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

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Circadian Rhythms ; 16: 13, 2018 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-30473715

RESUMO

In mammals, the master pacemaker driving circadian rhythms is thought to reside in the suprachiasmatic nuclei (SCN) of the anterior hypothalamus. A clear view of molecular clock mechanisms within the SCN neurons has been elucidated. In contrast, much less is known about the output mechanism by which the SCN circadian pacemaker sends timing information for eventual control of physiological and behavioral rhythms. Two secreted molecules, prokineticin 2 (PK2) and vasopressin, that are encoded by respective clock-controlled genes, have been indicated as candidate SCN output molecules. Several lines of evidence have emerged that support the role of PK2 as an output signal for the SCN circadian clock, including the reduced circadian rhythms in mice that are deficient in PK2 or its receptor, PKR2. In the current study, transgenic mice with the overexpression of PK2 have been generated. These transgenic mice displayed reduced oscillation of the PK2 expression in the SCN and decreased amplitude of circadian locomotor rhythm, supporting the important signaling role of PK2 in the regulation of circadian rhythms. Altered molecular rhythms were also observed in the SCN in the transgenic mice, indicating that PK2 signaling also regulates the operation of core clockwork. This conclusion is consistent with recent reports showing the likely signaling role of PK2 from the intrinsically photosensitive retinal ganglion cells to SCN neurons. Thus, PK2 signaling plays roles in both the input and the output pathways of the SCN circadian clock.

2.
J Biol Chem ; 289(22): 15518-26, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24753254

RESUMO

Mutations in the G protein-coupled prokineticin receptor 2 (PKR2) are known to cause Kallmann syndrome and idiopathic hypogonadotropic hypogonadism manifesting with delayed puberty and infertility. Some of the mutant receptors are not routed to the cell surface; instead, they are trapped in the cellular secretory pathway. The cell-permeant agonists/antagonists have been used to rescue some membrane receptors that are not targeted onto the cell membrane. Here, we chose three disease-associated mutations (W178S, G234D, and P290S), which all resulted in retention of PKR2 intracellularly. We show that a small molecule PKR2 antagonist (A457) dramatically increased cell surface expression and rescued the function of P290S PKR2, but had no effect on W178S and G234D PKR2. Furthermore, we also tested chemical chaperone glycerol on the cell surface expression and function of PKR2 mutants. Treatment with 10% glycerol significantly increased the cell surface expression and signaling of P290S and W178S PKR2. These data demonstrate that some Kallmann syndrome-associated, intracellularly retained mutant PKR2 receptors can be functionally rescued, suggesting a potential treatment strategy for patients bearing such mutations.


Assuntos
Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Síndrome de Kallmann/genética , Síndrome de Kallmann/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Animais , Células CHO , Cricetulus , Crioprotetores/farmacologia , Glicerol/farmacologia , Células HEK293 , Compostos Heterocíclicos de 4 ou mais Anéis/síntese química , Humanos , Síndrome de Kallmann/tratamento farmacológico , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação Puntual , Transporte Proteico/genética , Deficiências na Proteostase/genética , Deficiências na Proteostase/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de Peptídeos/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
3.
Am J Physiol Endocrinol Metab ; 309(4): E357-69, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26081281

RESUMO

PPARγ-deficient mice die at E9.5 due to placental abnormalities. The mechanism by which this occurs is unknown. We demonstrated that the new endocrine factor EG-VEGF controls the same processes as those described for PPARγ, suggesting potential regulation of EG-VEGF by PPARγ. EG-VEGF exerts its functions via prokineticin receptor 1 (PROKR1) and 2 (PROKR2). This study sought to investigate whether EG-VEGF mediates part of PPARγ effects on placental development. Three approaches were used: 1) in vitro, using human primary isolated cytotrophoblasts and the extravillous trophoblast cell line (HTR-8/SVneo); 2) ex vivo, using human placental explants (n = 46 placentas); and 3) in vivo, using gravid wild-type PPARγ(+/-) and PPARγ(-/-) mice. Major processes of placental development that are known to be controlled by PPARγ, such as trophoblast proliferation, migration, and invasion, were assessed in the absence or presence of PROKR1 and PROKR2 antagonists. In both human trophoblast cell and placental explants, we demonstrated that rosiglitazone, a PPARγ agonist, 1) increased EG-VEGF secretion, 2) increased EG-VEGF and its receptors mRNA and protein expression, 3) increased placental vascularization via PROKR1 and PROKR2, and 4) inhibited trophoblast migration and invasion via PROKR2. In the PPARγ(-/-) mouse placentas, EG-VEGF levels were significantly decreased, supporting an in vivo control of EG-VEGF/PROKRs system during pregnancy. The present data reveal EG-VEGF as a new mediator of PPARγ effects during pregnancy and bring new insights into the fine mechanism of trophoblast invasion.


Assuntos
PPAR gama/fisiologia , Placentação , Resultado da Gravidez/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Animais , Benzamidas/farmacologia , Células Cultivadas , Cricetinae , Implantação do Embrião/efeitos dos fármacos , Implantação do Embrião/genética , Embrião de Mamíferos , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , Placenta/metabolismo , Gravidez , Piridinas/farmacologia , Rosiglitazona , Tiazolidinedionas/farmacologia , Ativação Transcricional/efeitos dos fármacos , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo
4.
Proc Natl Acad Sci U S A ; 109(14): 5475-80, 2012 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-22431614

RESUMO

Stroke causes brain dysfunction and neuron death, and the lack of effective therapies heightens the need for new therapeutic targets. Here we identify prokineticin 2 (PK2) as a mediator for cerebral ischemic injury. PK2 is a bioactive peptide initially discovered as a regulator of gastrointestinal motility. Multiple biological roles for PK2 have been discovered, including circadian rhythms, angiogenesis, and neurogenesis. However, the role of PK2 in neuropathology is unknown. Using primary cortical cultures, we found that PK2 mRNA is up-regulated by several pathological stressors, including hypoxia, reactive oxygen species, and excitotoxic glutamate. Glutamate-induced PK2 expression is dependent on NMDA receptor activation and extracellular calcium. Enriched neuronal culture studies revealed that neurons are the principal source of glutamate-induced PK2. Using in vivo models of stroke, we found that PK2 mRNA is induced in the ischemic cortex and striatum. Central delivery of PK2 worsens infarct volume, whereas PK2 receptor antagonist decreases infarct volume and central inflammation while improving functional outcome. Direct central inhibition of PK2 using RNAi also reduces infarct volume. These findings indicate that PK2 can be activated by pathological stimuli such as hypoxia-ischemia and excitotoxic glutamate and identify PK2 as a deleterious mediator for cerebral ischemia.


Assuntos
Isquemia Encefálica/fisiopatologia , Hormônios Gastrointestinais/fisiologia , Neuropeptídeos/fisiologia , Animais , Hormônios Gastrointestinais/genética , Neuropeptídeos/genética , RNA Mensageiro/genética , Ratos , Regulação para Cima
5.
J Circadian Rhythms ; 13: 2, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27103928

RESUMO

The mammalian circadian clock is composed of single-cell oscillators. Neurochemical and electrical signaling among these oscillators is important for the normal expression of circadian rhythms. Prokineticin 2 (PK2), encoding a cysteine-rich secreted protein, has been shown to be a critical signaling molecule for the regulation of circadian rhythms. PK2 expression in the suprachiasmatic nucleus (SCN) is highly rhythmic, peaking during the day and being essentially absent during the night. Mice with disrupted PK2 gene or its receptor PKR2 display greatly reduced rhythmicity of broad circadian parameters such as locomotor activity, body temperature and sleep/wake patterns. PK2 has been shown to increase the firing rate of SCN neurons, with unknown molecular mechanisms. Here we report that TRPV2, an ion channel belonging to the family of TRP, is co-expressed with PKR2 in the SCN neurons. Further, TRPV2 protein, but not TRPV2 mRNA, was shown to oscillate in the SCN in a PK2-dependent manner. Functional studies revealed that TRPV2 enhanced signaling of PKR2 in calcium mobilization or ion current conductance, likely via the increased trafficking of TRPV2 to the cell surface. Taken together, these results indicate that TRPV2 is likely part of the downstream signaling of PK2 in the regulation of the circadian rhythms.

6.
J Biol Chem ; 288(37): 26865-77, 2013 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-23873935

RESUMO

MicroRNAs (miRNAs) are small, single-stranded, noncoding RNAs that function as negative regulators of gene expression. They are transcribed from endogenous DNA and form hairpin structures (termed as pre-miRNAs) that are processed to form mature miRNAs. It remains largely unknown as to the molecular consequences of the natural genetic variation in pre-miRNAs. Here, we report that an A→G polymorphism (rs71428439) is located in Homo sapiens miR-149 stem-loop region. This polymorphism results in a change in the structure of the miR-149 precursor. Our results showed that the genotype distribution of this polymorphism in myocardial infarction cases was significantly different from that in the control subjects. We examined the biological significance of this polymorphism on the production of mature miR-149, and we observed that the G-allelic miR-149 precursor displayed a lower production of mature miR-149 compared with the A-allelic one. Further investigations disclosed that miR-149 could withstand mitochondrial fission and apoptosis through targeting the pro-apoptotic factor p53-up-regulated modulator of apoptosis (Puma). Enforced expression of miR-149 promoted cell survival, whereas knockdown of miR-149 rendered cells to be sensitive to apoptotic stimulation. Intriguingly, the A to G variation led pre-miR-149 to elicit an attenuated effect on the inhibition of mitochondrial fission and apoptosis. Finally, this polymorphism exerts its influence on cardiac function in the mouse model of myocardial infarction. These data suggest that this polymorphism in the miR-149 precursor may result in important phenotypic traits of myocardial infarction. Our findings warrant further investigations on the relationship between miR-149 polymorphism and myocardial infarction.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , MicroRNAs/genética , Infarto do Miocárdio/genética , Polimorfismo de Nucleotídeo Único , Proteínas Proto-Oncogênicas/metabolismo , Adulto , Idoso , Animais , Doenças Cardiovasculares/metabolismo , Caspase 3/metabolismo , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Predisposição Genética para Doença , Vetores Genéticos , Genótipo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Miócitos Cardíacos/citologia , Fenótipo , Interferência de RNA
7.
J Neuroinflammation ; 9: 108, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22642848

RESUMO

BACKGROUND: Prokineticin 2 (PK2) is a secreted protein and causes potent hyperalgesia in vivo, and is therefore considered to be a new pronociceptive mediator. However, the molecular targets responsible for the pronociceptive effects of PK2 are still poorly understood. Here, we have found that PK2 potentiates the activity of acid-sensing ion channels in the primary sensory neurons. METHODS: In the present study, experiments were performed on neurons freshly isolated from rat dorsal root ganglion by using whole-cell patch clamp and voltage-clamp recording techniques. RESULTS: PK2 dose-dependently enhanced proton-gated currents with an EC50 of 0.22 ± 0.06 nM. PK2 shifted the proton concentration-response curve upwards, with a 1.81 ± 0.11 fold increase of the maximal current response. PK2 enhancing effect on proton-gated currents was completely blocked by PK2 receptor antagonist. The potentiation was also abolished by intracellular dialysis of GF109203X, a protein kinase C inhibitor, or FSC-231, a protein interacting with C-kinase 1 inhibitor. Moreover, PK2 enhanced the acid-evoked membrane excitability of rat dorsal root ganglion neurons and caused a significant increase in the amplitude of the depolarization and the number of spikes induced by acid stimuli. Finally, PK2 exacerbated nociceptive responses to the injection of acetic acid in rats. CONCLUSION: These results suggest that PK2 increases the activity of acid-sensing ion channels via the PK2 receptor and protein kinase C-dependent signal pathways in rat primary sensory neurons. Our findings support that PK2 is a proalgesic factor and its signaling likely contributes to acidosis-evoked pain by sensitizing acid-sensing ion channels.


Assuntos
Canais Iônicos Sensíveis a Ácido/fisiologia , Gânglios Espinais/metabolismo , Hormônios Gastrointestinais/fisiologia , Neurônios/química , Neuropeptídeos/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Peptídeos/fisiologia , Canais Iônicos Sensíveis a Ácido/metabolismo , Animais , Sinergismo Farmacológico , Gânglios Espinais/enzimologia , Gânglios Espinais/fisiologia , Hormônios Gastrointestinais/química , Masculino , Neurônios/enzimologia , Neurônios/metabolismo , Neuropeptídeos/química , Proteína Quinase C/química , Proteína Quinase C/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Receptores de Peptídeos/agonistas , Receptores de Peptídeos/química , Transdução de Sinais/fisiologia
8.
Biomedicines ; 9(3)2021 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-33802771

RESUMO

Endocrine gland derived vascular endothelial growth factor (EG-VEGF) is a canonical member of the prokineticin (PROKs) family. It acts via the two G-protein coupled receptors, namely PROKR1 and PROKR2. We have recently demonstrated that EG-VEGF is highly expressed in the human placenta; contributes to placental vascularization and growth and that its aberrant expression is associated with pregnancy pathologies including preeclampsia and fetal growth restriction. These findings strongly suggested that antagonization of its receptors may constitute a potential therapy for the pregnancy pathologies. Two specific antagonists of PROKR1 (PC7) and for PROKR2 (PKRA) were reported to reverse PROKs adverse effects in other systems. In the view of using these antagonists to treat pregnancy pathologies, a proof of concept study was designed to determine the biological significances of PC7 and PKRA in normal pregnancy outcome. PC7 and PKRA were tested independently or in combination in trophoblast cells and during early gestation in the gravid mouse. Both independent and combined treatments uncovered endogenous functions of EG-VEGF. The independent use of antagonists distinctively identified PROKR1 and PROKR2-mediated EG-VEGF signaling on trophoblast differentiation and invasion; thereby enhancing feto-placental growth and pregnancy outcome. Thus, our study provides evidence for the potential safe use of PC7 or PKRA to improve pregnancy outcome.

9.
Epilepsy Behav ; 14 Suppl 1: 74-80, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18849007

RESUMO

Neurogenesis persists in several regions of the adult mammalian brain. Although the hippocampus and olfactory bulb are most commonly studied in the context of adult neurogenesis, there is an increasing body of evidence in support of neurogenesis occurring outside of these two regions. The current study expands on previous data by showing newborn neurons with a mature phenotype are located in several olfactory and limbic structures outside of the hippocampus and olfactory bulb, where we previously described doublecortin/bromodeoxyuridine immature neurons. Notably, newborn neurons with a mature neuronal phenotype are found in the olfactory tubercles, anterior olfactory nuclei, tenia tecta, islands of Calleja, amygdala, and lateral entorhinal cortex. The appearance of newborn neurons with a mature phenotype in these regions suggests that these structures are destinations, and that newborn neurons are not simply passing through these structures. In light of the increasing body of evidence for neurogenesis in these and other olfactory, limbic, and striatal structures, we hypothesize that brain regions displaying adult neurogenesis are functionally linked.


Assuntos
Sistema Límbico/citologia , Neurônios/fisiologia , Bulbo Olfatório/citologia , Prosencéfalo/fisiologia , Animais , Animais Recém-Nascidos , Antimetabólitos , Bromodesoxiuridina , Proliferação de Células , Proteínas de Ligação a DNA , Lateralidade Funcional/fisiologia , Imuno-Histoquímica , Sistema Límbico/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Prosencéfalo/citologia
10.
J Clin Endocrinol Metab ; 93(9): 3551-9, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18559922

RESUMO

CONTEXT: Mice deficient in prokineticin 2(PROK2) and prokineticin receptor2 (PROKR2) exhibit variable olfactory bulb dysgenesis and GnRH neuronal migration defects reminiscent of human GnRH deficiency. OBJECTIVES: We aimed to screen a large cohort of patients with Kallmann syndrome (KS) and normosmic idiopathic hypogonadotropic hypogonadism (IHH) for mutations in PROK2/PROKR2, evaluate their prevalence, define the genotype/phenotype relationship, and assess the functionality of these mutant alleles in vitro. DESIGN: Sequencing of the PROK2 and PROKR2 genes was performed in 170 KS patients and 154 nIHH. Mutations were examined using early growth response 1-luciferase assays in HEK 293 cells and aequorin assays in Chinese hamster ovary cells. RESULTS: Four heterozygous and one homozygous PROK2 mutation (p.A24P, p.C34Y, p.I50M, p.R73C, and p.I55fsX1) were identified in five probands. Four probands had KS and one nIHH, and all had absent puberty. Each mutant peptide impaired receptor signaling in vitro except the I50M. There were 11 patients who carried a heterozygous PROKR2 mutation (p.R85C, p.Y113H, p.V115M, p.R164Q, p.L173R, p.W178S, p.S188L, p.R248Q, p.V331M, and p.R357W). Among them, six had KS, four nIHH, and one KS proband carried both a PROKR2 (p.V115M) and PROK2 (p.A24P) mutation. Reproductive phenotypes ranged from absent to partial puberty to complete reversal of GnRH deficiency after discontinuation of therapy. All mutant alleles appear to decrease intracellular calcium mobilization; seven exhibited decreased MAPK signaling, and six displayed decreased receptor expression. Nonreproductive phenotypes included fibrous dysplasia, sleep disorder, synkinesia, and epilepsy. Finally, considerable variability was evident in family members with the same mutation, including asymptomatic carriers. CONCLUSION: Loss-of-function mutations in PROK2 and PROKR2 underlie both KS and nIHH.


Assuntos
Hormônios Gastrointestinais/genética , Hormônio Liberador de Gonadotropina/deficiência , Hipogonadismo/genética , Mutação de Sentido Incorreto , Neuropeptídeos/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Adolescente , Equorina/genética , Animais , Células CHO , Cricetinae , Cricetulus , Análise Mutacional de DNA , Feminino , Frequência do Gene , Heterogeneidade Genética , Genótipo , Humanos , Síndrome de Kallmann/genética , Masculino , Modelos Biológicos , Linhagem , Transfecção
11.
J Neurosci ; 26(45): 11615-23, 2006 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-17093083

RESUMO

Circadian clocks drive daily rhythms in virtually all organisms. In mammals, the suprachiasmatic nucleus (SCN) is recognized as the master clock that synchronizes central and peripheral oscillators to evoke circadian rhythms of diverse physiology and behavior. How the timing information is transmitted from the SCN clock to generate overt circadian rhythms is essentially unknown. Prokineticin 2 (PK2), a clock-controlled gene that encodes a secreted protein, has been indicated as a candidate SCN clock output signal that regulates circadian locomotor rhythm. Here we report the generation and analysis of PK2-null mice. The reduction of locomotor rhythms in PK2-null mice was apparent in both hybrid and inbred genetic backgrounds. PK2-null mice also displayed significantly reduced rhythmicity for a variety of other physiological and behavioral parameters, including sleep-wake cycle, body temperature, circulating glucocorticoid and glucose levels, as well as the expression of peripheral clock genes. In addition, PK2-null mice showed accelerated acquisition of food anticipatory activity during a daytime food restriction. We conclude that PK2, acting as a SCN output factor, is important for the maintenance of robust circadian rhythms.


Assuntos
Ritmo Circadiano/genética , Hormônios Gastrointestinais/fisiologia , Neuropeptídeos/fisiologia , Núcleo Supraquiasmático/metabolismo , Análise de Variância , Animais , Comportamento Animal , Glicemia/genética , Temperatura Corporal/genética , Corticosterona/sangue , Criptocromos , Eletroencefalografia/métodos , Eletromiografia/métodos , Flavoproteínas/genética , Flavoproteínas/metabolismo , Privação de Alimentos/fisiologia , Hormônios Gastrointestinais/deficiência , Regulação da Expressão Gênica/fisiologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/fisiologia , Atividade Motora/genética , Neuropeptídeos/deficiência , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Sono/genética , Fatores de Tempo , Vigília/genética
12.
Sleep ; 30(3): 247-56, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17425220

RESUMO

STUDY OBJECTIVES: Sleep is regulated by circadian and homeostatic processes. Recent studies with mutant mice have indicated that circadian-related genes regulate sleep amount, as well as the timing of sleep. Thus a direct link between circadian and homeostatic regulation of sleep may exist, at least at the molecular level. Prokineticin 2 (PK2), which oscillates daily with high amplitude in the suprachiasmatic nuclei (SCN), has been postulated to be an SCN output molecule. In particular, mice lacking the PK2 gene (PK2-/-) have been shown to display significantly reduced rhythmicity for a variety of circadian physiological and behavioral parameters. We investigated the role of PK2 in sleep regulation. DESIGN: EEG/EMG sleep-wake patterns were recorded in PK2-/- mice and their wild-type littermate controls under baseline and challenged conditions. MEASUREMENTS AND RESULTS: PK2-/- mice exhibited reduced total sleep time under entrained light-dark and constant darkness conditions. The reduced sleep time in PK2-/- mice occurred predominantly during the light period and was entirely due to a decrease in non-rapid eye movement (NREM) sleep time. However, PK2-/- mice showed increased rapid eye movement (REM) sleep time in both light and dark periods. After sleep deprivation, compensatory rebound in NREM sleep, REM sleep, and EEG delta power was attenuated in PK2-/- mice. In addition, PK2-/- mice had an impaired response to sleep disturbance caused by cage change in the light phase. CONCLUSIONS: These results indicate that PK2 plays roles in both circadian and homeostatic regulation of sleep. PK2 may also be involved in maintaining the awake state in the presence of behavioral challenges.


Assuntos
Ritmo Circadiano/genética , Hormônios Gastrointestinais/deficiência , Hormônios Gastrointestinais/genética , Homeostase/genética , Neuropeptídeos/deficiência , Neuropeptídeos/genética , Fases do Sono/genética , Animais , Nível de Alerta/genética , Nível de Alerta/fisiologia , Ritmo Circadiano/fisiologia , Eletroencefalografia , Eletromiografia , Homeostase/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Privação do Sono/genética , Privação do Sono/fisiopatologia , Fases do Sono/fisiologia , Meio Social , Núcleo Supraquiasmático/fisiopatologia , Vigília/genética , Vigília/fisiologia
13.
Neuroreport ; 18(10): 981-5, 2007 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-17558281

RESUMO

Neurogenesis persists in the adult rodent olfactory epithelium and olfactory bulbs. Recent studies suggest that neurogenesis might also occur in the adult rodent piriform cortex, the primary cortical projection site of the olfactory bulbs. To determine whether olfactory enrichment influences neurogenesis in the mouse piriform cortex, olfactory enrichment was used in combination with bromodeoxyuridine labeling. Quantification of the number of bromodeoxyuridine-labeled cells in the piriform cortex that double label for either the immature neuronal marker, doublecortin, or the mature neuronal marker, neuronal nuclei or NeuN, showed that olfactory enrichment increases the survival of newborn neurons in the piriform cortex. These results confirm that neurogenesis occurs in the piriform cortex of rodents and suggest that it may play a neuroplastic role there.


Assuntos
Córtex Cerebral/citologia , Neurônios/fisiologia , Odorantes , Condutos Olfatórios/fisiologia , Organogênese/fisiologia , Animais , Mapeamento Encefálico , Bromodesoxiuridina/metabolismo , Contagem de Células/métodos , Diferenciação Celular/fisiologia , Proteínas do Domínio Duplacortina , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Bulbo Olfatório/fisiologia , Fosfopiruvato Hidratase/metabolismo , Fatores de Tempo
14.
Mol Pain ; 2: 35, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17107623

RESUMO

Prokineticins (PKs), consisting of PK1 and PK2, are a pair of newly identified regulatory peptides. Two closely related G-protein coupled receptors, PKR1 and PKR2, mediate the signaling of PKs. PKs/PKRs participate in the regulation of diverse biological processes, ranging from development to adult physiology. A number of studies have indicated the involvement of PKs/PKRs in nociception. Here we show that PK2 is a sensitizer for nociception. Intraplantar injection of recombinant PK2 resulted in a strong and localized hyperalgesia with reduced thresholds to nociceptive stimuli. PK2 mobilizes calcium in dissociated dorsal root ganglion (DRG) neurons. Mice lacking the PK2 gene displayed strong reduction in nociception induced by thermal and chemical stimuli, including capsaicin. However, PK2 mutant mice showed no difference in inflammatory response to capsaicin. As the majority of PK2-responsive DRG neurons also expressed transient receptor potential vanilloid (TRPV1) and exhibited sensitivity to capsaicin, TRPV1 is likely a significant downstream molecule of PK2 signaling. Taken together, these results reveal that PK2 sensitize nociception without affecting inflammation.


Assuntos
Hormônios Gastrointestinais/fisiologia , Hipestesia/genética , Hipestesia/fisiopatologia , Neuropeptídeos/fisiologia , Limiar da Dor/fisiologia , Animais , Comportamento Animal , Capsaicina , Gânglios Espinais/citologia , Hormônios Gastrointestinais/deficiência , Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/farmacologia , Hibridização In Situ/métodos , Inflamação/induzido quimicamente , Inflamação/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Neuropeptídeos/deficiência , Neuropeptídeos/genética , Neuropeptídeos/farmacologia , Dor/genética , Dor/fisiopatologia , Medição da Dor/métodos , Limiar da Dor/efeitos dos fármacos , Estimulação Física/métodos , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Estimulação Química , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
15.
J Comp Neurol ; 498(6): 796-809, 2006 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16927269

RESUMO

Prokineticins are a pair of regulatory peptides that have been shown to play important roles in gastrointestinal motility, angiogenesis, circadian rhythms, and, recently, olfactory bulb neurogenesis. Prokineticins exert their functions via activation of two closely related G-protein-coupled receptors. Here we report a comprehensive mRNA distribution for both prokineticins (PK1 and PK2) and their receptors (PKR1 and PKR2) in the adult mouse brain with the use of in situ hybridization. PK2 mRNA is expressed in discrete regions of the brain, including suprachiasmatic nucleus, islands of Calleja and medial preoptic area, olfactory bulb, nucleus accumbens shell, hypothalamic arcuate nucleus, and amygdala. PK1 mRNA is expressed exclusively in the brainstem, with high abundance in the nucleus tractus solitarius. PKR2 mRNA is detected throughout the brain, with prominent expression in olfactory regions, cortex, thalamus and hypothalamus, septum and hippocampus, habenula, amygdala, nucleus tractus solitarius, and circumventricular organs such as subfornical organ, median eminence, and area postrema. PKR2 mRNA is also detected in mammillary nuclei, periaqueductal gray, and dorsal raphe. In contrast, PKR1 mRNA is found in fewer brain regions, with moderate expression in the olfactory regions, dentate gyrus, zona incerta, and dorsal motor vagal nucleus. Both PKR1 and PKR2 are also detected in olfactory ventricle and subventricular zone of the lateral ventricle, both of which are rich sources of neuronal precursors. These extensive expression patterns suggest that prokineticins may have a broad array of functions in the central nervous system, including circadian rhythm, neurogenesis, ingestive behavior, reproduction, and autonomic function.


Assuntos
Encéfalo/metabolismo , Hormônios Gastrointestinais/biossíntese , Neuropeptídeos/biossíntese , Receptores Acoplados a Proteínas G/biossíntese , Receptores de Peptídeos/biossíntese , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/biossíntese , Animais , Expressão Gênica , Processamento de Imagem Assistida por Computador , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/análise
16.
Chronobiol Int ; 33(2): 191-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26818846

RESUMO

Prokineticin 2 (PK2) has been indicated as an output signaling molecule for the suprachiasmatic nucleus (SCN) circadian clock. Most of these studies were performed with nocturnal animals, particularly mice and rats. In the current study, the PK2 and its receptor, PKR2, was cloned from a species of diurnal macaque monkey. The macaque monkey PK2 and PKR2 were found to be highly homologous to that of other mammalian species. The mRNA expression of PK2 and PKR2 in the macaque brain was examined by in situ hybridization. The expression patterns of PK2 and PKR2 in the macaque brain were found to be quite similar to that of the mouse brain. Particularly, PK2 mRNA was shown to oscillate in the SCN of the macaque brain in the same phase and with similar amplitude with that of nocturnal mouse brain. PKR2 expression was also detected in known primary SCN targets, including the midline thalamic and hypothalamic nuclei. In addition, we detected the expression of PKR2 mRNA in the dorsal raphe nucleus (DR) of both macaque and mouse brains. As a likely SCN to dorsal raphe projection has previously been indicated, the expression of PKR2 in the raphe nuclei of both macaque and mouse brain signifies a possible role of DR as a previously unrecognized primary SCN projection target.


Assuntos
Relógios Biológicos/genética , Ritmo Circadiano/genética , Regulação da Expressão Gênica/fisiologia , Neuropeptídeos/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Hipotálamo/metabolismo , Hibridização In Situ/métodos , Luz , Macaca mulatta , RNA Mensageiro/metabolismo
17.
Mol Brain ; 9(1): 78, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27535380

RESUMO

The temporal organization of activity/rest or sleep/wake rhythms for mammals is regulated by the interaction of light/dark cycle and circadian clocks. The neural and molecular mechanisms that confine the active phase to either day or night period for the diurnal and the nocturnal mammals are unclear. Here we report that prokineticin 2, previously shown as a circadian clock output molecule, is expressed in the intrinsically photosensitive retinal ganglion cells, and the expression of prokineticin 2 in the intrinsically photosensitive retinal ganglion cells is oscillatory in a clock-dependent manner. We further show that the prokineticin 2 signaling is required for the activity and arousal suppression by light in the mouse. Between the nocturnal mouse and the diurnal monkey, a signaling receptor for prokineticin 2 is differentially expressed in the retinorecipient suprachiasmatic nucleus and the superior colliculus, brain projection targets of the intrinsically photosensitive retinal ganglion cells. Blockade with a selective antagonist reveals the respectively inhibitory and stimulatory effect of prokineticin 2 signaling on the arousal levels for the nocturnal mouse and the diurnal monkey. Thus, the mammalian diurnality or nocturnality is likely determined by the differential signaling of prokineticin 2 from the intrinsically photosensitive retinal ganglion cells onto their retinorecipient brain targets.


Assuntos
Nível de Alerta , Ritmo Circadiano , Hormônios Gastrointestinais/metabolismo , Neuropeptídeos/metabolismo , Transdução de Sinais , Animais , Nível de Alerta/efeitos da radiação , Relógios Biológicos/efeitos da radiação , Ritmo Circadiano/efeitos da radiação , Haplorrinos , Luz , Camundongos , Modelos Biológicos , Atividade Motora/efeitos da radiação , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/efeitos da radiação , Opsinas de Bastonetes/metabolismo , Transdução de Sinais/efeitos da radiação , Fatores de Tempo
18.
Nat Commun ; 7: 12932, 2016 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-27703142

RESUMO

Prokineticin-2 (PK2), a recently discovered secreted protein, regulates important physiological functions including olfactory biogenesis and circadian rhythms in the CNS. Interestingly, although PK2 expression is low in the nigral system, its receptors are constitutively expressed on nigrostriatal neurons. Herein, we demonstrate that PK2 expression is highly induced in nigral dopaminergic neurons during early stages of degeneration in multiple models of Parkinson's disease (PD), including PK2 reporter mice and MitoPark mice. Functional studies demonstrate that PK2 promotes mitochondrial biogenesis and activates ERK and Akt survival signalling pathways, thereby driving neuroprotection. Importantly, PK2 overexpression is protective whereas PK2 receptor antagonism exacerbates dopaminergic degeneration in experimental PD. Furthermore, PK2 expression increased in surviving nigral dopaminergic neurons from PD brains, indicating that PK2 upregulation is clinically relevant to human PD. Collectively, our results identify a paradigm for compensatory neuroprotective PK2 signalling in nigral dopaminergic neurons that could have important therapeutic implications for PD.


Assuntos
Sistema Nervoso Central/citologia , Neurônios Dopaminérgicos/metabolismo , Hormônios Gastrointestinais/metabolismo , Neuropeptídeos/metabolismo , Animais , Comportamento Animal , Morte Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hormônios Gastrointestinais/genética , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuropeptídeos/genética , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Reação em Cadeia da Polimerase , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Substância Negra/citologia
19.
J Neurosci ; 24(10): 2375-9, 2004 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-15014112

RESUMO

The recent discovery of prokineticin 2 (PK2) expression in the suprachiasmatic nucleus and its receptors in critical autonomic control centers of the brain, including the subfornical organ (SFO), suggests the intriguing possibility that PK2 regulates the excitability of SFO neurons and thus influences autonomic function. Using current-clamp techniques to record from dissociated SFO neurons, we examined the effects of PK2 on the excitability of these cells. PK2 (20 nm) induced depolarizations in 40% of SFO neurons (n = 45; mean, 7.5 +/- 1.7 mV), an effect that was reversible, PK2-specific, and concentration dependent. The depolarization was accompanied by an increase in action potential frequency from 0.4 +/- 0.1 to 1.4 +/- 0.5 Hz in responding cells (n = 10). This excitatory effect appears to be, in part, attributable to a PK2-induced decrease in the delayed rectifier potassium current (I(K)). In 10 SFO neurons recorded using perforated patch voltage-clamp techniques, six demonstrated a reversible decrease in I(K) (mean decrease, 26.7 +/- 6.4%) in response to 20 nm PK2, whereas artificial CSF alone was without an effect on these currents. These data are the first to show excitatory effects of PK2 on neurons and, in addition, demonstrate that this peptide modulates voltage-activated K(+) channels. The activation of SFO neurons by PK2 illustrates a mechanism through which this peptide may exert circadian control of autonomic functions.


Assuntos
Hormônios Gastrointestinais/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neuropeptídeos/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Órgão Subfornical/efeitos dos fármacos , Órgão Subfornical/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Polaridade Celular/efeitos dos fármacos , Canais de Potássio de Retificação Tardia , Relação Dose-Resposta a Droga , Técnicas In Vitro , Masculino , Técnicas de Patch-Clamp , Potássio/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Órgão Subfornical/citologia
20.
Endocrinology ; 146(9): 3950-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15932929

RESUMO

A highly vascular endocrine gland, the corpus luteum (CL) is an excellent model for the study of angiogenic factors. Prokineticins (PK-1 and -2), also termed endocrine-gland-derived vascular endothelial growth factor (VEGF) and BV8 are newly identified proteins described as selective angiogenic mitogens. We previously identified PK binding sites, two closely homologous G protein-coupled receptors (PK-R1 and PK-R2) in human and bovine ovarian cells, but their function remained unknown. In this study we examined the presence and effects of PK in CL-derived endothelial and steroidogenic cell types (LEC and LSC, respectively). PK-1 mRNA was identified in CL and follicles by real-time PCR, using primers specific for the bovine PK-1 sequence (retrieved from Bos taurus whole genome shotgun database). PK were potent angiogenic mitogens for LEC; they enhanced cell proliferation, elevated [3H]thymidine incorporation, MAPK activation, and c-jun/fos mRNA expression. The effects of PK proteins on cell survival were examined by nuclear morphology (4',6-diamidino-2-phenylindole dihydrochloride staining), measurement of DNA fragmentation (terminal dUTP nucleotide end labeling assay), and caspase-3 cleavage. Results obtained by these techniques demonstrated that PK protected LEC from serum starvation-induced apoptosis. Stress conditions such as serum withdrawal, TNF-alpha, and hypoxia markedly increased PK-R2 expression, whereas mRNA levels of PK-R1 remained unchanged. These suggest that the antiapoptotic effect of PK-1 on LEC may be mediated via PK-R2. PK-1 increased VEGF mRNA expression by LSC, implying that it could also indirectly, via VEGF, affect luteal angiogenesis. Together, these findings suggest an important role for PK-1 in luteal function by acting as a mitogen and survival factor in LEC.


Assuntos
Corpo Lúteo/citologia , Corpo Lúteo/fisiologia , Peptídeos/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Sequência de Bases , Bovinos , Divisão Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Feminino , Células da Granulosa/citologia , Células da Granulosa/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Mitógenos/genética , Mitógenos/metabolismo , Dados de Sequência Molecular , Peptídeos/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-jun/genética , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/genética , Células Tecais/citologia , Células Tecais/fisiologia , Timidina/farmacocinética , Trítio , Fator A de Crescimento do Endotélio Vascular/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA