Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R594-R606, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30758978

RESUMO

The stress peptide pituitary adenylate cyclase activating polypeptide (PACAP) and its specific receptor PACAP type 1 receptor (PAC1) have been implicated in sudden infant death syndrome (SIDS). PACAP is also critical to the neonatal cardiorespiratory response to homeostatic stressors identified in SIDS, including hypoxia. However, which of PACAP's three receptors, PAC1, vasoactive intestinal peptide receptor type 1 (VPAC1), and/or vasoactive intestinal peptide receptor type 2 (VPAC2), are involved is unknown. In this study, we hypothesized that PAC1, but not VPAC2, is involved in mediating the cardiorespiratory response to hypoxia during neonatal development. To test this hypothesis, head-out plethysmography and surface ECG electrodes were used to assess the cardiorespiratory variables of unanesthetized postnatal day 4 PAC1 and VPAC2-knockout (KO) and wild-type (WT) mice in response to a 10% hypoxic challenge. Our results demonstrate that compared with WT pups, the early and late hypoxic rate of expired CO2 (V̇co2), V̇co2 and ventilatory responses were blunted in PAC1-KO neonates, and during the posthypoxic period, minute ventilation (V̇e), V̇co2 and heart rate were increased, while the increase in apneas normally associated with the posthypoxic period was reduced. Consistent with impaired cardiorespiratory control in these animals, the V̇e/V̇co2 slope was reduced in PAC1-KO pups, suggesting that breathing was inappropriately matched to metabolism. In contrast, VPAC2-KO pups exhibited elevated heart rate variability during hypoxia compared with WT littermates, but the effects of the VPAC2-KO genotype on breathing were minimal. These findings suggest that PAC1 plays the principal role in mediating the cardiorespiratory effects of PACAP in response to hypoxic stress during neonatal development and that defective PACAP signaling via PAC1 may contribute to the pathogenesis of SIDS.


Assuntos
Sistema Cardiovascular/metabolismo , Frequência Cardíaca , Hipóxia/metabolismo , Pulmão/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Ventilação Pulmonar , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/deficiência , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Morte Súbita do Lactente/etiologia , Animais , Animais Recém-Nascidos , Sistema Cardiovascular/fisiopatologia , Modelos Animais de Doenças , Feminino , Humanos , Hipóxia/genética , Hipóxia/fisiopatologia , Recém-Nascido , Pulmão/fisiopatologia , Masculino , Camundongos Knockout , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Transdução de Sinais , Morte Súbita do Lactente/genética
2.
Exp Eye Res ; 169: 134-140, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29428294

RESUMO

The intraocular pressure of mice displays a daily rhythmicity being highest during the dark period. The present study was performed to elucidate the role of the circadian clock and light in the diurnal and the circadian variations in intraocular pressure in mice, by using animals with disrupted clock function (VPAC2 receptor knockout mice) or impaired light information to the clock (PACAP knockout mice). In wildtype mice, intraocular pressure measured under light/dark conditions showed a statistically significant 24 h sinusoidal rhythm with nadir during the light phase and peak during the dark phase. After transfer of the wildtype mice into constant darkness, the intraocular pressure increased, but the rhythmic changes in intraocular pressure continued with a pattern identical to that obtained during the light/dark cycle. The intraocular pressure in VPAC2 receptor deficient mice during light/dark conditions also showed a sinusoidal pattern with significant changes as a function of a 24 h cycle. However, transfer of the VPAC2 receptor knockout mice into constant darkness completely abolished the rhythmic changes in intraocular pressure. The intraocular pressure in PACAP deficient mice oscillated significantly during both 24 h light and darkness and during constant darkness. During LD conditions, the amplitude of PACAP deficient was significantly lower compared to wildtype mice, resulting in higher daytime and lower nighttime values. In conclusion, by studying the VPAC2 receptor knockout mouse which lacks circadian control and the PACAP knockout mouse which displays impaired light signaling, we provided evidence that the daily intraocular pressure rhythms are primarily generated by the circadian master clock and to a lesser extent by environmental light and darkness.


Assuntos
Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Pressão Intraocular/fisiologia , Luz , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/deficiência , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Receptores Tipo II de Peptídeo Intestinal Vasoativo/fisiologia , Tonometria Ocular
3.
PLoS One ; 12(11): e0188166, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29155851

RESUMO

The two sister peptides, pituitary adenylate cyclase activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) and their receptors, the PAC1 -and the VPAC2 receptors, are involved in regulation of the circadian timing system. PACAP as a neurotransmitter in the retinohypothalamic tract (RHT) and VIP as a neurotransmitter, involved in synchronization of SCN neurons. Behavior and physiology in VPAC2 deficient mice are strongly regulated by light most likely as a result of masking. Consequently, we used VPAC2 and PAC1/VPAC2 double mutant mice in comparison with PAC1 receptor deficient mice to further elucidate the role of PACAP in the light mediated regulation of behavior and physiology of the circadian system. We compared circadian rhythms in mice equipped with running wheels or implanted radio-transmitter measuring core body temperature kept in a full photoperiod ((FPP)(12:12 h light dark-cycles (LD)) and skeleton photo periods (SPP) at high and low light intensity. Furthermore, we examined the expression of PAC1- and VPAC2 receptors in the SCN of the different genotypes in combination with visualization of PACAP and VIP and determined whether compensatory changes in peptide and/or receptor expression in the reciprocal knockouts (KO) (PAC1 and VPAC2) had occurred. Our data demonstrate that in although being closely related at both ligand and receptor structure/sequence, PACAP/PAC1 receptor signaling are independent of VIP/VPAC2 receptor signaling and vice versa. Furthermore, lack of either of the receptors does not result in compensatory changes at neither the physiological or anatomical level. PACAP/PAC1 signaling is important for light regulated behavior, VIP/VPAC2signaling for stable clock function and both signaling pathways may play a role in shaping diurnality versus nocturnality.


Assuntos
Ritmo Circadiano/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Peptídeo Intestinal Vasoativo/genética , Sequência de Aminoácidos , Animais , Temperatura Corporal/fisiologia , Feminino , Regulação da Expressão Gênica , Luz , Transdução de Sinal Luminoso , Masculino , Camundongos , Camundongos Knockout , Fotoperíodo , Condicionamento Físico Animal , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/deficiência , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Corrida , Peptídeo Intestinal Vasoativo/metabolismo
4.
Sci Rep ; 5: 14044, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26370467

RESUMO

Individual neurons in the suprachiasmatic nuclei (SCN) contain an intracellular molecular clock and use intercellular signaling to synchronize their timekeeping activities so that the SCN can coordinate brain physiology and behavior. The neuropeptide vasoactive intestinal polypeptide (VIP) and its VPAC2 receptor form a key component of intercellular signaling systems in the SCN and critically control cellular coupling. Targeted mutations in either the intracellular clock or intercellular neuropeptide signaling mechanisms, such as VIP-VPAC2 signaling, can lead to desynchronization of SCN neuronal clocks and loss of behavioral rhythms. An important goal in chronobiology is to develop interventions to correct deficiencies in circadian timekeeping. Here we show that extended exposure to constant light promotes synchrony among SCN clock cells and the expression of ~24 h rhythms in behavior in mice in which intercellular signaling is disrupted through loss of VIP-VPAC2 signaling. This study highlights the importance of SCN synchrony for the expression of rhythms in behavior and reveals how non-invasive manipulations in the external environment can be used to overcome neurochemical communication deficits in this important brain system.


Assuntos
Relógios Circadianos/genética , Relógios Circadianos/efeitos da radiação , Ritmo Circadiano/genética , Ritmo Circadiano/efeitos da radiação , Luz , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Animais , Comportamento Animal , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Neurônios/efeitos da radiação , Esforço Físico/genética , Esforço Físico/efeitos da radiação , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/efeitos da radiação
5.
J Mol Neurosci ; 48(3): 584-96, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22622901

RESUMO

The circadian time-keeping system consists of clocks in the suprachiasmatic nucleus (SCN) and in peripheral organs including an adrenal clock linked to the rhythmic corticosteroid production by regulating steroidogenic acute regulatory protein (StAR). Clock cells contain an autonomous molecular oscillator based on a group of clock genes and their protein products. Mice lacking the VPAC2 receptor display disrupted circadian rhythm of physiology and behaviour, and therefore, we using real-time RT-PCR quantified (1) the mRNAs for the clock genes Per1 and Bmal1 in the adrenal gland and SCN, (2) the adrenal Star mRNA and (3) the serum corticosterone concentration both during a light/dark (L/D) cycle and at constant darkness in wild type (WT) and VPAC2 receptor-deficient mice (VPAC2-KO). We also examined if PER1 and StAR were co-localised in the adrenal steroidogenic cells. Per1 and Bmal1 mRNA showed a 24-h rhythmic expression in the adrenal of WT mice under L/D and dark conditions. During a L/D cycle, the adrenal clock gene rhythm in VPAC2-KO mice was phase-advanced by approximately 6 h compared to WT mice and became arrhythmic in constant darkness. A significant 24-h rhythmic variation in the adrenal Star mRNA expression and circulating corticosterone concentration was similarly phase-advanced during the L/D cycle. The loss of adrenal clock gene rhythm in the VPAC2 receptor knockout mice after transfer into constant darkness was accompanied by disappearance of rhythmicity in Star mRNA expression and serum corticosterone concentration. Double immunohistochemistry showed that the PER1 protein and StAR were co-localised in the same steroidogenic cells. Circulating corticosterone plays a role in the circadian timing system and the misaligned corticosterone rhythm in the VPAC2 receptor knockout mice could be involved in their abnormal rhythms of physiology.


Assuntos
Fatores de Transcrição ARNTL/genética , Córtex Suprarrenal/metabolismo , Ritmo Circadiano/fisiologia , Corticosterona/sangue , Regulação da Expressão Gênica/fisiologia , Proteínas Circadianas Period/genética , Fosfoproteínas/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Núcleo Supraquiasmático/metabolismo , Fatores de Transcrição ARNTL/biossíntese , Medula Suprarrenal/metabolismo , Animais , Ritmo Circadiano/genética , Ritmo Circadiano/efeitos da radiação , Corticosterona/metabolismo , Escuridão , Regulação da Expressão Gênica/efeitos da radiação , Hibridização In Situ , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Circadianas Period/biossíntese , Fosfoproteínas/biossíntese , RNA Mensageiro , Reação em Cadeia da Polimerase em Tempo Real , Receptores Tipo II de Peptídeo Intestinal Vasoativo/fisiologia
6.
Proc Natl Acad Sci U S A ; 108(34): 14306-11, 2011 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-21788520

RESUMO

The suprachiasmatic nucleus (SCN) is the principal circadian pacemaker of mammals, coordinating daily rhythms of behavior and metabolism. Circadian timekeeping in SCN neurons revolves around transcriptional/posttranslational feedback loops, in which Period (Per) and Cryptochrome (Cry) genes are negatively regulated by their protein products. Recent studies have revealed, however, that these "core loops" also rely upon cytosolic and circuit-level properties for sustained oscillation. To characterize interneuronal signals responsible for robust pacemaking in SCN cells and circuits, we have developed a unique coculture technique using wild-type (WT) "graft" SCN to drive pacemaking (reported by PER2::LUCIFERASE bioluminescence) in "host" SCN deficient either in elements of neuropeptidergic signaling or in elements of the core feedback loop. We demonstrate that paracrine signaling is sufficient to restore cellular synchrony and amplitude of pacemaking in SCN circuits lacking vasoactive intestinal peptide (VIP). By using grafts with mutant circadian periods we show that pacemaking in the host SCN is specified by the genotype of the graft, confirming graft-derived factors as determinants of the host rhythm. By combining pharmacological with genetic manipulations, we show that a hierarchy of neuropeptidergic signals underpins this paracrine regulation, with a preeminent role for VIP augmented by contributions from arginine vasopressin (AVP) and gastrin-releasing peptide (GRP). Finally, we show that interneuronal signaling is sufficiently powerful to maintain circadian pacemaking in arrhythmic Cry-null SCN, deficient in essential elements of the transcriptional negative feedback loops. Thus, a hierarchy of paracrine neuropeptidergic signals determines cell- and circuit-level circadian pacemaking in the SCN.


Assuntos
Ritmo Circadiano/fisiologia , Rede Nervosa/metabolismo , Comunicação Parácrina , Transdução de Sinais , Núcleo Supraquiasmático/metabolismo , Animais , Ritmo Circadiano/genética , Técnicas de Cocultura , Criptocromos/deficiência , Criptocromos/metabolismo , Regulação da Expressão Gênica , Camundongos , Comunicação Parácrina/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Transdução de Sinais/genética , Núcleo Supraquiasmático/citologia , Peptídeo Intestinal Vasoativo/deficiência , Peptídeo Intestinal Vasoativo/metabolismo
7.
Am J Physiol Regul Integr Comp Physiol ; 300(3): R519-30, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21178124

RESUMO

Neurons of the brain's biological clock located in the hypothalamic suprachiasmatic nucleus (SCN) generate circadian rhythms of physiology (core body temperature, hormone secretion, locomotor activity, sleep/wake, and heart rate) with distinct temporal phasing when entrained by the light/dark (LD) cycle. The neuropeptide vasoactive intestinal polypetide (VIP) and its receptor (VPAC2) are highly expressed in the SCN. Recent studies indicate that VIPergic signaling plays an essential role in the maintenance of ongoing circadian rhythmicity by synchronizing SCN cells and by maintaining rhythmicity within individual neurons. To further increase the understanding of the role of VPAC2 signaling in circadian regulation, we implanted telemetric devices and simultaneously measured core body temperature, spontaneous activity, and heart rate in a strain of VPAC2-deficient mice and compared these observations with observations made from mice examined by wheel-running activity. The study demonstrates that VPAC2 signaling is necessary for a functional circadian clock driving locomotor activity, core body temperature, and heart rate rhythmicity, since VPAC2-deficient mice lose the rhythms in all three parameters when placed under constant conditions (of either light or darkness). Furthermore, although 24-h rhythms for three parameters are retained in VPAC2-deficient mice during the LD cycle, the temperature rhythm displays markedly altered time course and profile, rising earlier and peaking ∼4-6 h prior to that of wild-type mice. The use of telemetric devices to measure circadian locomotor activity, temperature, and heart rate, together with the classical determination of circadian rhythms of wheel-running activity, raises questions about how representative wheel-running activity may be of other behavioral parameters, especially when animals have altered circadian phenotype.


Assuntos
Regulação da Temperatura Corporal , Ritmo Circadiano , Frequência Cardíaca , Atividade Motora , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Transdução de Sinais , Núcleo Supraquiasmático/metabolismo , Animais , Feminino , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Fotoperíodo , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Corrida , Telemetria , Fatores de Tempo
8.
J Biol Rhythms ; 25(4): 235-46, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20679493

RESUMO

Daily exercise promotes physical health as well as improvements in mental and neural functions. Studies in intact wild-type (WT) rodents have revealed that the brain's suprachiasmatic nuclei (SCN), site of the main circadian pacemaker, are also responsive to scheduled wheel running. It is unclear, however, if and how animals with a dysfunctional circadian pacemaker respond to exercise. Here, we tested whether scheduled voluntary exercise (SVE) in a running wheel for 6 hours per day could promote neural and behavioral rhythmicity in animals whose circadian competence is compromised through genetically targeted loss of vasoactive intestinal polypeptide (VIP(-/-) mice) or its VPAC(2) receptor (Vipr2(-/-) mice). We report that in constant dark (DD), rhythmic VIP(-/-) and Vipr2(-/-) mice show weak free-running rhythms with a period of <23 hours and all wild-type mice are strongly rhythmic with approximately 23.5-hour periodicity. VIP(-/-) and Vipr2(-/-) mice rapidly (<7 days) synchronize to daily SVE, while WT mice take much longer (>35 days). Following 21 to 50 days of SVE, WT mice show small changes in their rhythms, and most Vipr2(-/-) mice now sustain robust near 24-hour behavioral rhythms, whereas very few VIP(-/-) mice do. This study demonstrates that scheduled daily exercise can markedly improve circadian rhythms in behavioral activity and raises the possibility that this noninvasive approach may be useful as an intervention in clinical etiologies in which there are dysfunctions of circadian time keeping.


Assuntos
Ritmo Circadiano/genética , Atividade Motora/fisiologia , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Peptídeo Intestinal Vasoativo/deficiência , Animais , Ritmo Circadiano/fisiologia , Escuridão , Camundongos , Proteínas Proto-Oncogênicas c-fos/biossíntese , Núcleo Supraquiasmático/fisiologia
9.
Proc Natl Acad Sci U S A ; 107(34): 15240-5, 2010 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-20696890

RESUMO

Circadian pacemaking requires the orderly synthesis, posttranslational modification, and degradation of clock proteins. In mammals, mutations in casein kinase 1 (CK1) epsilon or delta can alter the circadian period, but the particular functions of the WT isoforms within the pacemaker remain unclear. We selectively targeted WT CK1epsilon and CK1delta using pharmacological inhibitors (PF-4800567 and PF-670462, respectively) alongside genetic knockout and knockdown to reveal that CK1 activity is essential to molecular pacemaking. Moreover, CK1delta is the principal regulator of the clock period: pharmacological inhibition of CK1delta, but not CK1epsilon, significantly lengthened circadian rhythms in locomotor activity in vivo and molecular oscillations in the suprachiasmatic nucleus (SCN) and peripheral tissue slices in vitro. Period lengthening mediated by CK1delta inhibition was accompanied by nuclear retention of PER2 protein both in vitro and in vivo. Furthermore, phase mapping of the molecular clockwork in vitro showed that PF-670462 treatment lengthened the period in a phase-specific manner, selectively extending the duration of PER2-mediated transcriptional feedback. These findings suggested that CK1delta inhibition might be effective in increasing the amplitude and synchronization of disrupted circadian oscillators. This was tested using arrhythmic SCN slices derived from Vipr2(-/-) mice, in which PF-670462 treatment transiently restored robust circadian rhythms of PER2::Luc bioluminescence. Moreover, in mice rendered behaviorally arrhythmic by the Vipr2(-/-) mutation or by constant light, daily treatment with PF-670462 elicited robust 24-h activity cycles that persisted throughout treatment. Accordingly, selective pharmacological targeting of the endogenous circadian regulator CK1delta offers an avenue for therapeutic modulation of perturbed circadian behavior.


Assuntos
Caseína Quinase 1 épsilon/antagonistas & inibidores , Caseína Quinase Idelta/antagonistas & inibidores , Ritmo Circadiano/fisiologia , Animais , Sequência de Bases , Caseína Quinase 1 épsilon/fisiologia , Caseína Quinase Idelta/deficiência , Caseína Quinase Idelta/genética , Caseína Quinase Idelta/fisiologia , Ritmo Circadiano/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Técnicas In Vitro , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Circadianas Period/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Núcleo Supraquiasmático/efeitos dos fármacos , Núcleo Supraquiasmático/fisiologia
10.
Peptides ; 31(6): 1068-75, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20226823

RESUMO

Allergic asthma is a T(H)2-mediated disease marked by airway inflammation, increased mucus production, and elevated serum IgE in response to allergen provocation. Among its ascribed functions, the neuropeptide vasoactive intestinal peptide (VIP) is believed to promote a T(H)2 phenotype when signaling through its VPAC(2) receptor. In this study, we assessed the requirement for the VIP/VPAC(2) axis in initiating the allergic pulmonary phenotype in a murine model of fungal allergic asthma. C57BL/6 wild-type (WT) and VPAC(2) knock-out (KO) mice were sensitized with Aspergillus fumigatus antigen and challenged with an aerosol of live conidia to induce allergic airways disease. WT and KO mice exhibited similar peribronchovascular inflammation, increased number of goblet cells, and elevated serum IgE. However, the absence of VPAC(2) receptor resulted in a marked enhancement of MUC5AC mRNA with an associated increase in goblet cells and a reduction in eosinophils in the airway lumen at day 3 when VIP mRNA was undetectable in the KO lung. Sustained elevation of serum IgE was noted in KO mice at day 14, while the level in WT mice declined at this time point. These data suggest that the absence of VPAC(2) does not protect mice from developing the signs and symptoms of allergic asthma.


Assuntos
Asma/imunologia , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Animais , Antígenos de Fungos/imunologia , Aspergillus/imunologia , Asma/patologia , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/patologia , Células Caliciformes/patologia , Metaplasia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucina-5AC/biossíntese , Receptores Tipo II de Peptídeo Intestinal Vasoativo/fisiologia
11.
Neuropharmacology ; 55(8): 1329-39, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18804119

RESUMO

Peptide histidine isoleucine (PHI) and vasoactive intestinal peptide (VIP) are neuropeptides synthesized from a common precursor, prepro-VIP, and share structural similarity and biological functions in many systems. Within the central nervous system and peripheral tissues, PHI and VIP have overlapping distribution. PHI-mediated functions are generally via activation of VIP receptors; however, the potency and affinity of PHI for VIP receptors are significantly lower than VIP. In addition, several studies suggest distinct PHI receptors that are independent of VIP receptors. PHI receptors have been cloned and characterized in fish, but their existence in mammals is still unknown. This study focuses on the functional role of PHI in the thalamus because of the localization of both PHI and VIP receptors in this brain region. Using extracellular multiple-unit recording techniques, we found that PHI strongly attenuated the slow intrathalamic rhythmic activity. Using intracellular recording techniques, we found that PHI selectively depolarized thalamic relay neurons via an enhancement of the hyperpolarization-activated mixed cation current, Ih. Further, the actions of PHI were occluded by VIP and dopamine, indicating these modulators converge onto a common mechanism. In contrast to previous work, we found that PHI was more potent than VIP in producing excitatory actions on thalamic neurons. We next used the transgenic mice lacking a specific VIP receptor, VPAC2, to identify its possible role in PHI-mediated actions in the thalamus. PHI depolarized all relay neurons tested from wild-type mice (VPAC2(+/+)); however, in knockout mice (VPAC2(-/-)), PHI produced no change in membrane potential in all neurons tested. Our findings indicate that excitatory actions of PHI are mediated by VPAC2 receptors, not by its own PHI receptors and the excitatory actions of PHI clearly attenuate intrathalamic rhythmic activities, and likely influence information transfer through thalamocortical circuits.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Interneurônios/efeitos dos fármacos , Peptídeo PHI/farmacologia , Tálamo/citologia , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Biofísica , Dopamina/farmacologia , Relação Dose-Resposta a Droga , Estimulação Elétrica , Técnicas In Vitro , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp/métodos , Periodicidade , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Peptídeo Intestinal Vasoativo/farmacologia
12.
J Neurochem ; 106(4): 1646-57, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18554318

RESUMO

Vasoactive intestinal polypeptide and its receptor, VPAC(2), play important roles in the functioning of the brain's circadian clock in the suprachiasmatic nuclei (SCN). Mice lacking VPAC(2) receptors (Vipr2(-/-)) show altered circadian rhythms in locomotor behavior, neuronal firing rate, and clock gene expression, however, the nature of molecular oscillations in individual cells is unclear. Here, we used real-time confocal imaging of a destabilized green fluorescent protein (GFP) reporter to track the expression of the core clock gene Per1 in live SCN-containing brain slices from wild-type (WT) and Vipr2(-/-) mice. Rhythms in Per1-driven GFP were detected in WT and Vipr2(-/-) cells, though a significantly lower number and proportion of cells in Vipr2(-/-) slices expressed detectable rhythms. Further, Vipr2(-/-) cells expressed significantly lower amplitude oscillations than WT cells. Within each slice, the phases of WT cells were synchronized whereas cells in Vipr2(-/-) slices were poorly synchronized. Most GFP-expressing cells, from both genotypes, expressed neither vasopressin nor vasoactive intestinal polypeptide. Pharmacological blockade of VPAC(2) receptors in WT SCN slices partially mimicked the Vipr2(-/-) phenotype. These data demonstrate that intercellular communication via the VPAC(2) receptor is important for SCN neurons to sustain robust, synchronous oscillations in clock gene expression.


Assuntos
Proteínas do Olho/biossíntese , Regulação da Expressão Gênica/fisiologia , Receptores Tipo II de Peptídeo Intestinal Vasoativo/biossíntese , Núcleo Supraquiasmático/metabolismo , Animais , Células Cultivadas , Proteínas do Olho/análise , Proteínas do Olho/genética , Feminino , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal/métodos , Atividade Motora/fisiologia , Proteínas Circadianas Period , Receptores Tipo II de Peptídeo Intestinal Vasoativo/análise , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/fisiologia , Núcleo Supraquiasmático/química
13.
Artigo em Inglês | MEDLINE | ID: mdl-18419265

RESUMO

A hierarchy of interacting, tissue-based clocks controls circadian physiology and behavior in mammals. Preeminent are the suprachiasmatic nuclei (SCN): central hypothalamic pacemakers synchronized to solar time via retinal afferents and in turn responsible for internal synchronization of other clocks present in major organ systems. The SCN and peripheral clocks share essentially the same cellular timing mechanism. This consists of autoregulatory transcriptional/posttranslational feedback loops in which the Period (Per) and Cryptochrome (Cry) "clock" genes are negatively regulated by their protein products. Here, we review recent studies directed at understanding the molecular and cellular bases to the mammalian clock. At the cellular level, we demonstrate the role of F-box protein Fbxl3 (characterized by the afterhours mutation) in directing the proteasomal degradation of Cry and thereby controlling negative feedback and circadian period of the molecular loops. Within SCN neural circuitry, we describe how neuropeptidergic signaling by VIP synchronizes and sustains the cellular clocks. At the hypothalamic level, signaling via a different SCN neuropeptide, prokineticin, is not required for pacemaking but is necessary for control of circadian behavior. Finally, we consider how metabolic pathways are coordinated in time, focusing on liver function and the role of glucocorticoid signals in driving the circadian transcriptome and proteome.


Assuntos
Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Animais , Perfilação da Expressão Gênica , Fígado/fisiologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Mutação , Neuropeptídeos/genética , Neuropeptídeos/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteoma , Receptores Tipo II de Peptídeo Intestinal Vasoativo/deficiência , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Transdução de Sinais , Núcleo Supraquiasmático/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA