Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
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.

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.
PLoS One ; 9(3): e90860, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24633064

RESUMO

The possible signaling role of prokineticin 2 (PK2) and its receptor, prokineticin receptor 2 (PKR2), on female reproduction was investigated. First, the expression of PKR2 and its co-localization with estrogen receptor (ERα) in the hypothalamus was examined. Sexually dimorphic expression of PKR2 in the preoptic area of the hypothalamus was observed. Compared to the male mice, there was more widespread PKR2 expression in the preoptic area of the hypothalamus in the female mice. The likely co-expression of PKR2 and ERα in the preoptic area of the hypothalamus was observed. The estrous cycles in female PK2-null, and PKR2-null heterozygous mice, as well as in PK2-null and PKR2-null compound heterozygous mice were examined. Loss of one copy of PK2 or PKR2 gene caused elongated and irregular estrous cycle in the female mice. The alterations in the estrous cycle were more pronounced in PK2-null and PKR2-null compound heterozygous mice. Consistent with these observations, administration of a small molecule PK2 receptor antagonist led to temporary blocking of estrous cycle at the proestrous phase in female mice. The administration of PKR2 antagonist was found to blunt the circulating LH levels. Taken together, these studies indicate PK2 signaling is required for the maintenance of normal female estrous cycles.


Assuntos
Ciclo Estral/fisiologia , Hormônios Gastrointestinais/metabolismo , Neuropeptídeos/metabolismo , Animais , Receptor alfa de Estrogênio/metabolismo , Ciclo Estral/efeitos dos fármacos , Feminino , Hormônios Gastrointestinais/antagonistas & inibidores , Hormônios Gastrointestinais/genética , Hipotálamo/metabolismo , Camundongos , Camundongos Knockout , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/genética
4.
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
5.
PLoS One ; 8(1): e54916, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23372791

RESUMO

Infiltration of myeloid cells in the tumor microenvironment is often associated with enhanced angiogenesis and tumor progression, resulting in poor prognosis in many types of cancer. The polypeptide chemokine PK2 (Bv8, PROK2) has been shown to regulate myeloid cell mobilization from the bone marrow, leading to activation of the angiogenic process, as well as accumulation of macrophages and neutrophils in the tumor site. Neutralizing antibodies against PK2 were shown to display potent anti-tumor efficacy, illustrating the potential of PK2-antagonists as therapeutic agents for the treatment of cancer. In this study we demonstrate the anti-tumor activity of a small molecule PK2 antagonist, PKRA7, in the context of glioblastoma and pancreatic cancer xenograft tumor models. For the highly vascularized glioblastoma, PKRA7 was associated with decreased blood vessel density and increased necrotic areas in the tumor mass. Consistent with the anti-angiogenic activity of PKRA7 in vivo, this compound effectively reduced PK2-induced microvascular endothelial cell branching in vitro. For the poorly vascularized pancreatic cancer, the primary anti-tumor effect of PKRA7 appears to be mediated by the blockage of myeloid cell migration/infiltration. At the molecular level, PKRA7 inhibits PK2-induced expression of certain pro-migratory chemokines and chemokine receptors in macrophages. Combining PKRA7 treatment with standard chemotherapeutic agents resulted in enhanced effects in xenograft models for both types of tumor. Taken together, our results indicate that the anti-tumor activity of PKRA7 can be mediated by two distinct mechanisms that are relevant to the pathological features of the specific type of cancer. This small molecule PK2 antagonist holds the promise to be further developed as an effective agent for combinational cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Transporte/metabolismo , Transformação Celular Neoplásica/efeitos dos fármacos , Hormônios Gastrointestinais/antagonistas & inibidores , Glioma/patologia , Células Mieloides/patologia , Neovascularização Patológica , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/antagonistas & inibidores , Neoplasias Pancreáticas/patologia , Animais , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Glioma/tratamento farmacológico , Glioma/mortalidade , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos , Células Mieloides/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/mortalidade , Receptores de N-Metil-D-Aspartato , Carga Tumoral/efeitos dos fármacos , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Clin Endocrinol Metab ; 97(4): E694-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22319038

RESUMO

CONTEXT: Kallmann syndrome (KS), combined pituitary hormone deficiency (CPHD), and septo-optic dysplasia (SOD) all result from development defects of the anterior midline in the human forebrain. OBJECTIVE: The objective of the study was to investigate whether KS, CPHD, and SOD have shared genetic origins. DESIGN AND PARTICIPANTS: A total of 103 patients with either CPHD (n = 35) or SOD (n = 68) were investigated for mutations in genes implicated in the etiology of KS (FGFR1, FGF8, PROKR2, PROK2, and KAL1). Consequences of identified FGFR1, FGF8, and PROKR2 mutations were investigated in vitro. RESULTS: Three patients with SOD had heterozygous mutations in FGFR1; these were either shown to alter receptor signaling (p.S450F, p.P483S) or predicted to affect splicing (c.336C>T, p.T112T). One patient had a synonymous change in FGF8 (c.216G>A, p.T72T) that was shown to affect splicing and ligand signaling activity. Four patients with CPHD/SOD were found to harbor heterozygous rare loss-of-function variants in PROKR2 (p.R85G, p.R85H, p.R268C). CONCLUSIONS: Mutations in FGFR1/FGF8/PROKR2 contributed to 7.8% of our patients with CPHD/SOD. These data suggest a significant genetic overlap between conditions affecting the development of anterior midline in the human forebrain.


Assuntos
Fator 8 de Crescimento de Fibroblasto/genética , Hipopituitarismo/genética , Síndrome de Kallmann/genética , Mutação , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Displasia Septo-Óptica/genética , Animais , Feminino , Fator 8 de Crescimento de Fibroblasto/metabolismo , Estudos de Associação Genética , Heterozigoto , Humanos , Hipopituitarismo/metabolismo , Síndrome de Kallmann/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Neuro-Hipófise/metabolismo , Neuro-Hipófise/patologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Displasia Septo-Óptica/metabolismo , Transdução de Sinais , Reino Unido , Estados Unidos
7.
Mol Cell Endocrinol ; 346(1-2): 44-50, 2011 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-21664414

RESUMO

Prokineticin, 1 (PROK1) and prokineticin 2 (PROK2), are two closely related proteins that were identified as the mammalian homologs of their two amphibian homologs, mamba intestinal toxin (MIT-1) and Bv8. MIT-1 was initially identified as a non-toxic constituent in the venom of the black mamba snake (Dendroaspis polylepis) (Joubert and Strydom, 1980) while Bv8 was identified in the skin secretion of the toad, Bombina variegate (Mollay et al., 1999). All three homologs stimulate gastrointestinal motility thus accounting for their family name "prokineticins" (Schweitz et al., 1990, 1999). However, since its initial description, both PROK1 and PROK2 have been found to regulate a dazzling array of biological functions throughout the body. In particular, PROK1 acts as a potent angiogenic mitogen on endocrine vascular epithelium, thus earning its other name, Endocrine gland-vascular endothelial factor (EG-VEGF) (LeCouter et al., 2002). In contrast, the PROK2 signaling pathway is a critical regulator of olfactory bulb morphogenesis and sexual maturation in mammals and this function is the focus of this review.


Assuntos
Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Reprodução/fisiologia , Animais , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Bulbo Olfatório/metabolismo , Bulbo Olfatório/fisiologia , Reprodução/genética , Maturidade Sexual/genética , Maturidade Sexual/fisiologia
8.
Sleep ; 34(1): 49-56, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21203371

RESUMO

OBJECTIVES: Vasoactive intestinal polypeptide (VIP) has been implicated in sleep regulation as a promoter of rapid eye movement (REM) sleep. Previous work has shown that the amount of time spent in REM sleep is increased by intracerebroventricular administration of VIP, and reduced by treatment with VIP antagonists or antibodies against VIP. A variety of evidence suggests that VIP is critical for normal expression of circadian rhythmicity of diverse physiological and behavioral parameters. In the present study, we investigated the role of this peptide in sleep regulation using VIP-deficient (VIP-/-) mice. METHODS: EEG/EMG sleep-wake patterns were recorded in VIP-/- mice and their wild-type littermate controls under normal light-dark (LD), constant darkness (DD) and sleep deprivation conditions. RESULTS: VIP-/- mice exhibited reduced REM sleep time over the 24-h cycle while total daily amounts of NREM sleep and wakefulness were not altered significantly. The reduced REM sleep time in VIP-/- mice occurred entirely during the day due to a reduction in the duration, but not the frequency, of REM sleep bouts. In response to sleep deprivation, compensatory rebounds in NREM sleep and REM sleep were also attenuated in VIP-/- mice. Finally, the loss of VIP altered the temporal distribution of sleep in that the VIP -/- mice exhibited smaller amplitude rhythms in total sleep, NREM sleep, and REM sleep under both LD and DD. CONCLUSIONS: These results indicate that VIP regulates the duration of REM sleep, sleep homeostatic mechanisms as well as the temporal patterning of sleep.


Assuntos
Ritmo Circadiano/fisiologia , Sono REM/fisiologia , Peptídeo Intestinal Vasoativo/fisiologia , Animais , Eletroencefalografia , Eletromiografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Privação do Sono/fisiopatologia
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.
Proc Natl Acad Sci U S A ; 104(44): 17447-52, 2007 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-17959774

RESUMO

Gonadotropin-releasing hormone (GnRH) deficiency in the human presents either as normosmic idiopathic hypogonadotropic hypogonadism (nIHH) or with anosmia [Kallmann syndrome (KS)]. To date, several loci have been identified to cause these disorders, but only 30% of cases exhibit mutations in known genes. Recently, murine studies have demonstrated a critical role of the prokineticin pathway in olfactory bulb morphogenesis and GnRH secretion. Therefore, we hypothesize that mutations in prokineticin 2 (PROK2) underlie some cases of KS in humans and that animals deficient in Prok2 would be hypogonadotropic. One hundred IHH probands (50 nIHH and 50 KS) with no known mutations were examined for mutations in the PROK2 gene. Mutant PROK2s were examined in functional studies, and the reproductive phenotype of the Prok2(-/-) mice was also investigated. Two brothers with KS and their sister with nIHH harbored a homozygous deletion in the PROK2 gene (p.[I55fsX1]+[I55fsX1]). Another asymptomatic brother was heterozygous for the deletion, whereas both parents (deceased) had normal reproductive histories. The identified deletion results in a truncated PROK2 protein of 27 amino acids (rather than 81 in its mature form) that lacks bioactivity. In addition, Prok2(-/-) mice with olfactory bulb defects exhibited disrupted GnRH neuron migration, resulting in a dramatic decrease in GnRH neuron population in the hypothalamus as well as hypogonadotropic hypogonadism. Homozygous loss-of-function PROK2 mutations cause both KS and nIHH.


Assuntos
Hormônios Gastrointestinais/metabolismo , Hipogonadismo/metabolismo , Hipogonadismo/patologia , Síndrome de Kallmann/metabolismo , Síndrome de Kallmann/patologia , Neuropeptídeos/metabolismo , Animais , Sequência de Bases , Movimento Celular , Feminino , Hormônios Gastrointestinais/deficiência , Hormônios Gastrointestinais/genética , Deleção de Genes , Regulação da Expressão Gênica , Genótipo , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Hipogonadismo/genética , Síndrome de Kallmann/genética , Masculino , Camundongos , Camundongos Knockout , Mutação/genética , Neurônios/citologia , Neurônios/metabolismo , Neuropeptídeos/deficiência , Neuropeptídeos/genética , Linhagem , Fenótipo , Reprodução
12.
Biol Reprod ; 76(5): 749-58, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17229935

RESUMO

Prokineticin 1 (PROK1), also termed endocrine gland-derived vascular endothelial growth factor (endocrine gland-derived VEGF), is a newly identified protein assigned with diverse biologic functions. It binds two homologous G protein-coupled receptors, PROKR1 and PROKR2. To better understand the roles of PROK1 and its receptors in ovarian function, their expression was determined in follicles and corpora lutea (CLs) at different developmental stages. PROK1 mRNA levels were low at early luteal stage and midluteal stage, but increased sharply during natural or induced luteolysis. High PROK1 mRNA levels also were found in atretic follicles. This profile of PROK1 expression was opposite to that of the well-established angiogenic factor VEGF. Of the two receptor-type expressions, PROKR1 but not PROKR2 was correlated positively with its ligand. Immunohistochemical staining revealed that PROK1 was located mainly within the muscular layer of arterioles, and during regression it also was localized to macrophages and steroidogenic cells. The expression pattern of ITGB2 mRNA, a leukocyte cell marker, overlapped that of PROK1, thus suggesting that leukocyte infiltration may explain the elevated expression of PROK1 in atretic follicles and regressing CL. Indeed, flow cytometry analyses showed that nearly all beta-2 integrin chain (ITGB2)-positive cells also were stained with anti-PROK1 and that significantly more ITGB2/PROK1 double-stained cells were present in degenerating follicles and CL. Furthermore, when challenged in vitro with PROK1, adherent, mononuclear cell numbers and TNF levels were elevated, indicating that PROK1 triggers monocyte activation. Together, these data suggest that PROK1, acting via PROKR1, may be involved in the recruitment of monocytes to regressing CL and atretic follicles and their consequent activation therein.


Assuntos
Corpo Lúteo/crescimento & desenvolvimento , Ciclo Estral/metabolismo , Atresia Folicular/fisiologia , Ovário/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/biossíntese , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Animais , Bovinos , Contagem de Células , Ceramidas/metabolismo , Enzima de Clivagem da Cadeia Lateral do Colesterol/biossíntese , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Dinoprosta/biossíntese , Dinoprosta/genética , Escherichia coli/metabolismo , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese
13.
J Biol Chem ; 282(10): 6917-21, 2007 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-17259180

RESUMO

Prokineticin 2, a cysteine-rich secreted protein, regulates diverse biological functions including the neurogenesis of olfactory bulb. Here we show that the PK2 gene is a functional target gene of proneural basic helix-loop-helix (bHLH) factors. Neurogenin 1 and MASH1 activate PK2 transcription by binding to E-box motifs on the PK2 promoter with the same set of E-boxes critical for another pair of bHLH factors, CLOCK and BMAL1, in the regulation of circadian clock. Our results establish PK2 as a common functional target gene for different bHLH transcriptional factors in mediating their respective functions.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Hormônios Gastrointestinais/genética , Neuropeptídeos/genética , Bulbo Olfatório/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Movimento Celular , Regulação da Expressão Gênica , Camundongos , Proteínas do Tecido Nervoso/fisiologia , Regiões Promotoras Genéticas , Células-Tronco/citologia
14.
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
15.
Mol Interv ; 6(6): 330-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17200460

RESUMO

Secreted peptides play broad regulatory roles in brain function and elsewhere in the body. Prokineticins are a pair of newly identified regulatory peptides that signal through two highly homologous G protein-coupled receptors. Prokineticins possess a unique structural motif of five disulfide bonds and a completely conserved N-terminal hexapeptide sequence that is essential to biological activity. Diverse biological functions, including roles in development and cell differentiation, have been assigned to the prokineticins. A network of genes, subject to various transcriptional factors, may functionally converge on the prokineticins as regulatory targets.


Assuntos
Peptídeos/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Sequência de Aminoácidos , Animais , Ritmo Circadiano/fisiologia , Humanos , Dados de Sequência Molecular , Contração Muscular , Músculo Liso/fisiologia , Peptídeos/química , Peptídeos/genética , Transdução de Sinais , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/química , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
16.
FEBS J ; 272(22): 5703-9, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16279936

RESUMO

Circadian timing from the suprachiasmatic nucleus (SCN) is a critical component of sleep regulation. Animal lesion and genetic studies have indicated an essential interaction between the circadian signals and the homeostatic processes that regulate sleep. Here we summarize the biological functions of prokineticins, a pair of newly discovered regulatory proteins, with focus on the circadian function of prokineticin 2 (PK2) and its potential role in sleep-wake regulation. PK2 has been shown as a candidate SCN output molecule that regulates circadian locomotor behavior. The PK2 molecular rhythm in the SCN is predominantly controlled by the circadian transcriptional/translational loops, but also regulated directly by light. The receptor for PK2 is expressed in the primary SCN output targets that regulate circadian behavior including sleep-wake. The depolarizing effect of PK2 on neurons that express PK2 receptor may represent a possible mechanism for the regulatory role of PK2 in circadian rhythms.


Assuntos
Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Hormônios Gastrointestinais/fisiologia , Regulação da Expressão Gênica/fisiologia , Neuropeptídeos/fisiologia , Sono/fisiologia , Sequência de Aminoácidos , Animais , Relógios Biológicos/genética , Relógios Biológicos/fisiologia , Cisteína/química , Hormônios Gastrointestinais/química , Luz , Camundongos , Dados de Sequência Molecular , Atividade Motora/fisiologia , Neuropeptídeos/química , Sinais Direcionadores de Proteínas , Núcleo Supraquiasmático/fisiologia , Transcrição Gênica
17.
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
18.
Mol Pharmacol ; 65(3): 582-8, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14978236

RESUMO

Prokineticins are cysteine-rich secreted proteins that regulate diverse biological processes, including gastrointestinal motility, angiogenesis, and circadian rhythms. Two closely related G protein-coupled receptors that mediate signal transduction of prokineticins have recently been cloned. The structural elements required for prokineticins' bioactivities are still unknown. We show here that both the N-terminal hexapeptide (AVITGA) and C-terminal cysteine-rich domains are critical for the bioactivities of prokineticins. Substitutions, deletions, and insertions to the conserved N-terminal hexapeptides result in the loss of agonist activity. Mutant prokineticins with the substitution of the first N-terminal alanine with methionine or the addition of a methionine to the N terminus inhibit the activation of prokineticin receptors and thus are considered as antagonists of prokineticin receptors. We have further shown that mutations in selected cysteine residues in the C-terminal domain result in prokineticins without biological activity. The essential role of C-terminal domain is reinforced by two observations: that peptides without the carboxyl domain and proteins with the N-terminal hexapeptide fused to the carboxyl domains of colipase or dickkopf are devoid of biological activity. We demonstrate that limited structural changes of C-terminal cysteine-rich regions of prokineticins are tolerable because chimeric prokineticins with swapped cysteine-rich domains between prokineticin 1 and prokineticin 2, as well as a splice variant of prokineticin 2 that contains extra 21 residue insertion in its C-terminal domain, are biologically active.


Assuntos
Processamento Alternativo , Hormônios Gastrointestinais/metabolismo , Neuropeptídeos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Animais , Células CHO , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Cricetinae , Cisteína/química , Hormônios Gastrointestinais/química , Hormônios Gastrointestinais/genética , Mutagênese Sítio-Dirigida , Neuropeptídeos/química , Neuropeptídeos/efeitos dos fármacos , Neuropeptídeos/genética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química
19.
Nature ; 417(6887): 405-10, 2002 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-12024206

RESUMO

The suprachiasmatic nucleus (SCN) controls the circadian rhythm of physiological and behavioural processes in mammals. Here we show that prokineticin 2 (PK2), a cysteine-rich secreted protein, functions as an output molecule from the SCN circadian clock. PK2 messenger RNA is rhythmically expressed in the SCN, and the phase of PK2 rhythm is responsive to light entrainment. Molecular and genetic studies have revealed that PK2 is a gene that is controlled by a circadian clock (clock-controlled). Receptor for PK2 (PKR2) is abundantly expressed in major target nuclei of the SCN output pathway. Inhibition of nocturnal locomotor activity in rats by intracerebroventricular delivery of recombinant PK2 during subjective night, when the endogenous PK2 mRNA level is low, further supports the hypothesis that PK2 is an output molecule that transmits behavioural circadian rhythm. The high expression of PKR2 mRNA within the SCN and the positive feedback of PK2 on its own transcription through activation of PKR2 suggest that PK2 may also function locally within the SCN to synchronize output.


Assuntos
Ritmo Circadiano/fisiologia , Hormônios Gastrointestinais/metabolismo , Atividade Motora/fisiologia , Neuropeptídeos , Núcleo Supraquiasmático/fisiologia , Células 3T3 , Animais , Relógios Biológicos/genética , Relógios Biológicos/fisiologia , Linhagem Celular , Ritmo Circadiano/genética , Hormônios Gastrointestinais/genética , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica
20.
J Biol Chem ; 277(22): 19276-80, 2002 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-11886876

RESUMO

We previously described two mammalian secreted proteins, prokineticin 1 and prokineticin 2, that potently contract gastrointestinal smooth muscle. Prokineticin 1 has also been shown to promote angiogenesis by stimulating proliferation, migration, and fenestration of endocrine organ-derived endothelial cells. Here we report the cloning and characterization of two closely related G protein-coupled receptors as receptors for prokineticins. Expression of prokineticin receptors in heterologous systems shows that these receptors bind to and are activated by nanomolar concentrations of recombinant prokineticins. Activation of prokineticin receptors leads to mobilization of calcium, stimulation of phosphoinositide turnover, and activation of p44/p42 MAPK signaling pathways that are consistent with the effects of prokineticins on smooth muscle contraction and angiogenesis. mRNA expression analysis reveals that prokineticin receptors are expressed in gastrointestinal organs, endocrine glands, and other tissues.


Assuntos
Glândulas Endócrinas/metabolismo , Fatores de Crescimento Endotelial/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Hormônios Gastrointestinais/metabolismo , Linfocinas/metabolismo , Neuropeptídeos , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Equorina/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Células COS , Cálcio/metabolismo , Clonagem Molecular , Cricetinae , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Fatores de Crescimento Endotelial/química , Humanos , Ligantes , Linfocinas/química , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , Fosforilação , Plasmídeos/metabolismo , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fatores de Tempo , Distribuição Tecidual , Transfecção , Fator A de Crescimento do Endotélio Vascular , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina , Fatores de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA