Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Mol Genet Metab ; 131(1-2): 277-283, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32747172

RESUMO

Rare genetic variants in LDLR, APOB and PCSK9 are known causes of familial hypercholesterolaemia and it is expected that rare variants in other genes will also have effects on hyperlipidaemia risk although such genes remain to be identified. The UK Biobank consists of a sample of 500,000 volunteers and exome sequence data is available for 50,000 of them. 11,490 of these were classified as hyperlipidaemia cases on the basis of having a relevant diagnosis recorded and/or taking lipid-lowering medication while the remaining 38,463 were treated as controls. Variants in each gene were assigned weights according to rarity and predicted impact and overall weighted burden scores were compared between cases and controls, including population principal components as covariates. One biologically plausible gene, HUWE1, produced statistically significant evidence for association after correction for testing 22,028 genes with a signed log10 p value (SLP) of -6.15, suggesting a protective effect of variants in this gene. Other genes with uncorrected p < .001 are arguably also of interest, including LDLR (SLP = 3.67), RBP2 (SLP = 3.14), NPFFR1 (SLP = 3.02) and ACOT9 (SLP = -3.19). Gene set analysis indicated that rare variants in genes involved in metabolism and energy can influence hyperlipidaemia risk. Overall, the results provide some leads which might be followed up with functional studies and which could be tested in additional data sets as these become available. This research has been conducted using the UK Biobank Resource.


Assuntos
Hiperlipidemias/genética , Hiperlipoproteinemia Tipo II/genética , Pró-Proteína Convertase 9/genética , Receptores de LDL/genética , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases/genética , Apolipoproteína B-100/genética , Bancos de Espécimes Biológicos , LDL-Colesterol/genética , Exoma/genética , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Testes Genéticos , Humanos , Hiperlipidemias/patologia , Hiperlipoproteinemia Tipo II/diagnóstico , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Proteínas Celulares de Ligação ao Retinol/genética , Fatores de Risco , Reino Unido , Sequenciamento do Exoma
2.
Endocrinology ; 162(5)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33564881

RESUMO

RFamide-related peptides (RFRPs, mammalian orthologs of gonadotropin-inhibitory hormone) convey circadian, seasonal, and social cues to the reproductive system. They regulate gonadotropin secretion by modulating gonadotropin-releasing hormone (GnRH) neurons via the RFRP receptor. Mice lacking this receptor are fertile but exhibit abnormal gonadotropin responses during metabolic challenges, such as acute fasting, when the normal drop in gonadotropin levels is delayed. Although it is known that these food intake signals to the reproductive circuit originate in the nucleus tractus solitarius (NTS) in the brainstem, the phenotype of the neurons conveying the signal remains unknown. Given that neuropeptide FF (NPFF), another RFamide peptide, resides in the NTS and can bind to the RFRP receptor, we hypothesized that NPFF may regulate GnRH neurons. To address this question, we used a combination of techniques: cell-attached electrophysiology on GnRH-driven green fluorescent protein-tagged neurons in acute brain slices; calcium imaging on cultured GnRH neurons; and immunostaining on adult brain tissue. We found (1) NPFF inhibits GnRH neuron excitability via the RFRP receptor and its canonical signaling pathway (Gi/o protein and G protein-coupled inwardly rectifying potassium channels), (2) NPFF-like fibers in the vicinity of GnRH neurons coexpress neuropeptide Y, (3) the majority of NPFF-like cell bodies in the NTS also coexpress neuropeptide Y, and (4) acute fasting increased NPFF-like immunoreactivity in the NTS. Together these data indicate that NPFF neurons within the NTS inhibit GnRH neurons, and thus reproduction, during fasting but prior to the energy deficit.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Receptores de Peptídeos/metabolismo , Animais , Tronco Encefálico/metabolismo , Jejum/metabolismo , Feminino , Glicoproteínas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeos/metabolismo , Oligopeptídeos/metabolismo
3.
Domest Anim Endocrinol ; 72: 106435, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32247990

RESUMO

The 2 structurally related peptides, neuropeptide FF (NPFF) and neuropeptide AF (NPAF), are encoded by the NPFF gene and have been identified as neuromodulators that regulate nociception and opiate-mediated analgesia via NPFF receptor (NPFFR2) in mammals. However, little is known about these 2 peptides in birds. In this study, we examined the structure, tissue expression profile, and functionality of NPAF and NPFF in chickens. Our results showed that: 1) unlike mammalian NPFF, NPFF from chicken and other avian species is predicted to produce a single bioactive NPAF peptide, whereas the putative avian NPFF peptide likely lacks activity due to the absence of functional RFamide motif at its C-terminus; 2) synthetic chicken (c-) NPAF can potently activate cNPFFR2 (and not cNPFFR1) expressed in HEK293 cells, as monitored by 3 cell-based luciferase reporter systems, indicating that cNPAF is a potent ligand for cNPFFR2, which activation could decrease intracellular cAMP levels and stimulate the MAPK/ERK signaling cascade; interestingly, gonadotropin-inhibitory hormone, a peptide sharing high structural similarity to NPAF, could specifically activate cNPFFR1 (but not cNPFFR2); 3) Quantitative real-time PCR revealed that cNPFF mRNA is widely expressed in chicken tissues with the highest level detected in the hypothalamus, whereas cNPFFR2 is expressed in all tissues examined with the highest level noted in the hypothalamus and anterior pituitary. Taken together, our data reveal that avian NPFF encodes a single bioactive NPAF peptide, which preferentially activates NPFFR2, and provides insights into potential structural and functional changes of NPFF-derived peptides during vertebrate evolution.


Assuntos
Galinhas/metabolismo , Regulação da Expressão Gênica/fisiologia , Oligopeptídeos/metabolismo , Receptores de Neuropeptídeos/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Células HEK293 , Humanos , Oligopeptídeos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Neuropeptídeos/genética
4.
ACS Med Chem Lett ; 2(1): 53-7, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-24900254

RESUMO

Kisspeptin is a member of the RFamide neuropeptide family that is implicated in gonadotropin secretion. Because kisspeptin-GPR54 signaling is implicated in the neuroendocrine regulation of reproduction, GPR54 ligands represent promising therapeutic agents against endocrine secretion disorders. In the present study, the selectivity profiles of GPR54 agonist peptides were investigated for several GPCRs, including RFamide receptors. Kisspeptin-10 exhibited potent binding and activation of neuropeptide FF receptors (NPFFR1 and NPFFR2). In contrast, short peptide agonists bound with much lower affinity to NPFFRs while showing relatively high selectivity toward GPR54. The possible localization of secondary kisspeptin targets was also demonstrated by variation in the levels of GnRH release from the median eminence and the type of GPR54 agonists used. Negligible affinity of the reported NPFFR ligands to GPR54 was observed and indicates the unidirectional cross-reactivity between both ligands.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA