies, we detected an enrichment of RLN3-immunoreactive nerve fibers in deep layers of the RSC, and a higher level of co-localization of RXFP3 mRNA with vesicular GABA transporter (vGAT) mRNA than with vesicular glutamate transporter-1 (vGLUT1) mRNA across the RSC, consistent with an effect of RLN3/RXFP3 signalling on the intrinsic, inhibitory circuits within the RSC. These findings suggest that contextual conditioning processes in the RSC involve, in part, RLN3 afferent modulation of local inhibitory neurons that provides a stronger memory acquisition which, in turn, retards the extinction process.


Assuntos
Extinção Psicológica , Medo , Receptores Acoplados a Proteínas G , Animais , Masculino , Medo/fisiologia , Medo/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Ratos , Extinção Psicológica/fisiologia , Extinção Psicológica/efeitos dos fármacos , Relaxina/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/efeitos dos fármacos , Giro do Cíngulo/metabolismo , Giro do Cíngulo/efeitos dos fármacos , Giro do Cíngulo/fisiologia , Receptores de Peptídeos
3.
Neuroendocrinology ; 111(12): 1201-1218, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33333517

RESUMO

INTRODUCTION: Food intake varies during the ovarian hormone/estrous cycle in humans and rodents, an effect mediated mainly by estradiol. A potential mediator of the central anorectic effects of estradiol is the neuropeptide relaxin-3 (RLN3) synthetized in the nucleus incertus (NI) and acting via the relaxin family peptide-3 receptor (RXFP3). METHODS: We investigated the relationship between RLN3/RXFP3 signaling and feeding behavior across the female rat estrous cycle. We used in situ hybridization to investigate expression patterns of Rln3 mRNA in NI and Rxfp3 mRNA in the hypothalamic paraventricular nucleus (PVN), lateral hypothalamic area (LHA), medial preoptic area (MPA), and bed nucleus of the stria terminalis (BNST), across the estrous cycle. We identified expression of estrogen receptors (ERs) in the NI using droplet digital PCR and assessed the electrophysiological responsiveness of NI neurons to estradiol in brain slices. RESULTS: Rln3 mRNA reached the lowest levels in the NI pars compacta during proestrus. Rxfp3 mRNA levels varied across the estrous cycle in a region-specific manner, with changes observed in the perifornical LHA, magnocellular PVN, dorsal BNST, and MPA, but not in the parvocellular PVN or lateral LHA. G protein-coupled estrogen receptor 1 (Gper1) mRNA was the most abundant ER transcript in the NI. Estradiol inhibited 33% of type 1 NI neurons, including RLN3-positive cells. CONCLUSION: These findings demonstrate that the RLN3/RXFP3 system is modulated by the estrous cycle, and although further studies are required to better elucidate the cellular and molecular mechanisms of estradiol signaling, current results implicate the involvement of the RLN3/RXFP3 system in food intake fluctuations observed across the estrous cycle in female rats.


Assuntos
Estradiol/metabolismo , Ciclo Estral/metabolismo , Região Hipotalâmica Lateral/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Área Pré-Óptica/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Relaxina/metabolismo , Núcleos Septais/metabolismo , Animais , Feminino , RNA Mensageiro/metabolismo , Ratos
4.
J Neuroendocrinol ; 32(5): e12844, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32307768

RESUMO

Recently, there has been a resurgence in regulatory peptide science as a result of three converging trends. The first is the increasing population of the drug pipeline with peptide-based therapeutics, mainly in, but not restricted to, incretin-like molecules for treatment of metabolic disorders such as diabetes. The second is the development of genetic and optogenetic tools enabling new insights into how peptides actually function within brain and peripheral circuits to accomplish homeostatic and allostatic regulation. The third is the explosion in defined structures of the G-protein coupled receptors to which most regulatory peptides bind and exert their actions. These trends have closely wedded basic systems biology to drug discovery and development, creating a "two-way street" on which translational advances travel from basic research to the clinic, and, equally importantly, "reverse-translational" information is gathered, about the molecular, cellular and circuit-level mechanisms of action of regulatory peptides, comprising information required for the fine-tuning of drug development through testing in animal models. This review focuses on a small group of 'influential' peptides, including oxytocin, vasopressin, pituitary adenylate cyclase-activating polypeptide, ghrelin, relaxin-3 and glucagon-like peptide-1, and how basic discoveries and their application to therapeutics have intertwined over the past decade.


Assuntos
Neuroendocrinologia , Biologia de Sistemas , Animais , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase
5.
Bipolar Disord ; 22(8): 841-848, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32216002

RESUMO

OBJECTIVES: Loss-of-function mutations in the gene encoding the calcium-calmodulin (Ca2+ -CaM)-dependent protein kinase kinase-2 (CaMKK2) enzyme are linked to bipolar disorder. Recently, a de novo arginine to cysteine (R311C) mutation in CaMKK2 was identified from a whole exome sequencing study of bipolar patients and their unaffected parents. The aim of the present study was to determine the functional consequences of the R311C mutation on CaMKK2 activity and regulation by Ca2+ -CaM. METHODS: The effects of the R311C mutation on CaMKK2 activity and Ca2+ -CaM activation were examined using a radiolabeled adenosine triphosphate (ATP) kinase assay. We performed immunoblot analysis to determine whether the R311C mutation impacts threonine-85 (T85) autophosphorylation, an activating phosphorylation site on CaMKK2 that has also been implicated in bipolar disorder. We also expressed the R311C mutant in CaMKK2 knockout HAP1 cells and used immunoblot analysis and an MTS reduction assay to study its effects on Ca2+ -dependent downstream signaling and cell viability, respectively. RESULTS: The R311C mutation maps to the conserved HRD motif within the catalytic loop of CaMKK2 and caused a marked reduction in kinase activity and Ca2+ -CaM activation. The R311C mutation virtually abolished T85 autophosphorylation in response to Ca2+ -CaM and exerted a dominant-negative effect in cells as it impaired the ability of wild-type CaMKK2 to initiate downstream signaling and maintain cell viability. CONCLUSIONS: The highly disruptive, loss-of-function impact of the de novo R311C mutation in human CaMKK2 provides a compelling functional rationale for being considered a potential rare monogenic cause of bipolar disorder.


Assuntos
Transtorno Bipolar/genética , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/genética , Cálcio/metabolismo , Calmodulina/metabolismo , Transtorno Bipolar/diagnóstico , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Calmodulina/genética , Variação Genética , Humanos , Mutação , Fosforilação , Transdução de Sinais/fisiologia
6.
IBRO Rep ; 8: 1-10, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31890981

RESUMO

Modern neuroscience utilizes transgenic techniques extensively to study the activity and function of brain neural networks. A key feature of this approach is its compatibility with molecular methods for selective transgene expression in neuronal circuits of interest. Until now, such targeted transgenic approaches have not been applied to the extensive circuitry involving the neuropeptide, relaxin-3. Pharmacological and gene knock-out studies have revealed relaxin-3 signalling modulates interrelated behaviours and cognitive processes, including stress and anxiety, food and alcohol consumption, and spatial and social memory, highlighting the potential of this system as a therapeutic target. In the present study, we aimed to identify a promoter sequence capable of regulating cell-type specific transgene expression from an adeno-associated viral (AAV) vector in relaxin-3 neurons of the rat nucleus incertus (NI). In parallel to relaxin-3 promoter sequences, we also tested an AAV vector containing promoter elements for the tropomyosin receptor kinase A (TrkA) gene, as TrkA is co-expressed with relaxin-3 in rat NI neurons. Stereotaxic injection of an mCherry-expressing AAV vector revealed widespread non-specific TrkA promoter (880 bp) activity in and adjacent to the NI at 8 weeks post-treatment. In contrast, mCherry expression was successfully restricted to relaxin-3 NI neurons with 98% specificity using a 1736 bp relaxin-3 promoter. In addition to detailed anatomical mapping of NI relaxin-3 networks, illustrated here in association with GABAergic medial septum neurons, this method for targeted transgene delivery offers a versatile tool for ongoing preclinical studies of relaxin-3 circuitry.

7.
Hippocampus ; 29(10): 905-920, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30891856

RESUMO

Anxiety disorders are highly prevalent in modern society and better treatments are required. Key brain areas and signaling systems underlying anxiety include prefrontal cortex, hippocampus, and amygdala, and monoaminergic and peptidergic systems, respectively. Hindbrain GABAergic projection neurons that express the peptide, relaxin-3, broadly innervate the forebrain, particularly the septum and hippocampus, and relaxin-3 acts via a Gi/o -protein-coupled receptor known as the relaxin-family peptide 3 receptor (RXFP3). Thus, relaxin-3/RXFP3 signaling is implicated in modulation of arousal, motivation, mood, memory, and anxiety. Ventral hippocampus (vHip) is central to affective and cognitive processing and displays a high density of relaxin-3-positive nerve fibers and RXFP3 binding sites, but the identity of target neurons and associated effects on behavior are unknown. Therefore, in adult, male rats, we assessed the neurochemical nature of hippocampal RXFP3 mRNA-expressing neurons and anxiety-like and social behavior following chronic RXFP3 activation in vHip by viral vector expression of an RXFP3-selective agonist peptide, R3/I5. RXFP3 mRNA detected by fluorescent in situ hybridization was topographically distributed across the hippocampus in somatostatin- and parvalbumin-mRNA expressing GABA neurons. Chronic RXFP3 activation in vHip increased anxiety-like behavior in the light-dark box and elevated-plus maze, but not the large open-field test, and reduced social interaction with a conspecific stranger. Our data reveal disruptive effects of persistent RXFP3 signaling on hippocampal GABA networks important in anxiety; and identify a potential therapeutic target for anxiety disorders that warrants further investigation in relevant preclinical models.


Assuntos
Ansiedade/metabolismo , Comportamento Animal/fisiologia , Neurônios GABAérgicos/metabolismo , Hipocampo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Comportamento Social , Animais , Comportamento Animal/efeitos dos fármacos , Neurônios GABAérgicos/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores de Peptídeos/agonistas
8.
Brain Struct Funct ; 224(1): 453-469, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30368554

RESUMO

In mammals, the extended amygdala is a neural hub for social and emotional information processing. In the rat, the extended amygdala receives inhibitory GABAergic projections from the nucleus incertus (NI) in the pontine tegmentum. NI neurons produce the neuropeptide relaxin-3, which acts via the Gi/o-protein-coupled receptor, RXFP3. A putative role for RXFP3 signalling in regulating social interaction was investigated by assessing the effect of intracerebroventricular infusion of the RXFP3 agonist, RXFP3-A2, on performance in the 3-chamber social interaction paradigm. Central RXFP3-A2, but not vehicle, infusion, disrupted the capacity to discriminate between a familiar and novel conspecific subject, but did not alter differentiation between a conspecific and an inanimate object. Subsequent studies revealed that agonist-infused rats displayed increased phosphoERK(pERK)-immunoreactivity in specific amygdaloid nuclei at 20 min post-infusion, with levels similar to control again after 90 min. In parallel, we used immunoblotting to profile ERK phosphorylation dynamics in whole amygdala after RXFP3-A2 treatment; and multiplex histochemical labelling techniques to reveal that after RXFP3-A2 infusion and social interaction, pERK-immunopositive neurons in amygdala expressed vesicular GABA-transporter mRNA and displayed differential profiles of RXFP3 and oxytocin receptor mRNA. Overall, these findings demonstrate that central relaxin-3/RXFP3 signalling can modulate social recognition in rats via effects within the amygdala and likely interactions with GABA and oxytocin signalling.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neurônios GABAérgicos/efeitos dos fármacos , Peptídeos/administração & dosagem , Receptores Acoplados a Proteínas G/agonistas , Receptores de Peptídeos/agonistas , Reconhecimento Psicológico/efeitos dos fármacos , Comportamento Social , Ácido gama-Aminobutírico/metabolismo , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/enzimologia , Animais , Neurônios GABAérgicos/enzimologia , Infusões Intraventriculares , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Ocitocina/metabolismo , Fosforilação , Ratos Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ocitocina/genética , Receptores de Ocitocina/metabolismo , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
9.
Neuroscience ; 390: 88-103, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30125684

RESUMO

Anxiety-related defensive behavior is controlled by a distributed network of brain regions and interconnected neural circuits. The dorsal raphe nucleus (DR), which contains the majority of forebrain-projecting serotonergic neurons, is a key brain region involved in fear states and anxiety-related behavior via modulation of this broad neural network. Evidence suggests that relaxin-3 neurons in the nucleus incertus (NI) may also interact with this network, however, the potential role of the NI in the control of anxiety-related defensive behavior requires further investigation. In this study, we examined the response of an anxiety-related neuronal network, including serotonergic neurons in the DR and relaxin-3-containing neurons in the NI, to administration of an anxiogenic drug and exposure to an aversive environment. We administered an anxiogenic dose of the adenosine receptor antagonist, caffeine (50 mg/kg, i.p.), or vehicle, to adult male Wistar rats and 30 min later exposed them to either an elevated plus-maze (EPM) or a home cage environment. Administration of caffeine and exposure to the EPM activated a broad network of brain regions involved in control of anxiety-like behaviors, including serotonergic neurons in the DR, as measured using c-Fos immunohistochemistry. However, only exposure to the EPM activated relaxin-3-containing neurons in the NI, and activation of these neurons was not correlated with changes in anxiety-like behavior. These data suggest activation of the NI relaxin-3 system is associated with expression of behavior in tests of anxiety, but may not be directly involved in the approach-avoidance conflict inherent in anxiety-related defensive behavior in rodents.


Assuntos
Ansiedade/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Núcleos da Rafe/metabolismo , Relaxina/metabolismo , Neurônios Serotoninérgicos/metabolismo , Animais , Ansiedade/induzido quimicamente , Cafeína/administração & dosagem , Masculino , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo , Ratos Wistar
10.
Behav Brain Res ; 336: 135-144, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-28864207

RESUMO

The neuropeptide relaxin-3 (RLN3) binds with high affinity to its cognate receptor, relaxin-family peptide receptor 3 (RXFP3), and with lower affinity to RXFP1, the cognate receptor for relaxin. Intracerebroventricular (icv) administration of RLN3 in rats strongly increases food and water intake and alters the activity of the hypothalamic-pituitary-adrenal (HPA) and gonadal (HPG) axes, but the relative involvement of RXFP3 and RXFP1 in these effects is not known. Therefore, the effects of icv administration of equimolar (1.1 nmol) amounts of RLN3 and the RXFP3-selective agonist RXFP3-A2 on food and water intake, plasma levels of corticosterone, testosterone, and oxytocin and c-fos mRNA expression in key hypothalamic regions in male rats were compared. Food intake was increased by both RLN3 and RXFP3-A2, but the orexigenic effects of RXFP3-A2 were significantly stronger than RLN3, 30 and 60min after injection. Water intake and plasma corticosterone and testosterone levels were significantly increased by RLN3, but not by RXFP3-A2. Conversely, RXFP3-A2 but not RLN3 decreased oxytocin plasma levels. RLN3, but not RXFP3-A2, increased c-fos mRNA levels in the parvocellular (PVNp) and magnocellular (PVNm) paraventricular and supraoptic (SON) hypothalamic nuclei, in the ventral medial preoptic area (MPAv), and in the organum vasculosum of the lamina terminalis (OVLT). A significant increase in c-fos mRNA expression was induced in the perifornical lateral hypothalamic area (LHApf) by RLN3 and RXFP3-A2. These results suggest that RXFP1 is involved in the RLN3 stimulation of water intake and activation of the HPA and HPG axes. The reduced food intake stimulation by RLN3 compared to RXFP3-A2 may relate to activation of both orexigenic and anorexigenic circuits by RLN3.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores de Peptídeos/agonistas , Relaxina/metabolismo , Animais , Corticosterona/sangue , Ingestão de Líquidos/efeitos dos fármacos , Alimentos , Sistema Hipotálamo-Hipofisário , Hipotálamo , Masculino , Proteínas do Tecido Nervoso/farmacologia , Neurônios/metabolismo , Ocitocina/sangue , Sistema Hipófise-Suprarrenal , Proteínas Proto-Oncogênicas c-fos/sangue , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Relaxina/farmacologia , Testosterona/sangue
11.
Acta Neuropathol Commun ; 5(1): 53, 2017 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-28659169

RESUMO

Elevated iron in the SNpc may play a key role in Parkinson's disease (PD) neurodegeneration since drug candidates with high iron affinity rescue PD animal models, and one candidate, deferirpone, has shown efficacy recently in a phase two clinical trial. However, strong iron chelators may perturb essential iron metabolism, and it is not yet known whether the damage associated with iron is mediated by a tightly bound (eg ferritin) or lower-affinity, labile, iron pool. Here we report the preclinical characterization of PBT434, a novel quinazolinone compound bearing a moderate affinity metal-binding motif, which is in development for Parkinsonian conditions. In vitro, PBT434 was far less potent than deferiprone or deferoxamine at lowering cellular iron levels, yet was found to inhibit iron-mediated redox activity and iron-mediated aggregation of α-synuclein, a protein that aggregates in the neuropathology. In vivo, PBT434 did not deplete tissue iron stores in normal rodents, yet prevented loss of substantia nigra pars compacta neurons (SNpc), lowered nigral α-synuclein accumulation, and rescued motor performance in mice exposed to the Parkinsonian toxins 6-OHDA and MPTP, and in a transgenic animal model (hA53T α-synuclein) of PD. These improvements were associated with reduced markers of oxidative damage, and increased levels of ferroportin (an iron exporter) and DJ-1. We conclude that compounds designed to target a pool of pathological iron that is not held in high-affinity complexes in the tissue can maintain the survival of SNpc neurons and could be disease-modifying in PD.


Assuntos
Antiparkinsonianos/farmacologia , Ferro/metabolismo , Fármacos Neuroprotetores/farmacologia , Transtornos Parkinsonianos/tratamento farmacológico , Quinazolinonas/farmacologia , alfa-Sinucleína/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular Tumoral , Cães , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Oxidopamina , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , Substância Negra/patologia , alfa-Sinucleína/genética
12.
Brain Struct Funct ; 222(1): 449-463, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27146679

RESUMO

The medial septum/diagonal band (MS/DB) is a relay region connecting the hypothalamus and brainstem with the hippocampus, and both the MS/DB and dorsal/ventral hippocampus receive strong topographic GABA/peptidergic projections from the nucleus incertus of the pontine tegmentum. The neuropeptide relaxin-3, released by these neurons, is the cognate ligand for a Gi/o-protein-coupled receptor, RXFP3, which is highly expressed within the MS/DB, and both cholinergic and GABAergic neurons in this region of rat brain receive relaxin-3 positive terminals/boutons. Comprehensive in vitro studies have demonstrated that the cell signaling pathways altered by RXFP3 stimulation, include inhibition of forskolin-activated cAMP levels and activation of ERK phosphorylation. In this study we investigated whether intracerebroventricular (icv) injection of RXFP3-A2, a selective relaxin-3 receptor agonist, altered ERK phosphorylation levels in the MS/DB of adult male rats. We subsequently assessed the neurochemical phenotype of phosphorylated (p) ERK-positive neurons in MS/DB after icv RXFP3-A2 administration by dual-label immunostaining for pERK and neuronal markers for cholinergic and GABAergic neurons. Central RXFP3-A2 injection significantly increased levels of pERK immunoreactivity (IR) in MS/DB at 20 and 90 min post-injection, compared to vehicle and naive levels. In addition, RXFP3-A2 increased the number of cells expressing pERK-IR in the MS/DB at 90 (but not 20) min post-injection in cholinergic (but not GABAergic) neurons, which also expressed putative RXFP3-IR. Moreover, icv injection of RXFP3-A2 impaired alternation in a delayed spontaneous T-maze test of spatial working memory. The presence of RXFP3-like IR and the RXFP3-related activation of the MAPK/ERK pathway in MS/DB cholinergic neurons identifies them as a key target of ascending relaxin-3 projections with implications for the acute and chronic modulation of cholinergic neuron activity and function by relaxin-3/RXFP3 signaling.


Assuntos
Neurônios Colinérgicos/metabolismo , Sistema de Sinalização das MAP Quinases , Memória de Curto Prazo/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Núcleos Septais/fisiologia , Memória Espacial/fisiologia , Animais , Neurônios GABAérgicos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Peptídeos/administração & dosagem , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Núcleos Septais/metabolismo
13.
J Med Chem ; 59(16): 7445-56, 2016 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-27464307

RESUMO

Structure-activity studies of the insulin superfamily member, relaxin-3, have shown that its G protein-coupled receptor (RXFP3) binding site is contained within its central B-chain α-helix and this helical structure is essential for receptor activation. We sought to develop a single B-chain mimetic that retained agonist activity. This was achieved by use of solid phase peptide synthesis together with on-resin ruthenium-catalyzed ring closure metathesis of a pair of judiciously placed i,i+4 α-methyl, α-alkenyl amino acids. The resulting hydrocarbon stapled peptide was shown by solution NMR spectroscopy to mimic the native helical conformation of relaxin-3 and to possess potent RXFP3 receptor binding and activation. Alternative stapling procedures were unsuccessful, highlighting the critical need to carefully consider both the peptide sequence and stapling methodology for optimal outcomes. Our result is the first successful minimization of an insulin-like peptide to a single-chain α-helical peptide agonist which will facilitate study of the function of relaxin-3.


Assuntos
Hidrocarbonetos/farmacologia , Peptídeos/farmacologia , Relaxina/agonistas , Animais , Linhagem Celular , Cricetulus , Relação Dose-Resposta a Droga , Humanos , Hidrocarbonetos/química , Masculino , Modelos Moleculares , Estrutura Molecular , Peptídeos/síntese química , Peptídeos/química , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
14.
Glia ; 63(6): 1005-20, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25639936

RESUMO

In order to further investigate the molecular mechanisms that regulate oligodendrocyte (OC) survival, we utilized microarrays to characterize changes in OC gene expression after exposure to the cytokines neurotrophin3, insulin, or leukemia inhibitory factor (LIF) in vitro. We identified and validated the induction and secretion of the neuropeptide galanin in OCs, specifically in response to LIF. We next established that galanin is an OC survival factor and showed that autocrine or paracrine galanin secretion mediates LIF-induced OC survival in vitro. We also revealed that galanin is up-regulated in OCs in the cuprizone model of central demyelination, and that oligodendroglial galanin expression is significantly regulated by endogenous LIF in this context. We also showed that knock-out of galanin reduces OC survival and exacerbates callosal demyelination in the cuprizone model. These findings suggest a potential role for the use of galanin agonists in the treatment of human demyelinating diseases.


Assuntos
Galanina/metabolismo , Fator Inibidor de Leucemia/metabolismo , Bainha de Mielina/fisiologia , Oligodendroglia/fisiologia , Animais , Astrócitos/patologia , Astrócitos/fisiologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Cuprizona , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/fisiopatologia , Modelos Animais de Doenças , Galanina/genética , Expressão Gênica , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Bainha de Mielina/patologia , Células-Tronco Neurais/patologia , Células-Tronco Neurais/fisiologia , Oligodendroglia/patologia , Nervo Óptico/patologia , Nervo Óptico/fisiologia , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley
15.
Pharmacol Rev ; 67(1): 118-75, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25428932

RESUMO

Galanin was first identified 30 years ago as a "classic neuropeptide," with actions primarily as a modulator of neurotransmission in the brain and peripheral nervous system. Other structurally-related peptides-galanin-like peptide and alarin-with diverse biologic actions in brain and other tissues have since been identified, although, unlike galanin, their cognate receptors are currently unknown. Over the last two decades, in addition to many neuronal actions, a number of nonneuronal actions of galanin and other galanin family peptides have been described. These include actions associated with neural stem cells, nonneuronal cells in the brain such as glia, endocrine functions, effects on metabolism, energy homeostasis, and paracrine effects in bone. Substantial new data also indicate an emerging role for galanin in innate immunity, inflammation, and cancer. Galanin has been shown to regulate its numerous physiologic and pathophysiological processes through interactions with three G protein-coupled receptors, GAL1, GAL2, and GAL3, and signaling via multiple transduction pathways, including inhibition of cAMP/PKA (GAL1, GAL3) and stimulation of phospholipase C (GAL2). In this review, we emphasize the importance of novel galanin receptor-specific agonists and antagonists. Also, other approaches, including new transgenic mouse lines (such as a recently characterized GAL3 knockout mouse) represent, in combination with viral-based techniques, critical tools required to better evaluate galanin system physiology. These in turn will help identify potential targets of the galanin/galanin-receptor systems in a diverse range of human diseases, including pain, mood disorders, epilepsy, neurodegenerative conditions, diabetes, and cancer.


Assuntos
Galanina/metabolismo , Neurônios/efeitos dos fármacos , Receptores de Galanina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Desenho de Fármacos , Galanina/genética , Galanina/história , História do Século XX , Humanos , Camundongos Transgênicos , Dados de Sequência Molecular , Terapia de Alvo Molecular , Neurônios/metabolismo , Receptores de Galanina/genética , Receptores de Galanina/história , Receptores de Galanina/metabolismo
16.
Behav Brain Res ; 268: 117-26, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24681162

RESUMO

Behavioural arousal in mammals is regulated by various interacting central monoamine- and peptide-neurotransmitter/receptor systems, which function to maintain awake, alert and active states required for performance of goal-directed activities essential for survival, including food seeking. Existing anatomical and functional evidence suggests the highly-conserved neuropeptide, relaxin-3, which signals via its cognate Gi/o-protein coupled receptor, RXFP3, contributes to behavioural arousal and feeding behaviour in rodents. In studies to investigate this possibility further, adult male C57BL/6J mice were treated with the selective RXFP3 antagonist peptides, R3(B1-22)R/I5(A) and R3(B1-22)R, and motivated food seeking and consumption was assessed as a reflective output of behavioural arousal. Compared to vehicle treatment, intracerebroventricular (icv) injection of RXFP3 antagonists reduced: (i) food anticipatory activity before meal time during food restriction; (ii) consumption of highly palatable food; (iii) consumption of regular chow during the initial dark phase, and; (iv) consumption of regular chow after mild (∼4-h) food deprivation. Effects were not due to sedation and appeared to be specifically mediated via antagonism of relaxin-3/RXFP3 signalling, as RXFP3 antagonist treatment did not alter locomotor activity in wild-type mice or reduce palatable food intake in relaxin-3 deficient (knock-out) mice. Notably, in contrast to similar studies in the rat, icv injection of RXFP3 agonists and infusion into the paraventricular hypothalamic nucleus did not increase food consumption in mice, suggesting species differences in relaxin-3/RXFP3-related signalling networks. Together, our data provide evidence that endogenous relaxin-3/RXFP3 signalling promotes motivated food seeking and consumption, and in light of the established biological and translational importance of other arousal systems, relaxin-3/RXFP3 networks warrant further experimental investigation.


Assuntos
Comportamento Apetitivo/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/administração & dosagem , Ingestão de Alimentos/efeitos dos fármacos , Peptídeos/administração & dosagem , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Animais , Antecipação Psicológica/efeitos dos fármacos , Antecipação Psicológica/fisiologia , Comportamento Apetitivo/fisiologia , Escuridão , Dieta , Ingestão de Alimentos/fisiologia , Privação de Alimentos , Masculino , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Receptores Acoplados a Proteínas G/metabolismo
17.
Behav Brain Res ; 244: 142-51, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23380674

RESUMO

Relaxin-3 is a recently discovered neuropeptide and the results of earlier anatomical and pharmacological studies suggest it plays a physiological role in modulating functions such as arousal, learning and memory, food intake and neuroendocrine homeostasis. Relaxin-3 is also postulated to modulate affective behaviour, based on high densities of the relaxin-3 G-protein coupled receptor (RXFP3) in brain areas involved in stress and mood/anxiety, including the central amygdala, bed nucleus of the stria terminalis and hypothalamic paraventricular nucleus (PVN); and strong activation of relaxin-3 neurons by stressors, via activation of corticotropin-releasing factor receptor-1 (CRF1). This study assessed the effect of central administration of a newly developed RXFP3-selective agonist, on anxiety- and depressive-like behaviour in rats. Adult, male Sprague-Dawley rats administered 5 µg [R3A(11-24,C15→A)B] (referred to as RXFP3-A2), intracerebroventricularly, demonstrated decreased anxiety-like behaviour in the light-dark box and elevated plus maze, but not in the open field. Notably, in the repeat forced swim test, central RXFP3-A2 administration decreased immobility in rats that had been subjected to the 'stress' of former exposure to the anxiety tests, but not in experimentally naïve rats. These data implicate relaxin-3/RXFP3 signalling in the modulation of effects of acute (anxiety) and cumulative (depression) neurogenic stressors on behaviour; and suggest a potential for RXFP3 agonists as anxiolytic and anti-depressant agents. In addition, our results demonstrate that exposure of adult Sprague-Dawley rats to tests of anxiety-like behaviour (∼10-14 days prior) can significantly increase immobility time in the repeat forced swim test.


Assuntos
Ansiedade/prevenção & controle , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Depressão/prevenção & controle , Peptídeos/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Peptídeos/agonistas , Receptores de Peptídeos/fisiologia , Animais , Injeções Intraventriculares , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Proteínas do Tecido Nervoso/fisiologia , Peptídeos/administração & dosagem , Ratos , Relaxina/fisiologia
18.
Front Chem ; 1: 30, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24790958

RESUMO

Relaxin, a heterodimeric polypeptide hormone, is a key regulator of collagen metabolism and multiple vascular control pathways in humans and rodents. Its actions are mediated via its cognate G-protein-coupled receptor, RXFP1 although it also "pharmacologically" activates RXFP2, the receptor for the related, insulin-like peptide 3 (INSL3), which has specific actions on reproduction and bone metabolism. Therefore, experimental tools to facilitate insights into the distinct biological actions of relaxin and INSL3 are required, particularly for studies of tissues containing both RXFP1 and RXFP2. Here, we chemically functionalized human (H2) relaxin, the RXFP1-selective relaxin analog H2:A(4-24)(F23A), and INSL3 to accommodate a fluorophore without marked reduction in binding or activation propensity. Chemical synthesis of the two chains for each peptide was followed by sequential regioselective formation of their three disulfide bonds. Click chemistry conjugation of Cy5.5 at the B-chain N-terminus, with conservation of the disulfide bonds, yielded analogs displaying appropriate selective binding affinity and ability to activate RXFP1 and/or RXFP2 in vitro. The in vivo biological activity of Cy5.5-H2 relaxin and Cy5.5-H2:A(4-24)(F23A) was confirmed in mice, as acute intracerebroventricular (icv) infusion of these peptides (but not Cy5.5-INSL3) stimulated water drinking, an established behavioral response elicited by central RXFP1 activation. The central distribution of Cy5.5-conjugated peptides was examined in mice killed 30 min after infusion, revealing higher fluorescence within brain tissue near-adjacent to the cerebral ventricle walls relative to deeper brain areas. Production of fluorophore-conjugated relaxin family peptides will facilitate future pharmacological studies to probe the function of H2 relaxin/RXFP1 and INSL3/RXFP2 signaling in vivo while tracking their distribution following central or peripheral administration.

19.
Behav Pharmacol ; 23(5-6): 516-25, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22854307

RESUMO

This paper provides a review of the effects of relaxin-3 and structurally related analogues on food intake and related behaviours, in relation to hypothalamic neural networks and chemical messengers known to control feeding, metabolism and body weight, including other neuropeptides and hormones. Soon after relaxin-3 was discovered, pharmacological studies identified the ability of the native peptide to stimulate feeding acutely in adult rats. Although interpretation of these data was confounded by ligand cross-reactivity at relaxin-family peptide (RXFP) receptors, studies with relaxin-3 analogues selective for the native relaxin-3 receptor, RXFP3, confirmed that acute and chronic activation of RXFP3 increased feeding and weight gain, and produced changes in plasma leptin and insulin. These studies also identified the hypothalamus as a locus of action. Studies are now required to identify RXFP3-positive neuron populations involved in the effects of relaxin-3/RXFP3 signalling on metabolic and neuroendocrine homeostasis, and to determine whether peptide-based, nonpeptide-based or gene-based RXFP3 treatments can alter food intake and body weight in animal models of obesity and eating disorders, as a reflection of the therapeutic potential of this newly identified transmitter system.


Assuntos
Estimulantes do Apetite/uso terapêutico , Transtornos da Alimentação e da Ingestão de Alimentos/tratamento farmacológico , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Peptídeos/uso terapêutico , Receptores Acoplados a Proteínas G/agonistas , Receptores de Peptídeos/metabolismo , Relaxina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Animais , Estimulantes do Apetite/administração & dosagem , Estimulantes do Apetite/química , Estimulantes do Apetite/farmacologia , Comportamento Animal/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Transtornos da Alimentação e da Ingestão de Alimentos/metabolismo , Transtornos da Alimentação e da Ingestão de Alimentos/patologia , Injeções Intraventriculares , Ligantes , Camundongos , Terapia de Alvo Molecular , Proteínas do Tecido Nervoso/química , Neurônios/metabolismo , Especificidade de Órgãos , Peptídeos/administração & dosagem , Peptídeos/química , Peptídeos/farmacologia , Ratos , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Relaxina/análogos & derivados , Relaxina/química , Relaxina/farmacologia , Relaxina/uso terapêutico , Aumento de Peso/efeitos dos fármacos
20.
PLoS One ; 7(8): e42300, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22876314

RESUMO

Relaxin-3, the most recently identified member of the relaxin peptide family, is produced by GABAergic projection neurons in the nucleus incertus (NI), in the pontine periventricular gray. Previous studies suggest relaxin-3 is a modulator of stress responses, metabolism, arousal and behavioural activation. Knockout mice and peptide infusions in vivo have significantly contributed to understanding the function of this conserved neuropeptide. Yet, a definitive role remains elusive due to discrepancies between models and a propensity to investigate pharmacological effects over endogenous function. To investigate the endogenous function of relaxin-3, we generated a recombinant adeno-associated viral (rAAV) vector expressing microRNA against relaxin-3 and validated its use to knock down relaxin-3 in adult rats. Bilateral stereotaxic infusion of rAAV1/2 EmGFP miR499 into the NI resulted in significant reductions in relaxin-3 expression as demonstrated by ablation of relaxin-3-like immunoreactivity at 3, 6 and 9 weeks and by qRT-PCR at 12 weeks. Neuronal health was unaffected as transduced neurons in all groups retained expression of NeuN and stained for Nissl bodies. Importantly, qRT-PCR confirmed that relaxin-3 receptor expression levels were not altered to compensate for reduced relaxin-3. Behavioural experiments confirmed no detrimental effects on general health or well-being and therefore several behavioural modalities previously associated with relaxin-3 function were investigated. The validation of this viral vector-based model provides a valuable alternative to existing in vivo approaches and promotes a shift towards more physiologically relevant investigations of endogenous neuropeptide function.


Assuntos
Núcleo Celular/metabolismo , Inativação Gênica , Neuropeptídeos/metabolismo , Relaxina/genética , Animais , Linhagem Celular , Dependovirus/genética , Expressão Gênica , Regulação da Expressão Gênica , Vetores Genéticos/genética , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Neurônios/metabolismo , Interferência de RNA , Ratos , Transdução Genética , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA